spoons full of natural ingredients to prevent Chronic Inflammation

Inflammation (Chronic)

Inflammation (Chronic)

Last Section Update: 11/2025

Contributor(s): Maureen Williams, ND; Colleen Mazin, MS/MPH; Debra Gordon, MS; Stephen Tapanes, PhD; Jennifer Pryor, M.D.

1 Introduction

Of the 10 leading causes of mortality in the United States, chronic, low-level inflammation contributes to the pathogenesis of at least seven. These include heart disease, cancer, chronic lower respiratory disease, stroke, Alzheimer’s disease, diabetes, and nephritis.1-9

Inflammation has classically been viewed as an acute (short-term) response to tissue injury that produces characteristic symptoms and usually resolves spontaneously. More contemporary revelations show chronic inflammation to be a major factor in the development of degenerative disease and loss of youthful functions.

Chronic inflammation can be triggered by cellular stress and dysfunction, such as that caused by excessive calorie consumption, elevated blood sugar levels, and oxidative stress. It is now clear that the destructive capacity of chronic inflammation is unprecedented among physiologic processes.10

The danger of chronic, low-level inflammation is that its silent nature belies its destructive power.

In fact, stress-induced inflammation, once triggered, can persist undetected for years, or even decades, propagating cell death throughout the body. Due to the fact that it contributes so greatly to deterioration associated with the aging process, this silent state of chronic inflammation has been coined “inflammaging.”

Chronic low-level inflammation may be threatening your health at this very moment, without you realizing it. In this protocol you will learn about low-cost blood tests that can assess the inflammatory state within your body. You will also discover novel approaches that combat chronic inflammation to help avoid age-related health decline.

2 The Inflammatory Process

The Acute Inflammatory Response

Inflammation, the adaptive immune response to tissue injury or infection, plays a central role in metabolism in a variety of organisms.11

At its most basic level, an acute inflammatory response is triggered by 1) tissue injury (trauma, exposure to heat or chemicals); or 2) infection by viruses, bacteria, parasites, or fungi. The classic manifestation of acute inflammation is characterized by four cardinal signs: Redness and heat result from the increased blood flow to the site of injury. Swelling results from the accumulation of fluid at the injury site, a consequence of the increased blood flow. Finally, swelling can compress nerve endings near the injury, causing the characteristic pain associated with inflammation. Pain is also important to make the organism aware of the tissue damage. Additionally, inflammation in a joint usually results in a fifth sign (impairment of function), which has the effect of limiting movement and forcing rest of the injured joint to aid in healing.

A well-controlled acute inflammatory response has several protective roles:

  • It prevents the spread of infectious agents and damage to nearby tissues;
  • helps to remove damaged tissue and pathogens; and
  • assists the body's repair processes

However, a third type of stimuli, cellular stress and malfunction, triggers chronic inflammation, which, rather than benefiting health, contributes to disease and age-related deterioration via numerous mechanisms.

Cellular Stress and Chronic, Low-Level Inflammation

Mitochondria—cellular organelles responsible for generating biochemical energy in the form of adenosine triphosphate (ATP)—are a fundamentally necessary component of life in higher organisms. In fact, in the case of sophisticated multicellular life forms, organismal viability depends upon optimal mitochondrial function. Paradoxically, mitochondrial processes can also bring about a tissue-destroying inflammatory mediator known as the inflammasome; this phenomenon is provoked by damaged and dysfunctional mitochondria.12

Mitochondrial dysfunction arises consequent of exposure to exogenous (eg, environmental toxins, tobacco smoke) and endogenous (eg, reactive oxygen species) stressors, and as a result of the aging process itself. For example, a byproduct of mitochondrial energy generation is the creation of free radical molecules. Free radicals can damage cellular structures and initiate a cascade of proinflammatory genetic signals that ultimately results in cell death (apoptosis), or worse, uncontrolled cell growth—the hallmark of cancer.

Aging is associated with declining mitochondrial efficiency and increased production of free radical molecules. Recent research identifies this age-associated aberration of mitochondrial function as a principle actuator of chronic inflammation.13 Specifically, mitochondrial dysfunction brings about inflammation as follows:

  1. Accumulation of free radicals induces mitochondrial membrane permeability;
  2. Molecular components normally contained within the mitochondria leak into the cytoplasm (intracellular fluid in which cellular organelles are suspended);
  3. Cytoplasmic pattern recognition receptors (PRR's), which detect and initiate an immune response against intracellular pathogens, recognize the leaked mitochondrial molecules as potential threats;
  4. Upon detection of the potential threat, PRR's form a complex called the inflammasome that activates the inflammatory cytokine interleukin-1β, which then recruits components of the immune system to destroy the “infected” cell.14

These four steps represent a simplified scheme of mitochondrial dysfunction leading to cellular destruction; however, intracellular free radicals are not the only inducers of inflammatory cell death.

Circulating sugars, primarily glucose and fructose, are culprits as well. When these “blood sugars” come in contact with proteins and lipids a damaging reaction occurs forming compounds called advanced glycation end products (AGEs). AGEs bind to the cell-surface receptor called receptor for advanced glycation end products, or RAGE. Upon activation, RAGE triggers the movement of the inflammatory mediator nuclear factor kappa-B (NF-κB) to the nucleus, where it activates numerous inflammatory genes.15 AGEs are primarily formed in vivo, and glycation is exacerbated by elevated blood sugar levels. However, dietary AGEs also contribute to inflammation; they are abundant in foods cooked at high temperatures, especially red meat.16,17

Additional biochemical inducers of a chronic inflammatory response include:

  • Uric acid (urate) crystals, which can be deposited in joints during gouty arthritis; elevated levels are a risk factor for kidney disease, hypertension, and metabolic syndrome18,19;
  • Oxidized lipoproteins (such as LDL), a significant contributor to atherosclerotic plaques20; and
  • Homocysteine, a non-protein-forming amino acid that is a marker and risk factor for cardiovascular disease, and may increase bone fracture risk.21

Together, these proinflammatory instigators promote a perpetual low-level chronic inflammatory state called para-inflammation.11

Although it progresses silently, para-inflammation presents a major threat to the health and longevity of all aging humans. Chronic, low-level inflammation is associated with common diseases including cancer, type II diabetes, osteoporosis, cardiovascular diseases, and others. Thus, by targeting the myriad physiological variables that can inaugurate an inflammatory response, one can effectively temper chronic inflammation and reduce their risk for inflammatory diseases.

3 Inflammation: Lab Tests, Biomarkers & Mediators

Systemic inflammation depends on pro-inflammatory communication between damaged or infected cells and immune cells, as well as between different types of immune cells. A broad array of molecules participate in the inflammatory signaling network. For example, cytokines are a large family of circulating molecules produced by virtually every cell in the body, including immune cells, to regulate inflammation.303 Eicosanoids, molecules made from polyunsaturated fatty acids (PUFAs) in the cell membrane, influence local immune activity by regulating intracellular inflammatory signaling.304 Blood levels of key cytokines and eicosanoids can be measured to assess the degree of systemic inflammation. In addition, certain enzymes and proteins involved in synthesizing, regulating, or responding to inflammatory messengers can be useful indicators of inflammatory status.305

The following section covers several of the inflammatory factors that can participate in chronic, systemic inflammation and which can be measured through lab testing. This list is presented in alphabetical order.

Blood Tests

The following blood tests for markers of systemic inflammation are used to assess inflammatory status. They can also be useful for monitoring treatment response to anti-inflammatory therapies, including diet and lifestyle changes, nutrient supplements, and condition-appropriate medications.306-309

Table 1: Lab Tests for Assessing Chronic Inflammation

Pro-Inflammatory Marker LabCorp Methodology & Reference Range Quest Methodology & Reference Range
Antinuclear antibodies (ANAs)

Indirect fluorescent antibody (IFA)

Negative: <1:80

Immunofluorescence assay (IFA)

Negative: <1:40

C-reactive protein (high-sensitivity; hs-CRP)

Immunochemiluminometric assay (ICMA)

(mg/L)

Low risk: <1.00

Average risk: 1.00−3.00

Immunoturbidimetric

(mg/L)

Lower relative CV risk: < 1.0

1.0–3.0: Average relative CV risk

Fibrinogen activity

Clauss method

193−507 mg/dL

Clauss method

175–425 mg/dL

Galectin-3

Enzyme immunoassay (EIA)

<22.2 ng/mL - in patients without heart disease

<17.8 ng/mL lower risk, in heart failure patients

Enzyme-linked immunosorbent assay (ELISA)

<17.9 ng/mL

GlycA

Nuclear magnetic resonance (NMR)

<400 µmol/L

N/A
Interleukin-6 (IL-6)

Electrochemiluminescence immunoassay (ECLIA)

0‒13 pg/mL

Immunoassay (IA)

< 5 pg/mL

Neutrophil-to-lymphocyte

ratio (available as part of a complete blood count [CBC] with differential)

0.0–2.9 As listed on CBC report

Omega-3 index

(not a Quest or LabCorp test; available from OmegaQuant)

8–11%
Serum amyloid A (SAA)

Electrochemiluminescence (MSD)

0.4–5.0 mcg/mL

Tumor necrosis factor-alpha (TNF-α)

Enzyme-linked immunosorbent assay (ELISA)

<8.1 pg/mL

Immunoassay (IA)

TNF-α, highly sensitive

0.56–1.40 pg/mL

White blood cell count (component of CBC) 3.4–10.8 x 103/µL

Antinuclear Antibodies

Relevant lab testing: Antinuclear antibodies (indirect fluorescent antibody)

Optimal result: Negative

Antinuclear antibodies (ANAs) are immune proteins that target cells in the body. Certain ANA subtypes are characteristic of autoimmune connective tissue disorders like systemic lupus erythematosus (SLE), Sjögren syndrome, and mixed connective tissue disease (MCTD). In patients with clinical symptoms, ANA positivity can help confirm a diagnosis. ANAs may also be present in patients with other autoimmune diseases, including celiac disease, inflammatory bowel disease, psoriasis, type 1 diabetes, Addison’s disease, and autoimmune thyroid disease. In addition, they can be detected in some patients with latent infections, alcoholic liver disease, blood cancers, and other inflammatory conditions, and are thought to reflect general immune dysregulation rather than an autoimmune process.310 Detection of ANAs can prompt further investigation into the potential presence of specific underlying inflammatory conditions.311

Evidence shows some healthy individuals also express ANAs.310 One study in more than 25,000 healthy subjects found 14.01% had positive ANA titers (amounts) > 1:100. This means ANAs were still detectable at dilutions of 1:100 or greater. Even at a level of 1:320, positive ANA titers were found in women at more than 2.5 times the rate compared with men.312 Other evidence suggests as many as 45% of healthy people have positive, though low, ANA titers.310 Asymptomatic ANA positivity has been correlated with markers of metabolic disturbance and inflammation.313 Although some ANA-positive individuals go on to develop autoimmune disorders, many do not, and evidence suggests imbalanced inflammatory signaling may trigger the transition from asymptomatic to symptomatic ANA carrier.314 Analysis of data from the National Health and Nutrition Examination Survey (NHANES) suggests the prevalence of ANA positivity is increasing over time in the general U.S. population, ranging from 11% in 1988–1991 to just over 16% in 2011–2012.315

C-reactive Protein

Relevant lab testing: High-sensitivity CRP (hs-CRP)

Optimal level: < 1 mg/L

C-reactive protein (CRP), an acute-phase protein, is one of several proteins rapidly produced primarily by the liver during an inflammatory response. CRP production can be triggered by IL-6 and interleukin 1- beta (IL-1β). CRP plays an important role in the immune response by facilitating immune recognition of damaged or infected cells and cellular debris. CRP may also contribute to blood clotting.316-320

CRP production rises and decreases rapidly in response to inflammatory signaling, but chronic elevation can result from ongoing inflammatory immune activation as well as tissue damage.320 Chronically elevated CRP levels (detectable by a high-sensitivity CRP or hs-CRP test) are associated with age-related conditions including heart disease, type 2 diabetes, cancer, sarcopenia, and frailty, as well as all-cause mortality.321 In a study involving 2,437 middle-aged and older individuals (47–87 years) whose CRP levels were monitored over a decade, rising levels were correlated with adverse age-related health outcomes.322 High-sensitivity CRP levels are routinely measured as part of cardiovascular risk assessment and high levels are a stand-alone risk factor for heart disease-related death.320 CRP levels are also used to monitor disease progression in patients with cancer, autoimmune diseases, and some viral infections.323

Cyclooxygenases and Lipoxygenases

Relevant lab testing: Reasonably reflected in the omega-3 index

Optimal level: About 8%

Cyclooxygenases (COX) and lipoxygenases (LOX), along with cytochrome P450 oxidases, are the enzymes that catalyze conversion of omega-3 and omega-6 fatty acid derivatives into eicosanoids, specialized pro-resolving mediators (SPMs), and related metabolites.324 COX enzymes are the target of common anti-inflammatory drugs (including aspirin, naproxen, and ibuprofen) used to treat inflammation, fever, and pain, and a LOX-inhibiting drug (zileuton [Zyflo]) is available and approved for treating asthma. Whereas the products of COX and LOX enzymes are most often associated with pro-inflammatory processes, it is important to remember these enzymes also produce factors that inhibit or resolve inflammation, inhibit blood clotting, promote tissue repair, and support other cell and tissue functions. By blocking formation of these byproducts, COX- and LOX-inhibiting drugs can cause serious adverse side effects, many of which are known to be attributable to a decrease in beneficial effects of PUFA metabolites.325-331 Whereas levels or activity of COX and LOX enzymes are generally not assessed directly in common lab tests of inflammatory status, sometimes assessments of products of COX and LOX enzymatic activity may332 be undertaken, typically in research settings.333 Also, the omega-3 index is relevant in this context as it reflects the red blood cell membrane content of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) as a percentage of total fatty acids. Omega-3 fatty acids can be converted to inflammation-resolving or less-inflammatory compounds than omega-6 fatty acids via COX and LOX pathways.334-336

Eicosanoids

Relevant lab testing: Reasonably reflected in the omega-3 index

Optimal level: About 8%

Eicosanoids are a family of lipid compounds that modulate local immune activity and have multiple other effects on cell and organ function. Classic eicosanoid categories include prostaglandins, prostacyclins, leukotrienes, and thromboxanes. Related compounds produced from the same precursors are also thought to be important immune modulators. Eicosanoids and their related compounds are synthesized by nearly all cell types from PUFAs in cell membranes: the essential omega-6 fatty acid linoleic acid and its derivative arachidonic acid, as well as the essential omega-3 fatty acid alpha-linolenic acid (ALA) and its derivatives EPA and DHA. Eicosanoids have complex, sometimes contradictory, effects on inflammatory signaling; but in general, the most potent inflammatory eicosanoids are produced from omega-6 fatty acids, whereas eicosanoids derived from omega-3 fatty acids are less inflammatory or anti-inflammatory.324

Production of eicosanoids can be stimulated by oxidative stress and cytokines. Eicosanoids, in turn, can influence the immune response by upregulating or downregulating signal transduction pathways, such as NF-kappaB, and triggering or suppressing inflammasome activation.304 Other products of PUFA metabolism also modulate inflammatory processes. Among these are a group of compounds referred to as specialized pro-resolving mediators (SPMs), produced mainly from omega-3 fatty acids and are thought to have an important role in ending immune reactions.324

The omega-3 index is a measure of the EPA and DHA content of red blood cell membranes, expressed as a percentage of total fatty acid content. It is an indirect measure of inflammatory potential related to eicosanoids and SPMs. Low omega-3 index has been associated with increased risk of chronic inflammatory diseases including cardiovascular disease, type 2 diabetes, and chronic kidney disease, as well as all-cause mortality.337

Fibrinogen

Relevant lab testing: Fibrinogen (activity) blood test

Normal levels: 193–507 mg/dL

Fibrinogen, an acute-phase protein produced mainly in the liver, is involved in blood clotting and tissue repair through its conversion to the structural protein, fibrin. Its production is regulated largely by inflammatory signaling.338 Fibrinogen and fibrin stimulate inflammatory processes and elevated levels have been associated with venous thrombosis, atherosclerosis, coronary artery disease, stroke, and cardiovascular and all-cause mortality.338-340 In addition, higher fibrinogen levels have been linked to worse outcomes in patients with coronary artery disease,341-343 cerebrovascular disease,344 and breast cancer.345 Elevated fibrinogen has also been implicated as a contributor to metabolic disease, neurological disorders, and cancer progression.338 Levels are generally higher in women than men, and observational studies suggest elevated levels in women may be an indicator of early, asymptomatic, coronary artery disease.344

There are several methods for determining fibrinogen levels. The most common technique, called the Clauss method, involves adding a high concentration of thrombin to a plasma sample. Thrombin stimulates the conversion of fibrinogen into fibrin, which then triggers clot formation. Fibrinogen concentration can be estimated based on the time required for clotting to occur: a longer clotting time indicates a low fibrinogen level, and a short clotting time indicates a high fibrinogen level.346 The Clauss method is recommended as the initial screening test for functional fibrinogen, but its accuracy may be reduced in patients using certain anticoagulants.347 Normal ranges of fibrinogen may vary from lab to lab, partly due to different assessment techniques.

Regarding estimation of an optimal fibrinogen level, the differing lab methods used in different studies make this challenging. To keep things as straightforward as possible, the studies summarized in the following paragraph are limited to those that reported using the Clauss method or a similar method for assaying fibrinogen level. We have omitted discussion of specific fibrinogen levels from studies that reported using a different assay method.

A study that followed 4,730 subjects from the third National Health and Nutrition Examination Survey (NHANES III) for a median of almost 20 years found those with fibrinogen levels of ≤305 mg/dL had the lowest risk of all-cause, cardiovascular, and cancer-related mortality; those whose levels were between 306 and 409 mg/dL had a higher risk; and those with levels ≥410 mg/dL had the highest all-cause mortality risk—81% higher than those with the lowest levels. Fibrinogen was assessed in this study using a method that appears to be similar to the Clauss method.348

Another study evaluated data from 13,195 coronary artery disease patients who had undergone percutaneous coronary intervention in Germany between 2000 and 2009. The researchers measured fibrinogen activity using a modified Clauss method. The lowest all-cause mortality rate was seen in those with fibrinogen levels between 295 and 369 mg/dL; both higher and lower fibrinogen levels were associated with increased mortality risk.342 In a study involving subjects with cerebrovascular disease, those with fibrinogen levels greater than 400 mg/dL were at markedly higher risk of cognitive impairment compared with those whose fibrinogen levels were in the range of 200-400 mg/dL.349 In 855 postmenopausal women who underwent breast cancer surgery, those with pre-surgery, Clauss method-determined fibrinogen levels ≤320 mg/dL had the highest 10-year overall and disease-free survival rates, whereas those with levels ≥373 mg/dL had the lowest 10-year survival rate.345

Very low levels of fibrinogen are also problematic. In 3,049 patients with traumatic brain injury (TBI), Clauss method-determined fibrinogen levels of 150 mg/dL or lower were associated with a 75% increased chance of in-hospital mortality.350

Galectin-3

Relevant lab testing: Galectin-3, blood (serum or plasma) test

Lab methodology: Enzyme immunoassay (EIA)

Optimal level: <17.8 g/mL

Galectin-3 is a lectin, or binding protein, that interacts with the carbohydrate portions of glycoproteins; such as those in cell membranes, extracellular matrix, biological fluids, and inside cells. Galectin-3 helps initiate and amplify the acute inflammatory response. It also takes part in perpetuating chronic inflammatory signaling. Galectin-3 is therefore a biomarker of inflammatory activity and high levels are associated with inflammatory conditions including cardiovascular, kidney, lung, liver, neurological, autoimmune, metabolic, and infectious diseases.351-353 Elevated galectin-3 levels have also been linked to increased mortality among patients on maintenance hemodialysis.354

Galectin-3 overexpression has been shown to play a unique role in cancer cell development, proliferation, survival, and metastasis.355,356 In the heart, galectin-3 also appears to be directly involved in formation of fibrotic (scarred) tissue, leading to atrial fibrillation and heart failure.357 Indeed, heart problems like atrial fibrillation and heart failure have been correlated with high circulating levels of galectin-3.358-360 Because of its role in inflammation and fibrosis, emerging research suggests galectin-3 may be a useful predictor of outcomes following heart surgery and may be a target for treatment of a range of inflammatory diseases, including heart failure, neuro-inflammatory disorders, and cancer.355,356,361-364

Glycoprotein Acetyls (GlycA)

Relevant lab testing: GlycA blood test

Lab methodology: Nuclear magnetic resonance (NMR)

Optimal level: <400 μmol/L

Glycoprotein acetyls (GlycA) is a composite biomarker of inflammation measured through nuclear magnetic resonance testing of blood samples. GlycA quantifies the degree of glycosylation of multiple acute-phase proteins. Acute-phase proteins are released in response to acute inflammatory signaling, making it difficult to distinguish chronic from short-term inflammation. Because GycA is a composite marker, it is thought to better reflect chronic inflammation, as distinct from acute inflammation.365-367 One study that followed about 14,000 subjects from birth to age 24 years found GlycA levels were more stable than hs-CRP levels over time, but the two markers were similar in their correlations with other markers of inflammation.368 Another study that examined 1,518 blood samples from adult twins, aged 43–88 years, found GlycA was a better predictor of, and more closely linked to, other inflammatory marker levels than CRP.369

Multiple observational studies have shown higher GlycA levels are strongly correlated with a broad range of chronic inflammatory conditions, including atherosclerosis and coronary artery disease,366,370,371 cognitive impairment and Alzheimer disease,372,373 obesity,367 chronic kidney disease,374 depression,375 rheumatoid arthritis,376 chronic respiratory diseases,377 irritable bowel syndrome,378 metabolic-associated fatty liver disease (formerly called non-alcoholic fatty liver disease),379 and type 2 diabetes and its complications.380,381 One study that followed 5,526 adults for an average of 12.6 years found higher GlycA levels were associated with increased all-cause mortality rates.382 Another study in 3,306 adolescents and young adults found those with higher baseline GlycA levels had a greater risk of developing hypertension or metabolic syndrome during 9–10 years of follow-up.365

Interleukins

Relevant lab testing: IL-6 serum (blood) test (ECLIA)

Normal reference range: 0.0–13.0 pg/mL

Interleukins are a large family of cytokines produced mainly by immune cells that have many functions in the promotion and resolution of inflammation. Interleukin (IL)-1 (including IL-1α and IL-1β), IL-6, and IL-8 have well studied pro-inflammatory effects; whereas IL-4, IL-10, and IL-13 are anti-inflammatory.305 Immune cells release pro-inflammatory interleukins to accelerate the inflammatory process and anti-inflammatory interleukins to downregulate inflammation and promote healing. Several pro-inflammatory interleukins can also be released by epithelial and endothelial cells to initiate an immune response to infection or injury.303 A subset of pro-inflammatory interleukins act as chemokines, orchestrating the movement of circulating immune cells to the blood vessel wall and out of the bloodstream toward stressed cells and tissues.305

Interleukin-1β is activated by inflammasome-triggered production of the enzyme caspase-1. Evidence suggests inflammasome activation of IL-1β may be involved in a range of inflammatory conditions including cardiovascular, liver, lung, neurological, and autoimmune diseases; as well as periodontal disease, a condition closely linked to cardiovascular and other chronic inflammatory disorders.383-385

Interleukin-6 has complex effects on inflammatory signaling: low or controlled levels support healthy cell function and metabolism, whereas high levels exert pro-inflammatory and pro-oxidative actions.386 High levels of IL-6 have been reported in people with a range of age-related conditions including cardiovascular disease, cognitive decline, vascular dementia, metabolic disorders, sarcopenia, and frailty, as well as autoimmune conditions.303,387-389 In fact, in a meta-analysis of studies that compared cytokine levels in 8,154 older subjects with chronic disease to those in 33,967 healthy older subjects found those with chronic disease had higher levels of IL-6 (but not IL-1β).390 Because of its association with age-related ailments, IL-6 has been referred to as a “gerokine.”386

Interleukin-8 is a strong up-regulator of inflammation and, along with IL-1β and IL-6, a major contributor to age-related inflammation.391 Higher IL-8 levels have been correlated with postmenopausal osteoporosis and worse outcomes in patients with severe acute conditions.303

Nuclear Factor Kappa B

Relevant lab testing: Generally not assessed directly. Other inflammation-related tests such as hs-CRP may be viewed as proxies.

Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) is a nuclear protein complex that helps regulate cytokine production and other critical cellular functions in almost all cell types. NF-kappaB is activated by stimuli such as pathogen-related particles or inflammatory cytokines like TNF-α, IL-1, and C-reactive protein. In turn, it upregulates the expression of proteins involved in the inflammatory response.305,392 This includes proteins that make up inflammasomes—multi-protein complexes in the cell cytoplasm that, when stimulated, trigger an immune response.392,393 Dysregulated inflammasome activity is a contributing factor in chronic inflammatory conditions including type 2 diabetes, metabolic disease, cardiovascular disease, neurodegenerative disorders, and cancer.393

Serum Amyloid A

Relevant lab testing: Serum amyloid A (blood) test

Normal reference range: 0.5–5.0 μg/mL

Serum amyloid A is a highly sensitive acute-phase reactant synthesized predominantly by the liver in response to inflammatory stimuli. Serum amyloid A can be helpful in assessing chronic low-grade inflammation due to its sensitivity and ability to reflect subclinical inflammatory states.394-396 It is particularly useful in conditions where traditional markers like CRP may not be elevated. For example, serum amyloid A has been shown to correlate well with disease activity in inflammatory bowel disease, Crohn’s disease, and rheumatoid arthritis, even in patients with low CRP levels.397-399 In chronic inflammatory conditions, elevated serum amyloid A levels can indicate persistent inflammation and are associated with increased cardiovascular and kidney risk in patients with rheumatoid arthritis.398

Several studies have shown that higher levels of serum amyloid A predict increasingly poor cardiovascular outcomes and may be causally involved in the pathogenesis of atherosclerosis.400,401 A systematic review and meta-analysis also confirmed that high serum amyloid A levels are significantly associated with an increased risk of coronary heart disease, particularly in older adults.402

Tumor Necrosis Factor-alpha and Its Receptors

Relevant lab testing: Tumor necrosis factor-alpha serum (blood) test

Normal reference range: 0.0–2.2 pg/mL

Tumor necrosis factor-alpha (TNF-α) is a cytokine that activates inflammatory pathways by interacting with TNF receptors, TNFR1 and TNFR2. It was originally identified as an antitumor compound produced by macrophages (a type of immune cell), and is now known to be required for proper overall immune surveillance and responsiveness.303,305 TNF-α is released by all types of immune cells as well as epithelial cells (superficial cells lining surfaces exposed to the environment) and endothelial cells (cells comprising the inner lining of blood vessels).303 It transmits inflammation-regulating signals via TNFR1, which is expressed by nearly all the cells in the body; and TNFR2, which occurs on immune cells, neurons, and endothelial cells. TNFRs activate NF-kappaB, but the nature of the response depends on factors that are not fully understood. Because it can activate multiple signaling pathways with broad systemic effects, TNF-α is sometimes referred to as the master pro-inflammatory cytokine.403

Excessive or persistent TNF-α signaling is involved in chronic inflammation and may play a role in the development of cancer.303 High TNF-α levels have been associated with certain chronic inflammatory conditions, particularly metabolic conditions, such as obesity and type 2 diabetes; and autoimmune diseases, like rheumatoid arthritis and inflammatory bowel disease.403-406 One study that looked at genetic predictors of TNF-α expression found evidence suggesting increased TNF-α levels were causally linked with increased risk of coronary artery disease and stroke, but also with decreased risk of cancer.407 Nevertheless, a meta-analysis of studies that compared cytokine levels in 8,154 older subjects with chronic disease to those in 33,967 healthy older subjects found high TNF-α levels were not consistently correlated with the presence of chronic disease.390

Activation of TNFRs can result in the release of soluble forms of TNFRs that are detectable in the blood and have been shown to be indicators of inflammation. Higher soluble TNFR1 levels have been linked to coronary artery inflammation and increased risks of cardiovascular disease and type 2 diabetes, as well as faster progression of kidney disease.408-410 Studies in elderly subjects have found higher blood levels of soluble TNFR1 were correlated with deleterious effects of aging on DNA and functional ability.321,411 A meta-analysis of observational studies found higher levels of soluble TNFR2 were consistently seen in patients with a variety of cancers compared with healthy subjects.412

White Blood Cells and the Neutrophil-to-Lymphocyte Ratio

Relevant lab testing: Standard complete blood count (CBC) blood tests include white blood cell (WBC) counts and absolute neutrophil and absolute lymphocyte counts; the neutrophil-to-lymphocyte ratio (NLR) is calculated as (absolute neutrophils) / (absolute lymphocytes). Some CBC panels may include a calculated NLR ratio.

Normal reference range for WBC: 3.4–10.8 x103/µL.

Normal reference range for NLR: 0.0–2.9

White blood cells (WBCs) are circulating immune cells. They include leukocytes (mainly neutrophils), which are responsible for perpetuating the innate or generalized response to infection or injury; and lymphocytes (mainly T and B cells), which engage in specific responses and the ability to adapt to or learn from one’s unique history of exposures. WBC levels fluctuate rapidly depending on cytokine signaling. In chronic inflammation, prolonged pro-inflammatory cytokine stimulation generally upregulates the innate immune response, increasing not only the total number of WBCs but also the number of neutrophils relative to lymphocytes.413-415

The NLR has emerged as a highly sensitive marker of inflammation, including chronic systemic inflammation. Elevated NLR has been associated with increased risk of a range of chronic disorders including cardiovascular disease, type 2 diabetes, and chronic obstructive pulmonary disease.413 Metabolic syndrome,416 obesity,417 multiple sclerosis,418 psoriasis incidence,419 periodontal disease,420 ankylosing spondylitis421 and a variety of cancers422 are among the other chronic inflammatory conditions correlated with higher NLR. Higher NLR has also been associated with higher all-cause, cancer, and cardiovascular mortality.423-425 In addition, monitoring NLR may provide insights into disease progression.413,422 It is important to note that NLR is highly sensitive to factors such as stress, trauma, gender, and age. Such factors should be taken into consideration when interpreting individual NLR values.426

Emerging research suggests the platelet-to-lymphocyte (PLR) ratio may also be a useful indicator of inflammation, including due to cardiovascular or autoimmune disease.427-430 Another calculation, in which the numbers of neutrophils and platelets are multiplied and their product is divided by the number of lymphocytes ([N x P]/L), is being explored as another possible measure of systemic inflammation. This number is called the systemic inflammation index.431

4 Risk Factors for Chronic Inflammation

There are several risk factors which increase the likelihood of establishing and maintaining a low-level inflammatory response.

Age

In contrast to younger individuals (whose levels of inflammatory cytokines typically increase only in response to infection or injury), older adults can have consistently elevated levels of several inflammatory molecules, especially IL-6 and TNF-α.9 These elevations are observed even in healthy older individuals. While the reasoning for this age-associated increase in inflammatory markers is not thoroughly understood, it may reflect cumulative mitochondrial dysfunction and oxidative damage, or may be the result of other risk factors associated with age (such as increases in visceral body fat or reductions in sex hormones).

Obesity

Fat tissue is an endocrine organ, storing and secreting multiple hormones and cytokines into circulation and affecting metabolism throughout the body. For example, fat cells produce and secrete both TNF-α and IL-6, and visceral (abdominal) fat can produce these inflammatory molecules at levels sufficient to induce a strong inflammatory response.29,30 Visceral fat cells can produce three times the amount of IL-6 as fats cells elsewhere,31 and in overweight individuals, may be producing up to 35% of the total IL-6 in the body.32 Fat tissue can also be infiltrated by macrophages, which secrete pro-inflammatory cytokines. This accumulation of macrophages appears to be proportional to body mass index (BMI), and appear to be a major cause of low-grade, systemic inflammation and insulin resistance in obese individuals.33,34

Diet

A diet high in saturated fat is associated with higher pro-inflammatory markers, particularly in diabetic or overweight individuals.35,36 This effect was absent in healthy individuals.37-39 Diets high in synthetic trans fats (such as those produced by hydrogenation) have been associated with increases in inflammatory markers (IL-6, TNF-α, IL-8, CRP) in some studies,40,41 but had no effect in others.42,43 The increases in markers of inflammation due to synthetic trans fats may be more pronounced in overweight individuals.42

General dietary over-consumption is a major contributor to inflammation and other detrimental age-related processes in the modern world. Therefore, eating a calorie-restricted diet is an effective means of relieving physiologic stressors. Indeed, several studies show that calorie restriction provides powerful protection against inflammation.44,45 For more information about the metabolic benefits of eating fewer calories, readers should refer to the “Caloric Restriction” protocol.

Low Sex Hormones

Amongst their many roles in biology, sex hormones also modulate the immune/inflammatory response. The cells that mediate inflammation (such as neutrophils and macrophages) have receptors for estrogens and androgens that enable them to selectively respond to sex hormone levels in many tissues.46 A notable example is that of osteoclasts, the macrophages that reside in skeletal tissue and are responsible for breaking down and recycling old bone. Estrogens turn down osteoclast activity. Following menopause, lowered estrogen levels cause these bone depleting cells to maintain their activity, breaking down bone faster than it is rebuilt. This is one of the factors in the progression of osteoporosis.

Experiments in cell culture have demonstrated that testosterone and estrogen can repress the production and secretion of several pro-inflammatory markers, including IL-1β, IL-6, TNF-α, and the activity of NF-κB.47-49 These observations have been corroborated by observational studies that have linked lower testosterone levels in elderly men to increases in inflammatory markers (IL-6 and IL-6 receptor).50,51 Several studies have shown an increase in inflammatory IL-1β, IL-6, and TNF-α following surgical or natural menopause.9,52 Conversely, the preservation of sex hormone levels is associated with reductions in the risk of several inflammatory diseases, including atherosclerosis, asthma in women, and rheumatoid arthritis in men.46 Hormone replacement therapy (HRT) may partially exert its protective effects through an attenuation of the inflammatory response. Reductions in the risks of coronary heart disease and inflammatory bowel disease in some individuals, as well as levels of some circulating inflammatory cytokines (including IL-1B, IL-8, and TNF-α) has been observed in some studies of women on HRT.53-55

Smoking

Cigarette smoke contains several inducers of inflammation, particularly reactive oxygen species. Chronic smoking increases production of several pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-8), while simultaneously reducing production of anti-inflammatory molecules.56 Smoking also increases the risk of periodontal disease, an independent risk factor for increasing systemic inflammation.57

Sleep Disorders

Production of inflammatory cytokines (TNF-α and IL-1β) appears to follow a circadian rhythm and may be involved in the regulation of sleep in animals and humans.58 Disruption of normal sleep can lead to daytime elevations of these pro-inflammatory molecules. Plasma levels of TNF-α and/or IL-6 were elevated in patients with excessive daytime sleepiness, including those with sleep apnea and narcolepsy.58 These elevations in cytokines were independent of BMI or age,59,60 although persons with higher visceral body fat were more likely to have sleep disorders.61

Other Inciting Factors

Periodontal disease can produce a systemic inflammatory response that may affect several other systems, such as the heart and kidneys.62,63 It is by this mechanism that periodontal disease is thought to be a risk factor for cardiovascular diseases.64

Stress (both physical and emotional) can lead to inflammatory cytokine release (IL-6); stress is also associated with decreased sleep and increased body mass (stimulated by release of the stress hormone cortisol), both of which are independent causes of inflammation.65

The maintenance of a proper inflammatory response may also involve the central nervous system. The recently identified vagal immune reflex senses inflammatory molecules through a network of nerves (branches of the vagus nerve), and sends this information to the brain. If the brain determines that the inflammatory response is too great, it sends signals to the site(s) of inflammation to attenuate the response.66 Preliminary data suggest depressed nerve activity may be associated with exaggerated inflammatory responses seen in sepsis.67 Smoking, itself a risk factor for inflammation, also decreases activity of the vagus nerve.68

Excess Blood Glucose Fuels Inflammatory Fires

When glucose is properly utilized, our cells produce energy efficiently. As cellular sensitivity to insulin diminishes, excess glucose accumulates in our bloodstream. Like spilled gasoline, excess blood glucose creates a highly combustible environment from which oxidative and inflammatory fires chronically erupt.

Excess glucose not used for energy production converts to triglycerides that are either stored as unwanted body fat or accumulate in the blood where they contribute to the formation of atherosclerotic plaque.

As an aging human, you face a daily onslaught of excess glucose that poses a grave risk to your health and longevity. Surplus glucose relentlessly reacts with your body’s proteins, causing damaging glycation reactions while fueling the fires of chronic inflammation and inciting the production of destructive free radicals.69-71

Avert Glycation and Inflammation by Controlling Glucose Levels with Green Coffee Extract

Unroasted coffee beans, once purified and standardized, produce high levels of chlorogenic acid and other beneficial polyphenols that can suppress excess blood glucose levels. Human clinical trials support the role of chlorogenic acid-rich green coffee bean extract in promoting healthy blood sugar control and reducing disease risk.

Scientists have discovered that chlorogenic acid found abundantly in green coffee bean extract inhibits the enzyme glucose-6-phosphatase that triggers new glucose formation and glucose release by the liver.72,73 Glucose-6-phosphatase is involved in dangerous postprandial (after-meal) spikes in blood sugar.

In another significant mechanism, chlorogenic acid increases the signal protein for insulin receptors in liver cells.74 That has the effect of increasing insulin sensitivity, which in turn drives down blood sugar levels.

In a clinical trial, 56 healthy volunteers were challenged with an oral glucose tolerance test before and after a supplemental dose of green coffee extract. The oral glucose tolerance test is a standardized way of measuring a person's after-meal blood sugar response. In subjects not taking green coffee bean extract, the oral glucose tolerance test showed the expected rise of blood sugar to an average of 144 mg/dL after a 30-minute period. But in subjects who had taken 200 mg of the green coffee bean extract, that sugar spike was significantly reduced, to just 124 mg/dL—a 14% decrease.75 When a higher dose (400 mg) of green coffee bean extract was supplemented, there was an even greater average reduction in blood sugar—up to nearly 28% at one hour.

Maintaining a fasting glucose level of 80 – 86 mg/dL, and that two-hour post-meal glucose levels remain under 125 mg/dL, may help combat chronic inflammation.

5 Diseases Associated with Chronic Inflammation

Cardiovascular Diseases (CVD)

Inflammation is an integral part of atherosclerosis (recall that oxidized low-density lipoprotein cholesterol stimulates the inflammatory response). Circulating inflammatory cytokines are predictive of peripheral arterial disease, heart failure, atrial fibrillation, stroke, and coronary heart disease.9,26

Cancer

Several studies have established links between chronic low-level inflammation and many types of cancer, including lymphoma, prostate, ovarian, pancreatic, colorectal and lung.7,76 There are several mechanisms by which inflammation may contribute to carcinogenesis, including alterations in gene expression, DNA mutation, epigenetic alterations, promotion of tumor vascularization, and the expression of pro-inflammatory cytokines that have roles in cancer cell proliferation.7,77

Diabetes

The infiltration of macrophages into fat tissue and their subsequent release of pro-inflammatory cytokines into circulation occur at a greater rate in type II diabetics than in non-diabetics.33,35,78 Pro-inflammatory cytokines clearly decrease insulin sensitivity.2

Age-Related Macular Degeneration (AMD)

An evaluation of 11 population-based studies encompassing over 41,000 patients demonstrated a clear association between elevated serum CRP levels (> 3 mg/L) and the incidence of late onset AMD.79 The risk of AMD in these high-CRP patients was increased over 2-fold compared with patients with CRP levels < 1 mg/L.

Chronic Kidney Disease (CKD)

The chronic, low-grade inflammation in CKD can lead to the retention of several pro-inflammatory molecules in the blood (including cytokines, AGEs, and homocysteine).6 The reduced excretion of pro-inflammatory factors by the diseased kidney can accelerate the progression of chronic inflammatory disturbances elsewhere in the body, such as the cardiovascular system.

Osteoporosis

Inflammatory cytokines (TNF-α, IL-1β, IL-6) are involved in normal bone metabolism. Osteoclasts, the cells that break down (resorb) bone tissue, are a type of macrophage and can be stimulated by pro-inflammatory factors. Systemic elevations in pro-inflammatory cytokines push bone metabolism towards resorption, and have been observed to induce bone loss in persons with periodontal disease, pancreatitis, inflammatory bowel disease, and rheumatoid arthritis.3 An increase in the levels of inflammatory cytokines is also a mechanism by which menopause stimulates bone loss.

Depression

There is a small, but significant association between elevated IL-6 and CRP in depressed patients, which has been observed in many population studies.80 It is unclear whether inflammation leads to stress or vice versa, and there is data supporting both hypotheses.81,82

Cognitive Decline

Several observational studies have linked chronic low-level inflammation in older adults to cognitive decline and dementia, including vascular dementia and Alzheimer’s disease.9 One study found people with the highest CRP and IL-6 levels (> 2.4 pg/mL) had a ~30‒40% increased risk of cognitive decline compared to those with the lowest levels (< 1.4 pg/mL).83 Inflammatory markers can be elevated before the onset of cognitive dysfunction, indicating their potential relevance as a prognostic tool in high-risk individuals.9

Others

Elevations in circulating inflammatory cytokines are associated with several other conditions, both inflammatory (rheumatoid arthritis, IBD/Crohn’s disease, pancreatitis) and non-inflammatory (anemia, fibromyalgia, frailty, sarcopenia/cachexia/muscle wasting).4,5,84-86 Again, whether inflammation incites these conditions or results from them is unclear, and requires further investigation.

6 Drug Strategies to Combat Chronic Inflammation

Pentoxifylline

Pentoxifylline is a drug used to treat conditions involving poor circulation to the brain, limbs, and other areas perfused by small blood vessels. The drug effectively decreases blood viscosity and improves peripheral tissue oxygenation. Pentoxifylline acts as a phosphodiesterase inhibitor and has immunomodulatory and anti-inflammatory actions.1 Preclinical studies have shown that pentoxifylline modulates TNF-α signaling and enhances nitric oxide synthase activity as well.91,286 Pentoxifylline has been studied in a wide range of conditions, ranging from diabetic complications and non-alcoholic liver disease to endometriosis and cardiac surgery.87-90

In a randomized clinical trial, 400 mg of pentoxifylline taken twice daily for 12 months significantly suppressed hs-CRP, fibrinogen, and TNF-α levels in patients with chronic kidney disease. And subjects’ renal function did not change with treatment whereas the control group worsened.92 Administered at the same dose for one month to 30 diabetic individuals with high blood pressure, pentoxifylline reduced hs-CRP levels by 20% and improved erythrocyte sedimentation rate (a measure of inflammatory tendency of a blood sample) by 18%. Plasma antioxidant status was increased as well, as evidenced by a 20% reduction in malondialdehyde levels (a measure of oxidative stress) and a nearly 5% increase in glutathione levels.95 Pentoxifylline, at dosages ranging from 400 mg once daily to three times per day, has also demonstrated anti-inflammatory effects in clinical studies287-290; a meta-analysis of 15 randomized controlled trials including 739 participants concluded that these dosages of pentoxifylline significantly reduced TNF-α and hs-CRP levels.291 And in a small clinical study, pentoxifylline administered intravenously to patients with sepsis demonstrated some benefit, including decreasing TNF-α levels.292 However, a different small trial with intravenous pentoxifylline did not show a reduction in TNF-α levels, although it did improve cardiopulmonary dysfunction.293 Intravenous pentoxifylline has also been studied in septic infants, and has generally been found to offer some benefits as an adjunct to antibiotic treatment.294,295

Cabergoline

Cabergoline, a derivative of ergot, binds the D2 dopamine receptor leading to sustained decreases in prolactin secretion from the pituitary gland. It is used as a first-line treatment for pituitary adenomas and has been used in the treatment of Parkinson disease.296 Cabergoline has been reported to have anti-inflammatory activity; in an animal study, cabergoline protected the blood–brain barrier and reduced lipopolysaccharide-induced gene expression of TNF-α and IL-6.297

Prolactin levels have been linked to inflammatory status in males,298 and high prolactin levels may be a risk factor for delirium in patients with sepsis, a condition that involves a pronounced systemic inflammatory response.299 Prolactin levels also appear to be increased in synovial and periodontal tissues in patients with rheumatoid arthritis and periodontitis, respectively—two common chronic inflammatory diseases.300

A clinical trial in 15 patients with hyperprolactinemia found that 12 weeks of cabergoline treatment at 0.5–1 mg twice weekly reduced hs-CRP, prolactin, and insulin levels.301 In another small clinical study, 21 patients with prolactinoma were given 0.5 mg/day cabergoline and tapered as necessary over six months. Insulin sensitivity, homocysteine, and hs-CRP all improved, although independently of prolactin levels.302 Further clinical studies are necessary to elucidate prolactin and cabergoline’s potential roles in inflammatory conditions.

Metformin

The regulation of energy metabolism and inflammation are closely associated; this is evidenced by the co-incidence of metabolic disorders (obesity, diabetes) and low-grade inflammation.96 Metformin may reduce the activity of inflammatory cytokines by increasing the production of IL-1βreceptor antagonist (IL1Rn), a protein factor which interferes with pro-inflammatory signaling of IL-1β.97 It may also promote favorable CRP levels, although not to the same extent as weight loss.96,98 A randomized controlled trial of hypertensive and dyslipidemia patients taking 1,700 mg/day of metformin for 12 weeks demonstrated a 26.7% reduction in IL-6 and 8.3% reduction in TNF-α from baseline levels, a degree of reduction similar to that of the potent statin drug rosuvastatin (Crestor).99 The anti-inflammatory effects of metformin appear to be rapid; reductions in circulating TNF-α, IL-1β, CRP, and fibrinogen were observed after only 30 days in a larger study of 128 type II diabetic patients with dyslipidemia.100

Aspirin

Aspirin has been used as an anti-inflammatory therapy long before the molecular mechanics of inflammation had been discovered; it is now well characterized as an inhibitor of cyclooxygenase enzymes. The modification of COX molecules by aspirin has important implications for cardiovascular health. Blood platelets use cyclooxygenase to produce thromboxane A2, a pro-inflammatory molecule that is an important signal during the initial stages of the clotting process. The inhibitory effect of aspirin on COX enzymes in platelets can partially explain its protective effects against the complications of several disorders, including hypertension, heart attack, and stroke.101 Aspirin's inhibition of cyclooxygenase also helps explain its potential effect on cancer risk reduction as observed in several studies,102-105 as COX-2 also appears to have roles in increasing the proliferation of mutated cells, tumor formation, tumor invasion, and metastasis, and may contribute to drug resistance in some cancers.106 Aspirin has also been shown to reduce the activity of NF-kB in vitro,107 and lower levels of multiple inflammatory markers (TNF-α, CRP, IL-6) in patients with cardiovascular disease.108-111

Unlike many other NSAIDs, the effects of aspirin on COX enzymes are permanent for the life of the COX enzyme. Interestingly, it appears that rather than rendering the enzyme inactive, aspirin modifies the function of COX. Aspirin stops the enzyme from producing pro-inflammatory prostaglandins, and enables it to begin producing anti-inflammatory molecules called resolvins.112

Low-Dose Statin Drugs

Statins are thought to reduce inflammation by a mechanism distinct from their effects on cholesterol metabolism; they interfere with the function of cytokine receptors on the surface of white blood cells. Therefore, pro-inflammatory signals in the blood are unable to provoke a response from white blood cells, and they are prevented from further stimulating inflammation.113,114 Results of the JUPITER trial presented the strongest evidence for statins as anti-inflammatory therapy. In this study of over 17,000 healthy middle-aged men and women with elevated levels of the inflammatory marker CRP but normal levels of blood lipids, 20 mg/day of rosuvastatin reduced CRP levels by over half, in addition to reducing heart attack and stroke incidence.115 Smaller studies have looked at the effect of statins on other inflammatory markers as well. A randomized controlled trial of hypertensive and dyslipidemia patients taking a lower dose (10 mg/day) of rosuvastatin for 12 weeks demonstrated a ~22% reduction in IL-6 and 13% reduction in TNF-α from baseline levels.99 A second uncontrolled study of simvastatin demonstrated more modest reductions in IL-6, but no changes in TNF-α from the statin treatment.116 To generate a substantial anti-inflammatory effect using statin drugs alone requires a high dose that is more likely to induce side effects than lower dose statin therapy.

7 Dietary and Lifestyle Approaches to Reduce Chronic Inflammation

Inflammation itself is not a disease, but is featured, to varying degrees, in adverse health conditions. Information on strategies and research regarding the reduction of inflammation characteristic to specific health conditions are featured in their respective Life Extension protocols: "Allergies," "Macular Degeneration," "Cancer Adjuvant Therapy," "Atherosclerosis and Cardiovascular Disease," "Gout and Hyperuricemia," "Inflammatory Bowel Disease (Crohn’s and Ulcerative Colitis)," "Osteoarthritis," "Arthritis–Rheumatoid," and "Osteoporosis." What follows is a summary of dietary and supplemental approaches to addressing general chronic inflammation and para-inflammation. As many types of general inflammation often occur without additional symptoms, most of the strategies listed below are based on their ability to reduce circulating inflammatory cytokines, the hallmark of the para-inflammatory state.

Macronutrients and Energy Balance

Macronutrient content (particularly the types and levels of carbohydrates and fats) can have a significant effect on the progression of inflammation (as measured by increases in pro-inflammatory markers). Diets with relatively high glycemic index (GI) and glycemic load (GL) have been associated with elevated risk of coronary heart disease, stroke, and type 2 diabetes mellitus, particularly among overweight individuals, and have been associated with modest increases in proinflammatory markers in multiple studies.117 In a study of over 18,000 healthy women ≥45 years old without diagnosed diabetes, high GI and GL diets resulted in a small but significant increase in hs-CRP (+12% for high GI) over low GI diets.118 In the Danish Hoorne study,119 for every 10 unit increase in dietary glycemic index, circulating CRP was increased by 29%. As discussed previously, some dietary fats (particularly saturated and synthetic trans fats) increase inflammation occurrence, while omega-3 polyunsaturated fats appear to be anti-inflammatory.40

Since fat tissue (especially abdominal fat) expresses inflammatory cytokines, obesity can be a major cause of low-grade, systemic inflammation.33,34 Thus, it is important that total energy intake be proportional to energy expenditure, to avoid the deposition of abdominal fat. Obesity-induced increases in inflammatory cytokines appear to be reversible with fat loss.120 In a dramatic example, weight loss (by adjustable gastric banding) in a group of 20 severely obese individuals reduced IL-6 by 22% and CRP by almost half.121

An inflammatory index, developed by a group from the Arnold School of Public Health at the University of South Carolina, scored 42 common dietary constituents based on their ability to raise serum CRP.122 Constituents (such as saturated fat, tea polyphenols, or vitamin D) were given either a positive (anti-inflammatory) or negative (pro-inflammatory) score, the magnitude of which was weighted based on the volume of inflammation research on the isolated ingredient. Human clinical data was weighted more than animal data, and clinical trials more than observational studies. The scores were then verified by comparing them to nutrient intakes and CRP levels from a group of 494 volunteers over the course of one year. Amongst the most anti-inflammatory nutrients (based on the model and study data) are magnesium, beta-carotene, turmeric (curcumin), genistein, and tea; the most pro-inflammatory included carbohydrates, total- and saturated fat, and cholesterol. The index may provide a useful metric for accessing the overall inflammatory potential of an individual diet.

Exercise

Energy expenditure through exercise lowers multiple cytokines and pro-inflammatory molecules independently of weight loss. While muscle contraction initially results in a pro-inflammatory state, it paradoxically lowers systemic inflammation. This effect has been observed in dozens of human trials of exercise training in both healthy and unhealthy individuals across many age groups.123

Fiber

In an analysis of seven studies on the relationship between weight loss and hs-CRP, increased fiber consumption correlated with significantly greater reductions in hs-CRP concentrations. In these studies, daily fiber intakes ranging from 3.3 to 7.8 grams/MJ (equivalent to about 27 to 64 grams/day for a standard 2,000 kcal diet) reduced CRP from 25%‒54% in a dose-dependent fashion. These results should be interpreted carefully, as only two of the seven studies were specifically designed to examine the effects of fiber independently.120 The Women’s Health Initiative failed to detect an effect of fiber consumption on hs-CRP, but found that greater intake of dietary soluble and insoluble fiber (over 24 grams/day) was associated with lower levels of IL-6 and TNF-α.124

8 Nutrients

Black Cumin Seed Oil

Reported dosage: Approximately 1,300–2,300 mg daily

Black cumin (Nigella sativa) has a long history of use as a culinary herb and a traditional medicine for treating a wide range of conditions such as headaches, joint pain, back pain, asthma, skin problems, and high blood pressure. Research suggests black cumin seed, its oil, and its main active constituent thymoquinone can help balance immune and inflammatory responses and have antioxidant, antimicrobial, and anticancer properties.432

Numerous randomized controlled trials have confirmed the anti-inflammatory effect of black cumin. For example, in one randomized controlled trial involving 106 subjects with osteoarthritis of the knee, 2.5 mL of black cumin seed oil (about 2.3 grams) three times daily for one month improved pain and arthritis index scores and reduced the need for pain medications better than placebo.433 In another randomized placebo-controlled trial in 60 patients with coronary artery disease, 2 grams of black cumin seed oil daily for eight weeks reduced blood levels of adhesion molecules, which play a key role in immune cell migration during the inflammatory process compared with a sunflower oil placebo.434

In a meta-analysis of 20 randomized controlled trials involving a total of 1,086 participants, black cumin (mean dosage approx.1,745 mg/day) was found to reduce levels of CRP and TNF-α, but not IL-6, and improve indicators of oxidative stress.435 Another meta-analysis pooled findings from 16 randomized controlled trials involving 1,033 subjects with metabolic syndrome and associated cardiometabolic conditions (eg, obesity, type 2 diabetes, hypertension). The analysis found black cumin, at daily doses ranging from 900–2,300 mg, decreased levels of CRP, TNF-α, and IL-6, and reduced oxidative stress.436 An umbrella analysis that incorporated findings from seven meta-analyses found, overall, black cumin effectively reduced CRP and TNF-α levels and improved markers of oxidative stress. The dosages used in the studies in the included meta-analyses ranges from 1,300–1,900 mg daily.437 Furthermore, multiple clinical trials have reported black cumin can improve markers of cardiometabolic health, including blood lipids and markers of glycemic control. Dosages reported in the trials vary widely, but the median reported dosage was about 2,000 mg/day.438

Probiotics

Reported dosage: Approximately 4 billion to 35 billion colony forming units (CFUs) daily

Healthy gut bacteria regulate local and systemic immune function by interacting with epithelial and immune cells in the intestinal walls.439 They also produce short-chain fatty acids (SCFAs) that promote balanced immune function. SCFAs like butyrate, propionate, and acetate can affect immune cell activity by suppressing expression of inflammatory cytokines and increasing expression of anti-inflammatory cytokines.440 Probiotic supplements contain microorganisms that can have similar beneficial effects and encourage a healthy gut microbiome.441,442 An umbrella analysis of findings from 39 meta-analyses of randomized controlled trials found treatment with probiotics reduced CRP, TNF-α, and IL-6 levels in patients with a variety of chronic inflammatory disorders. Most of the trials used supplements containing mixed probiotic strains of Lactobacillus, Bifidobacterium, and/or Streptococcus species and used daily doses between about 4 billion and 35 billion CFUs.443

Resveratrol and Pterostilbene

Reported dosage: 500–3,000 mg daily

Resveratrol is a type of polyphenolic compound known as a stilbene. It has demonstrated antioxidant, anti-inflammatory, and anti-aging activities that have been attributed to its ability to regulate cell metabolism.444 Pterostilbene is a naturally occurring resveratrol analog that has shown similar properties in preclinical research.445 Resveratrol inhibits pro-inflammatory eicosanoid production and NF-kappaB signaling, downregulating inflammasome and cytokine signaling.444,446 It also appears to improve gut microbiome composition and gut barrier function.444,447

A meta-analysis that included findings from 35 randomized controlled trials with a total of 1,128 participants found resveratrol reduced hs-CRP and CRP levels, and was more effective when used for 10 weeks or longer and at daily doses of 500 mg or more.448 Furthermore, an umbrella meta-analysis that pooled data from 19 meta-analyses involving a total of 4,088 participants found resveratrol lowered CRP and TNF-α levels but had no significant effect on IL-6 levels. The analysis also showed treatment with at least 500 mg of resveratrol daily for at least 12 weeks led to weight loss, which may have been related to reduced inflammatory signaling.449

Resveratrol may have anti-inflammatory effects in people with specific chronic inflammatory disorders. A meta-analysis that pooled findings from six randomized controlled trials in subjects with cardiovascular disease found treatment with resveratrol lowered levels of CRP and TNF-α, but not IL-6, and only when used at doses higher than 15 mg daily.450 Similarly, meta-analyses of randomized controlled trials have shown resveratrol reduced hs-CRP and TNF-α (but not IL-6) levels in patients with metabolic-associated fatty liver disease (MAFLD)451 and those with metabolic syndrome.452 Another meta-analysis combined findings from six randomized controlled trials involving a total of 533 subjects with type 2 diabetes and found resveratrol, at doses of 40–1,000 mg daily, reduced CRP levels but did not change IL-6 or TNF-α levels.453

Curcumin

Reported dosage: 250–3,000 mg curcumin daily

Curcumin is a potent antioxidant carotenoid found in turmeric. Its anti-inflammatory effects are well established and attributable in part to its ability to suppress NF-kappaB, thereby downregulating inflammatory cytokine signaling.454 Numerous clinical trials have indicated curcumin may be helpful in treating a variety of chronic inflammatory disorders, particularly metabolic diseases, osteoarthritis, and rheumatic conditions.455

A systematic review and meta-analysis that included data from 66 randomized controlled trials found curcumin decreased levels of most biomarkers of inflammation; specifically, curcumin lowered CRP, IL-6, and TNF-α, but not IL-1β levels. In general, trials that used more absorbable forms of curcumin used lower doses (80–120 mg per day), whereas most trials that used common curcumin extracts used higher doses (250–3,000 mg per day).456 In a meta-analysis of 20 randomized controlled trials involving 1,394 subjects with metabolic syndrome, nano-curcumin (a form developed for increased absorbability), at doses of 80–180 mg daily, was found to decrease levels of hs-CRP, IL-6, and TNF-α. Notably, the analysis also found nano-curcumin improved markers of cardiovascular and metabolic health.457 Curcumin was also found to reduce inflammatory biomarker levels in two meta-analyses of trials in chronic kidney disease patients, including those requiring hemodialysis.458,459 A meta-analysis of six randomized controlled trials in patients with rheumatoid arthritis and ulcerative colitis showed curcumin, at doses of 250–1,500 mg per day for 8–12 weeks, reduced CRP levels.460 A randomized placebo-controlled trial in 70 patients with SLE found 1,000 mg of curcumin daily for 10 weeks reduced levels of IL-6, but not other inflammatory markers.461

Omega-3 PUFAs

Reported dosage: 900–2,000 mg daily

Fish oil is a rich source of the omega-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). These fatty acids can be incorporated into cell membranes and used to synthesize anti-inflammatory eicosanoids (prostaglandins and leukotrienes) as well as SPMs, which have a vital role in resolving inflammatory reactions.462,463 Omega-3 PUFAs also exert anti-inflammatory effects by modulating the gut microbiome.462 EPA and DHA supplements have been shown to improve outcomes in clinical trials involving patients with a wide range of inflammatory disorders, such as coronary artery disease, type 2 diabetes, obesity, metabolic-associated liver disease, Alzheimer disease, SLE, and rheumatoid arthritis.464-466

Higher fish oil or omega-3 PUFA intake has been associated with lower inflammatory marker levels, in observational research.467 One study in 402 subjects found a higher omega-6:omega-3 ratio was associated with higher CRP levels in those with chronic pain.468 Another study involving 101 subjects with advanced coronary artery disease prior to surgery found higher intake of omega-3 PUFAs, and a lower ratio of omega-6:omega-3 PUFAs, was correlated with a lower NLR, as well as a lower systemic inflammatory index value (a number calculated by multiplying the numbers of neutrophils by platelets and dividing by lymphocytes).469 In 105 men who had already undergone surgery for coronary artery disease and completed a dietary intake questionnaire, higher omega-3 PUFA intake was linked to lower PLR, but only those with a lower omega-6:omega-3 ratio had reduced CRP levels.470 Large observational studies have also reported correlations between higher omega-3 intake and lower levels of traditional inflammatory biomarkers.168,169 In one study that included 2,920 Greek men and women without known cardiovascular disease, participants who consumed over 300 grams (about 10 oz.) of fish per week had, on average, 33% lower CRP, 33% lower IL-6, and 21% lower TNF-α levels than participants who did not consume fish.170 In a sample of 5,677 men and women without cardiovascular disease participating in the Multi-Ethnic Study of Atherosclerosis (MESA), higher EPA and DHA intake was associated with reduced plasma concentrations of multiple inflammatory markers, including hs-CRP, IL-6, and soluble TNFR1.171

Numerous randomized controlled trials and multiple meta-analyses have shown omega-3 fatty acid supplementation can improve inflammatory marker levels. For example, in a randomized controlled trial in 60 patients receiving standard treatment following a heart attack, adding 2 grams of omega-3 PUFAs per day to treatment for 30 days lowered hs-CRP levels compared with placebo.475 A large meta-analysis of 48 randomized controlled trials including 8,489 subjects with metabolic syndrome found omega-3 PUFA supplementation led to decreased IL-1, IL-6, CRP, and TNF-α levels and reduced some cardiovascular risk factors. The analysis also showed the effects were stronger when the dose was 2 grams or less and increased over time with ongoing use.476 A meta-analysis of 13 clinical trials involving 853 patients with coronary artery disease found supplementing with omega-3 fatty acids reduced hs-CRP, IL-6, and TNF-α levels and improved blood vessel function.477 Another meta-analysis of 10 randomized controlled trials investigating the effects of 0.9–8 grams of fish oil per day in heart failure patients found supplementing with omega-3 fatty acids, particularly at doses ≤ 2 grams per day, reduced IL-6 and TNF-α levels, but not CRP or IL-1 levels.478

Specialized Pro-Resolving Mediators

Specialized pro-resolving mediators (SPMs) are cell-signaling molecules derived from the metabolism of PUFAs. They include resolvins, protectins, maresins, and others derived from the omega-3 PUFAs eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and docosapentaenoic acid (DPA), as well as lipoxins derived from the omega-6 PUFA arachidonic acid. Rather than decreasing inflammation, SPMs actively resolve inflammatory processes and help return tissues to a normal state.463,479 Resolution is achieved by recruiting macrophages to clear cellular debris and pathogens, promoting wound healing and tissue regeneration, and restoring the balance between anti-inflammatory and pro-inflammatory immune cell activity. Failure to resolve persistent inflammation, such as through using anti-inflammatory drugs, can result in scarring and tissue dysfunction.479,480

Clinical trials have shown supplementing with, or increasing intake of, omega-3 fatty acids can raise SPM and SPM-precursor levels in subjects with various health conditions as well as in healthy individuals.481-485,651 One trial, which used a crossover design, involved 21 subjects with chronic inflammation, nine of whom were men over 50 years of age and 12 were postmenopausal women. After a four-week washout period in which the subjects received sunflower oil to establish a baseline, they transitioned into the crossover phase of the study. Subjects received three grams of EPA per day in one 10-week experimental phase and 3 grams of DHA per day in another 10-week experimental phase, in random order, with a 10-week washout in between. The primary outcome was to assess how subjects’ immune cells (monocytes) responded to an inflammatory stimulus (a bacterial toxin called lipopolysaccharide [LPS]) in the lab. Blood levels of pro- and anti-inflammatory cytokines did not change during the study; however, after each supplement phase, monocytes taken from the participants’ blood samples showed reduced pro-inflammatory cytokine gene expression in response to an inflammatory stimulus in the lab: EPA decreased expression of the gene associated with the pro-inflammatory cytokine TNF-alpha, while DHA decreased expression of genes related to pro-inflammatory cytokines TNF-alpha, IL-6, and MCP-1 (monocyte chemoattractant protein-1), as well as the anti-inflammatory cytokine IL-10. Moreover, EPA and DHA were each found to increase plasma levels of SPMs and SPM derivatives.270 Interestingly, the effects of supplementation on SPM levels were significantly stronger in male than female participants.487

A randomized controlled trial in 40 healthy young individuals showed consuming omega-3 fatty acid-enriched hen eggs not only increased SPM levels but also decreased the inflammatory capacity of immune cells compared with eating regular hen eggs. In the trial, participants ate three hard-boiled eggs per day for three weeks: one group ate regular eggs and the other ate enriched eggs providing approximately 1,053 mg of omega-3 fatty acids (~45 mg EPA, 315 mg DHA, and 690 mg of alpha-linolenic acid) per day. At the end of the trial, blood levels of SPMs were higher in the enriched egg group. In addition, immune cells retrieved from participants’ blood samples showed eating the omega-3 fatty acid-enriched eggs, but not regular eggs, led to greater secretion of the immune response modulator tumor growth factor (TGF)-beta-1 and less secretion of the pro-inflammatory cytokine IL-6 following activation with an immune cell stimulant.652

The effects of SPMs on traditional markers of inflammation are not yet fully understood. In a preliminary trial in 44 patients with chronic pain, participants took 1,500 mg per day of a marine lipid supplement standardized to have high levels of SPM precursors for two weeks. Doses were then adjusted to 1,000 or 2,000 mg per day, depending on changes in subjective pain levels, for two more weeks. At the end of the study, measures of pain intensity, pain interference, anxiety, depression, and overall quality of life were improved, but hs-CRP levels were unchanged.488 Similarly, in a randomized placebo-controlled trial in 24 peripheral artery disease patients, 4.4 grams of fish oil per day for three months improved the omega-3 index and increased production of SPMs and SPM precursors, but did not reduce hs-CRP levels.489 In 21 older subjects with chronic inflammation, higher DHA concentrations (indicating greater intake) were associated with lower IL-6 and TNF-alpha levels. Interestingly, they were also associated with lower levels of anti-inflammatory IL-10. In addition, higher levels of the SPM precursors made from DHA were associated with lower levels of an inflammation activator called monocyte chemoattractant protein-1 (MCP-1).490

Vitamin D

Reported dosage: 12.5–100 mcg (500–4,000 IU) daily

Vitamin D appears to exert anti-inflammatory activity by lowering inflammatory cytokine levels, reducing pro-inflammatory prostaglandins, and inhibiting activation of NF-kappaB.491 Multiple meta-analyses of randomized controlled trials have shown vitamin D supplementation can reduce CRP and TNF-α levels in various populations.492,493 Vitamin D was found to significantly reduce hs-CRP and TNF-α levels and marginally lower IL-6 levels in a meta-analysis of randomized controlled trials involving type 2 diabetes patients. Some trials in this analysis used periodic or single high doses of vitamin D, whereas others used lower daily doses. In general, trials in which inflammatory markers decreased used dosing schemes averaging 12.5–100 mcg (500–4,000 IU) per day.494 In a meta-analysis that included eight trials in 592 patients with cancer or precancerous lesions, vitamin D was found to decrease TNF-α levels. It also reduced CRP and IL-6 levels, although these changes were not statistically significant. All but one of the studies in this analysis used doses averaging 20–100 mcg (800–4,000 IU) daily.495 One meta-analysis that pooled findings from six randomized controlled trials involving 758 postmenopausal women found, at doses averaging 10–70 mcg (400–2,800 IU) daily, vitamin D reduced CRP levels. The analysis also showed the effect was strongest in trials lasting three months or longer, trials that used ≤ 25 mcg (1,000 IU) of vitamin D per day, and trials that used vitamin D3.496 A meta-analysis of 11 trials in a total of 3,049 patients with rheumatoid arthritis found vitamin D at doses higher than 100 mcg (4,000 IU) daily had a more significant CRP-lowering effect than lower doses.497

Multinutrient Supplementation Targets Inflammation in Older Adults

A randomized double-blind trial at a single center in Germany enrolled 112 healthy adults aged 70 years and older and tested a daily multimicronutrient plus omega-3 supplement for 12 weeks.498 The supplement contained, among other nutrients, 400 mcg folic acid, 100 mcg vitamin B12, 50 mcg (2,000 IU) vitamin D3, and 1,000 mg omega-3 fatty acids (500 mg EPA + 500 mg DHA) per day. In an exploratory analysis, researchers assessed the effects of this multinutrient intervention on biomarkers of low-grade inflammation. The primary outcome was the INFLA score, a composite index derived from C-reactive protein, white blood cell count, platelet count, and the granulocyte-to-lymphocyte ratio; scores range from −16 (least inflammation) to +16 (most inflammation). After 12 weeks, the supplement group’s INFLA score decreased compared with placebo, indicating their inflammatory profile shifted toward lower low-grade inflammation.

The intervention also improved nutrient status. The omega-3 index (EPA+DHA as a percentage of red blood cell fatty acids) increased from 6.0% to 7.9% in the supplement group yet remained unchanged in the placebo group, and the proportion of participants below the desirable omega-3 index threshold (<8%) decreased from 91% to 59% in the supplement group. Vitamin D, folate, and B12 status also rose only with supplementation. The size of the inflammation score change tracked with changes in fatty acids: for every 1% increase in omega-3 index, the INFLA score dropped by about 1.55 points, whereas increases in red blood cell arachidonic acid were linked to higher INFLA scores. Benefits were larger in older participants and in those whose usual diets were more pro-inflammatory at baseline.

Key limitations in this trial included the modest sample size, short 12-week duration, lack of validated INFLA score cut-offs to translate score changes into specific clinical risk categories, exclusion of very low CRP values due to the assay’s detection limit, and a relatively healthy, highly educated cohort; factors that may limit generalizability.

Vitamin E

Reported dosage: 400–1,800 IU alpha-tocopherol or 250 mg tocotrienols daily

Vitamin E is a fat-soluble antioxidant that protects lipids throughout the body from free radical damage and reduces inflammation.499 A meta-analysis that included findings from 26 randomized controlled trials with a total of 2,102 participants found vitamin E reduced CRP levels. Vitamin E was also found to lower IL-6 levels when alpha-tocopherol form of vitamin E was used and in trials involving subjects with insulin resistance. It also reduced TNF-α levels when used at doses ≥ 700 mg (approximately 1,000–1,500 IU, depending on the type of vitamin E) per day. Most of the trials in the analysis used doses of 400–1,800 IU.500 In a meta-analysis of seven randomized placebo-controlled trials with a total of 504 participants, supplementation with vitamin E plus omega-3 fatty acids reduced hs-CRP levels. Along with 400 IU of vitamin E, treatment groups in five trials received 400 mg of alpha-linolenic acid from flaxseed oil and in the other two trials they received 1,000 or 4,000 mg of omega-3 fats from fish oil.501 A systematic review found the tocotrienol fraction of vitamin E, at 250 mg daily, reduced markers of inflammation; specifically, CRP and IL-1 levels were reduced with tocotrienol supplementation.502

Vitamin C

Reported dosage: 400–2,000 mg oral vitamin C daily

Vitamin C is an antioxidant nutrient with an important role in immune regulation and tissue repair. Insufficient and deficient vitamin C levels have been linked to elevated hs-CRP levels in observational studies.503-506 One study compared outcomes in 42 patients hospitalized with sepsis (systemic infection) who received standard care plus intravenous vitamin C (3 grams once daily for three days) to 42 similar patients who received standard care alone. Those who received vitamin C had lower levels of hs-CRP and TNF-α, less heart muscle damage, and lower risk of death (9.52% vs. 29.27%).507

Multiple clinical trials have indicated vitamin C can reduce inflammatory marker levels. A meta-analysis of 12 studies with a total of 893 participants found that vitamin C supplementation reduced CRP levels. A detailed analysis found the effect was stronger in clinical trials that measured hs-CRP, in subjects with baseline CRP levels of ≥3 mg/L, in subjects aged <60 years, and in trials that used intravenous vitamin C.508 In a randomized, controlled, crossover trial, 58 healthy young adults exposed to air pollution in a large Chinese city received 2,000 mg vitamin C daily and placebo, each for one week, in random order. Compared with placebo, vitamin C reduced CRP levels by 34.01%, TNF-α levels by 17.30%, and IL-6 levels by 19.47%.509 A small randomized placebo-controlled trial in 24 elderly women engaged in an exercise training program found 1,000 mg of vitamin C daily for six weeks lowered IL-6 gene expression levels yet increased expression of the anti-inflammatory cytokine IL-10 compared with placebo, although these effects were not statistically significant.510 A meta-analysis of six randomized controlled trials with 111 participants found vitamin C supplementation, at doses ranging from 400–1,500 mg daily, mitigated an acute rise in IL-6 levels brought on by a single bout of exercise; however, pooled findings from three randomized controlled trials with a total of 70 participants did not find a significant effect on CRP levels.511

Zinc

Reported dosage: 15–100 mg elemental zinc daily

Zinc is a critical nutrient for balanced immune function, and a growing body of research indicates that zinc has a role in downregulating NF-kappaB signaling.512 Low zinc status has been associated with high CRP levels in multiple observational studies.513,514 One report examined the effect of zinc supplementation on immune parameters (including IL-6, CRP, and hs-CRP levels) by performing a series of meta-analyses on data from 35 randomized controlled trials involving 1,995 subjects. The analyses found zinc lowered CRP, hs-CRP, and TNF-α levels. It also showed zinc supplementation lowered IL-6 levels, but only when used at doses of 50 mg per day or less. Finally, zinc reduced circulating neutrophils without affecting numbers of lymphocytes, indicating a reduction in the NLR. Although the trials included in this report used a variety of forms and doses of zinc, most used doses providing 15–100 mg of elemental zinc per day.515 Two other meta-analyses that included findings from randomized controlled trials involving subjects with type 2 diabetes showed zinc supplementation reduced CRP levels and improved other markers of cardiometabolic health.516,517 Another meta-analysis included 75 randomized controlled trials: by pooling findings from 24 trials, the analysis found zinc supplementation reduced CRP levels. Furthermore, pooled findings from 15 trials showed zinc decreased levels of IL-6 (in trials using 30 mg zinc or more) and TNF-α (in trials lasting up to eight weeks). Meta-analysis of other trials showed zinc also improved some markers of oxidative stress and cardiovascular risk.518

Carotenoids

Reported dosage: 4–30 mg lycopene, 10–20 mg lutein, 5 mg zeaxanthin, 4–20 mg astaxanthin, or 30 mg crocin daily

Carotenoids are a family of lipid-soluble phytonutrients that provide pigmentation to plants and have important antioxidant, anti-inflammatory, and anti-aging properties.519 Carotenoids can suppress NF-kappaB pathways, reducing pro-inflammatory cytokine levels. They also support intestinal health and function and improve microbiome balance, which may further decrease inflammation.520,521 Some of the most studied dietary carotenoids are519,522:

  • Beta-carotene, mainly from yellow and orange vegetables
  • Lycopene, mainly from tomatoes
  • Lutein, mainly from egg yolk and green leafy vegetables
  • Astaxanthin, mainly from pink to red seafood (salmon and crustaceans) and algae extracts

A large systematic review included findings from 17 correlational studies, 12 clinical trials involving dietary interventions, and nine clinical trials involving supplements. The review determined carotenoids, in general, decreased levels of inflammatory biomarkers and reduced cardiovascular risk, and dietary sources may have been more effective than supplements.522 A meta-analysis that pooled findings from 26 randomized controlled trials found carotenoid supplements led to reduced levels of CRP and IL-6, but not TNF-α; however, a more detailed analysis found only lycopene reduced IL-6 levels, whereas lutein, zeaxanthin, astaxanthin, and cryptoxanthin reduced CRP levels.523 Dosages of carotenoids used in the studies summarized in the two meta analyses just described were 5–30 mg/d for lycopene, 10–20 mg/d for lutein, 5 mg/d of zeaxanthin, 15–22 mg/d of beta-carotene, and about 16 mg/d of alpha-carotene. Another meta-analysis included 17 controlled trials investigating the effects of lycopene on cardiovascular disease markers. The analysis showed lycopene, at doses ranging from 4–30 mg per day, lowered levels of IL-6, but not CRP, and improved several other cardiovascular risk factors.524 A separate meta-analysis that included data from 12 randomized controlled trials showed that astaxanthin, another carotenoid, reduced IL-6 levels in people with type 2 diabetes. This analysis also showed that astaxanthin reduced some markers of oxidative stress, such as malondialdehyde, with this effect more pronounced in those with type 2 diabetes. In trial trials included in the meta-analysis, astaxanthin dosage ranged from 4 mg to 20 mg daily and the trial duration ranged from four weeks to 12 months.525

Crocin is a carotenoid from saffron (Crocus sativus) that has demonstrated positive health effects related to its anti-inflammatory and free radical-scavenging properties. A meta-analysis that included data from 13 randomized controlled trials determined crocin was effective for lowering CRP, IL-6, and TNF-α levels, and the effects were stronger when doses of at least 30 mg per day were used for at least 12 weeks.526 For example, in randomized controlled trials, 15 mg of crocin twice daily for 12 weeks resulted in lower IL-6 and TNF-α levels compared with placebo in patients with chronic obstructive pulmonary disease (COPD) and in women with polycystic ovary syndrome (PCOS).527,528 The same crocin intervention was found to reduce hs-CRP and TNF-α levels, as well as NF-kappaB activity, in a randomized placebo-controlled trial in 50 participants with type 2 diabetes.529

Polyphenols

Reported dosage: Greater than 450 mg cocoa flavanols or 300–1,500 mg green tea extract daily

Polyphenols are a large family of plant compounds (including flavonoids) that are well-known for their free radical-scavenging and anti-inflammatory properties. Tea, coffee, berries, and other fruits are among the most widely consumed sources of polyphenols; and diets rich in these foods have been associated with lower levels of CRP, IL-6, and IL-8.530-532 Preclinical research has shown dietary polyphenols have anti-inflammatory actions: they suppress NF-kappaB, thereby reducing inflammatory cytokine signaling; they inhibit the cyclooxygenase enzymes COX1 and COX 2, thereby limiting synthesis of pro-inflammatory eicosanoids; they decrease migration of white blood cells; and they modulate gut microbiome and improve gut barrier function.533,534

Higher intake of dietary polyphenols was linked to lower levels of CRP and fibrinogen in an observational study that used data from over 9,000 adults.535 Drinking a polyphenol-rich tea made from yerba mate (Ilex paraguariensis) for eight days lowered IL-6 and TNF-α levels compared with eight days of drinking water in a crossover trial involving nine men.536

Cocoa is a source of polyphenols known as flavanols. A meta-analysis of data from 33 controlled trials indicated dark chocolate or cocoa interventions, such as dark chocolate, cocoa powder, and cocoa beverages, reduced CRP levels when the amount of cocoa flavanols exceeded 450 mg per day, particularly in non-healthy subjects.537

Green tea is high in polyphenols called catechins. A randomized placebo-controlled trial in 28 postmenopausal women with overweight or mild obesity found supplementing with 150 mg of a polyphenol-rich green tea extract twice daily for 60 days lowered CRP levels and improved some markers of metabolic health.538 One meta-analysis included five randomized controlled trials that investigated changes in CRP levels in 383 subjects with type 2 diabetes; the analysis found CRP levels dropped 5.51 mg/dL more in those treated with green tea, at daily doses of 500–1,500 mg, than in those who served as controls.539 A separate meta-analysis of findings from 11 randomized controlled trials found epigallocatechin-gallate (EGCG), the main active polyphenol in green tea, had no effect on CRP levels, regardless of dose, duration, or participant health status.540

Boswellia

Reported dosage: 100–1,000 mg Boswellia extract or about 133–800 mg boswellic acids daily

Boswellia (Boswellia serrata), also known as frankincense, is a traditional anti-arthritis herb used in Ayurvedic medicine. Boswellic acid found in the gum resin has been found to modulate activity of lipoxygenase enzymes, shifting fatty acid metabolism in favor of less-inflammatory eicosanoids and anti-inflammatory SPMs.541

In a randomized controlled trial that included 70 subjects with osteoarthritis of the knee, 100 mg daily of a Boswellia gum resin extract for 30 days reduced hs-CRP and TNF-α levels and decreased joint stiffness and pain.542 A randomized placebo-controlled trial in 47 patients hospitalized with a severe, acute, viral respiratory infection found treatment with 333 mg of Boswellia extract (standardized to provide 133 mg of boswellic acids) three times daily reduced multiple markers of inflammation, including NLR and CRP, IL-6, and TNF-α levels. It also led to greater symptom improvement and shorter hospitalization compared with placebo.543 In a randomized controlled trial that compared 800 mg of boswellic acid three times daily to placebo in 80 ischemic stroke patients, boswellic acid-treated participants had decreased levels of TNF-α, IL-1β, IL-6, and IL-8, as well as prostaglandin E2, after seven days and greater neurological recovery after 30 days.544

Boswellia is often combined with other anti-inflammatory nutrients. In a randomized controlled trial in 90 subjects with chronic low back pain, 300 mg of combined Boswellia plus curcumin daily for 90 days reduced hs-CRP, IL-6, and TNF-α levels, alleviated pain and disability, and improved quality of life scores compared with placebo.545 A randomized placebo-controlled trial that included 62 participants with knee osteoarthritis found supplementing with 300 mg of Boswellia extract plus 250 mg of celery (Apium graveolens) seed extract twice daily for 90 days decreased levels of hs-CRP, IL-1, IL-6, IL-7, and TNF-α; reduced pain, stiffness, and swelling; and reduced biomarkers of cartilage degeneration.546

Protein Supplements

Reported dosage: 30–87 grams of whey protein or 15–40 grams soy protein daily

Protein supplements contain bioactive peptides that, when released, may reduce markers of inflammation.547 One meta-analysis included 31 randomized controlled trials investigating the effects of protein supplements (18 using whey protein and 13 using soy protein) on inflammatory marker levels in elderly subjects. Pooled analysis of results from trials that used whey protein found 30 grams or more of whey protein per day reduced IL-6 levels, particularly in those with sarcopenia and pre-frailty. Whey protein was also found to reduce CRP levels but only in subjects without obesity. The analysis also showed soy protein, at doses of 15–40 grams daily, reduced TNF-α levels, and the effect was enhanced when soy isoflavones were present in the soy supplement.548 Whey protein dosages in the included studies ranged from 15 to 87 grams per day, and the reported soy protein dosages ranged from 15 to 52 grams per day. On the other hand, a meta-analysis that included 11 randomized controlled trials found whey protein did not affect levels of IL-6 or TNF-α, regardless of health status.549

Magnesium

Reported dosage: 250–500 mg daily

Higher magnesium intake and blood levels have been associated with lower hs-CRP levels in multiple observational studies.550-552 A study in 5,775 subjects, aged 40–73 years, found higher magnesium intake was correlated with lower hs-CRP and GlycA levels, as well as a lower number of leukocytes (a type of white blood cell, mainly consisting of neutrophils).553 Another study found a link between higher magnesium intake and lower IL-6 levels in participants with a gene variant known to affect magnesium metabolism.554

Clinical trials investigating magnesium’s effect on markers of inflammation have yielded mixed results. One placebo-controlled trial in 60 coronary artery disease patients found 300 mg of magnesium sulfate daily for three months lowered serum levels, as well as gene expression levels, of the pro-inflammatory cytokines IL-18 and TNF-α.555 A meta-analysis of 17 randomized controlled trials with a combined total of 889 participants found magnesium supplementation, in doses ranging from 250–500 mg per day, decreased CRP levels compared with placebo, but had no effect on IL-6 or TNF-α. Some trials in the analysis found magnesium lowered IL-1 and fibrinogen levels, but these effects were not measured in enough trials to be included in the meta-analysis.556 However, another meta-analysis of 18 trials with 927 participants found no reduction in CRP, IL-6, or TNF-α levels with magnesium supplementation.557 People with existing inflammation may benefit more from magnesium treatment than those without inflammation. A meta-analysis that included 11 randomized controlled trials found that whereas magnesium did not lower CRP levels in the overall analysis, it did lower CRP levels in a subset of participants whose baseline CRP levels were ≥3.0 mg/dL.558

Selenium

Reported dosage: 150–200 mcg daily

Selenium is a vital nutrient for reduction–oxidation balance, and some evidence suggests selenium may help regulate inflammatory activity in the immune system.559,560 Selenium levels have been reported to be lower in patients with autoimmune and cardiovascular diseases, compared to the overall population.561 An observational study that included 606 elderly subjects with cardiovascular disease and 858 healthy elderly individuals found selenium deficiency was correlated with increased CRP levels and neutrophil-to-lymphocyte ratios, as well as increased likelihood of cardiovascular disease.562 A meta-analysis that included four randomized controlled trials investigating CRP changes in coronary heart disease patients found treatment with selenium, at doses of 150–500 mcg daily, reduced CRP levels, though it did not affect mortality.563 Another meta-analysis that included 13 randomized controlled trials also found selenium supplementation was effective for reducing CRP levels. Most of the included studies used 200 mcg of selenium; however, the range of doses used was 50–500 mcg per day.564

The effect of intravenous selenium on markers of inflammation appears to be somewhat different than with oral selenium supplementation. A meta-analysis that pooled findings from 24 randomized controlled trials found intravenous selenium administration reduced CRP and IL-6 levels but increased TNF-α levels, whereas oral selenium supplementation only had an effect on IL-6.565

N-acetylcysteine

Reported dosage: 400–2,000 mg daily

N-acetylcysteine (NAC), a form of the amino acid cysteine, can be used to synthesize glutathione. Glutathione is a major biological oxidative stress modulator and has been shown to regulate immune cell activity and inhibit release of pro-inflammatory cytokines.566

In a controlled study involving 32 patients with acute myocardial infarction (heart attack), those who received standard therapies plus 600 mg of NAC every eight hours for three days had reduced galectin-3 and hs-CRP levels compared with those who receive standard therapies alone.567 One meta-analysis that pooled findings from 24 randomized controlled trials with a total of 1,057 participants found treatment with NAC, at doses of 400–2,000 mg daily for up to 80 weeks, reduced CRP and IL-6 levels but did not affect other markers of inflammation.568 A meta-analysis of 28 randomized controlled trials found NAC significantly improved TNF-α, IL-6, IL-8, and homocysteine levels, though not CRP.569 A systematic review and meta-analysis of 20 randomized controlled trials found treatment with NAC before exercise decreased post-exercise levels of IL-6, but not TNF-α, and raised glutathione levels.566

Bromelain

Reported dosage: 90–500 mg daily

Typically derived from pineapple stem, bromelain—a mixture of proteolytic enzymes—has demonstrated a variety of anti-inflammatory properties in preclinical research.272,571 Bromelain has been shown to reduce NF-kappaB activation, pro-inflammatory cytokine signaling, and inflammatory immune cell activity. It also reduces COX-2 enzyme expression, decreasing the synthesis of prostaglandins,572,573 and modulates the inflammatory response through its effects on fibrin and fibrinogen.272,572

Human trials of bromelain for inflammatory conditions have yielded promising results.273 Bromelain has been reported to reduce swelling and relieve pain after dental surgery and improve symptoms in subjects with knee pain and arthritis.274,275,277,576 In addition, a combination of bromelain plus trypsin (an enzyme that breaks down proteins) and rutoside (a flavonoid) reduced pain and inflammation comparably with a non-steroidal anti-inflammatory drug (NSAID) (diclofenac), and also enhanced the effectiveness of this drug.274,276

A systematic review examined the effect of bromelain on levels of inflammatory biomarkers using findings from seven randomized controlled trials. Although the trials had substantial differences in forms of bromelain used, biomarkers measured, and participant populations, bromelain appeared generally to have beneficial effects on markers of inflammation including reductions in CRP, IL-6, IL-5, IL-12, and PGE2 levels. Trials in this review that found reductions in inflammatory marker levels used doses of 90–500 mg bromelain daily, sometimes in divided doses and/or combined with other nutrients.580

Bromelain and Vocal Cord Inflammation

One context in which natural anti-inflammatories such as bromelain may be useful is for vocal cord inflammation that often occurs in singers.

Singers, teachers, and other individuals who regularly use or overuse their voice (sometimes referred to as misuse or even abuse) are at high risk of straining their vocal cords, which can lead to inflammation and swelling.278-280 This chronic irritation can lead to the development of polyps and nodules, and even a type of vocal cord hemorrhage.282 These problems are usually addressed by resting the voice (if possible), proper use of the voice, oral hydration, and use of an indoor humidifier. Oral or injected corticosteroids, or surgical removal, may be necessary in some cases.278-283 It is not uncommon for these lesions to reoccur when the activity leading to their development (eg, singing or yelling) continues; thus, non-invasive natural support is often sought.

On the basis of the evidence supporting bromelain’s ability to attenuate inflammation, it has garnered popularity among singers and vocalists as a remedy for swelling and inflammation of the vocal cords.284 This is especially important and valuable considering that NSAIDs are a risk factor for a common cause of vocal cord problems.282,285

A clinical study examining 56 people who underwent surgery for benign vocal cord conditions found that post-operative treatment with a combination of bromelain (80 mg) and Boswellia (750 mg) twice daily for one week was effective in promoting tissue repair and recovery of vocal performance. This treatment was well tolerated and produced fewer side effects than treatment with inhaled steroids.281

Sesame

Reported dosage: 200 mg of sesamin or 40 grams of ground sesame seeds daily

Sesame oil and its lignans (sesamin, sesamolin, sesaminol, and sesamol) have demonstrated anti-inflammatory and immune-regulating effects in numerous preclinical studies. Actions attributed to sesame oil constituents include inhibiting cyclooxygenase (COX)-2 enzyme, thereby decreasing production of inflammatory eicosanoids, and downregulating NF-kappaB, thereby suppressing production of pro-inflammatory cytokines.332,589,590 In a randomized controlled trial in 44 women with rheumatoid arthritis, 200 mg of sesamin daily for six weeks lowered levels of hs-CRP, TNF-α, and COX-2, but not IL-1β or IL-6, and decreased joint tenderness and pain compared with placebo.591 Another randomized controlled trial in 50 subjects with knee osteoarthritis found 40 grams of ground sesame seeds per day for two months reduced levels of IL-6, but not hs-CRP.592 However, in a randomized crossover trial in 30 adults with overweight or obesity, 25 grams of sesame daily for five weeks had a similar effect to placebo on levels of CRP, IL-6, and TNF-α.593

Modified Citrus Pectin

Reported dosage: 20 grams daily

Citrus pectin is a dietary fiber and a large complex molecule that is not digested or absorbed by the human digestive system. Modified citrus pectin (MCP) is an enzymatically processed form of pectin made up of smaller molecules that are absorbed into circulation.594 Pectin and MCP bind to and inactivate galectin-3, a binding protein involved in inflammation, fibrosis, and cancer metastasis.595 MCP has also been shown to downregulate NF-kappaB and reduce expression of pro-inflammatory cytokines.596 Furthermore, MCP can be metabolized by intestinal bacteria, resulting in products that may also help regulate inflammation.597 The anti-inflammatory effects of MCP are thought to underlie the positive health effects it has demonstrated in preclinical research, such as decreasing cardiovascular inflammation, protecting against cardiac fibrosis, reducing atherosclerotic plaques, and enhancing cartilage healing.598-602 In addition, a growing body of evidence suggests it may improve certain outcomes in cancer patients.594

In a pilot clinical trial in 29 healthy adults, 20 grams of modified citrus pectin daily for four weeks reduced levels of IL-1β, IL-6, and TNF-α; raised levels of anti-inflammatory IL-10; and reduced anxiety scores significantly more than placebo.603

Mitochondrial Support

Reactive oxygen species generated during mitochondrial respiration contribute to inflammation, as outlined previously in the protocol. Aging individuals are especially susceptible to mitochondria-related oxidative stress since mitochondria become increasingly dysfunctional with age.604,605 Taking steps to support mitochondrial integrity and efficiency can help alleviate some of the systemic oxidative and inflammatory burden caused by poorly functioning mitochondria.606 Two nutrients, coenzyme Q10 and pyrroloquinoline quinone, are powerful mitochondrial protectants, and studies support an anti-inflammatory role for these compounds.607,608

Pyrroloquinoline quinone

Reported dosage: 300 mcg per kg body weight or approximately 20 mg daily

Pyrroloquinoline quinone (PQQ) is a cofactor for enzymes critically important for cellular energy homeostasis and redox balance. Several studies have shown that PQQ promotes new mitochondria formation, supports mitochondrial function, and reduces oxidative stress.608 Preclinical research has suggested PQQ can reduce joint inflammation; suppress release of IL-1β, IL-6, and TNF-α by injured joint cells; and protect mitochondria in cartilage cells from cytokine-induced damage.609,610 In 10 healthy adults, 300 mcg per kg body weight of PQQ daily for three days increased antioxidant status, decreased CRP and IL-6 levels, and raised markers of mitochondrial function.611 In addition, randomized controlled trials have found 20–21.5 mg of PQQ daily for 6–12 weeks increased brain blood flow, brain oxygenation, brain cell activity, and cognitive performance in older individuals, and improved the mitochondrial response to endurance training in healthy men.612-615

Coenzyme Q10

Reported dosage: 100–400 mg daily

Coenzyme Q10 (CoQ10) is found in lipid cell membranes, especially in mitochondrial membranes, where it is needed for ATP production. It is also a free radical scavenger that protects lipids from oxidative damage and participates in lipid metabolism.607 Studies have shown CoQ10 levels are low in individuals with a broad range of age-related and inflammatory conditions, including cardiovascular, metabolic, neurological, and kidney diseases; and lower levels are associated with increased mortality risk.616,617 Furthermore, supplementation has been shown to be helpful in these and other inflammatory conditions, including rheumatic diseases.616,618

CoQ10 supplementation has been found to reduce levels of inflammatory biomarkers in numerous clinical trials. One systematic review and meta-analysis that examined data from 31 randomized controlled trials involving a total of 1,517 participants concluded that CoQ10, at daily doses of 300–400 mg, was effective for reducing levels of CRP, IL-6, and TNF-α.619 A meta-analysis that pooled data from six randomized placebo-controlled trials involving 318 participants with metabolic syndrome found 100–200 mg of CoQ10 daily increased levels of adiponectin (an anti-inflammatory cytokine produced by fat cells), improved oxidative stress indicators, and lowered levels of inflammatory biomarkers.620 On the other hand, a meta-analysis that included 13 randomized controlled trials involving subjects with coronary artery disease found CoQ10 did not lower levels of CRP, IL-6, or TNF-α, though it did reduce oxidative stress.621

Counteracting Proinflammatory Glycation Reactions

Interactions between advanced glycation end products (AGEs) and their receptor (RAGE) play a key role in promoting acute and chronic inflammation by activating inflammatory signaling pathways such as NF-kappaB and stimulating the release of pro-inflammatory cytokines.622,623 Fortunately, in addition to dietary measures for controlling blood glucose concentrations and avoiding high-AGE foods, some natural compounds inhibit glycation and may help rein in the inflammatory cascade.622

Benfotiamine

Reported dosage: 100–600 mg daily along with other B vitamins

Benfotiamine is a synthetic fat-soluble derivative of vitamin B1 (thiamine). Thiamine and benfotiamine activate an enzyme involved in the breakdown of AGE precursors, thereby decreasing AGE production, AGE/RAGE interactions, and related inflammatory signaling.624 It has demonstrated potent antioxidant and anti-inflammatory properties and has been shown to help prevent diabetes complications related to inflammation in animal studies.625,626 Moreover, laboratory experiments have indicated benfotiamine may regulate inflammation by modulating COX and LOX enzyme activity.627

In a randomized controlled trial in 70 patients with mild cognitive impairment or mild dementia related to Alzheimer disease, 300 mg of benfotiamine twice daily for 12 months slowed the progression of cognitive decline and the rise in circulating AGE levels compared with placebo.628 In a randomized placebo-controlled trial, 30 subjects with osteoarthritis were given a supplement providing 50 mg of benfotiamine, 50 mg of pyridoxamine (vitamin B6), and 500 mcg of methylcobalamin (vitamin B12) or placebo to take three times daily for 24 weeks. The supplement group had reduced arthritis pain and disability scores, decreased AGE levels, and lower CRP levels compared with the placebo group at the end of the trial.629 An uncontrolled trial involving 24 rheumatoid arthritis patients found the same supplement, taken twice daily for 12 weeks, improved a measure of blood vessel function called flow-mediated vasodilation, decreased CRP levels, and lowered disease activity scores.630 In a randomized controlled trial, 165 subjects with diabetic neuropathy were randomized to receive 300 or 600 mg of benfotiamine per day or placebo for six weeks. Neuropathy symptom scores were reduced in those taking benfotiamine, and the effect was stronger in those taking the higher dose.220

However, not all clinical trials have found benfotiamine to improve inflammation in individuals with type 1 or type 2 diabetes. In one randomized placebo-controlled trial in patients with type 1 diabetes, 300 mg of benfotiamine daily for 24 months did not affect inflammatory biomarker levels or nerve function.632 Another randomized placebo-controlled trial in 39 type 2 diabetes patients with evidence of kidney damage found benfotiamine, at 300 mg three times daily for 12 weeks, did not reduce levels of AGEs or inflammatory biomarkers and did not improve vascular function.633

Carnosine

Reported dosage: 500–2,000 mg daily

Carnosine is a dipeptide made from the amino acids beta-alanine and histidine and found mainly in meat. Carnosine has been shown to scavenge free radicals, downregulate inflammatory signaling, and inhibit glycation.634,635 A meta-analysis of nine randomized controlled trials with a total of 350 participants found supplementing with carnosine, at doses ranging from 500 to 2,000 mg per day, or its amino acid precursors reduced levels of CRP and TNF-α, but not IL-6.636 In one of the trials, 54 subjects with type 2 diabetes received 1,000 mg of carnosine or placebo daily for 12 weeks; carnosine was found to reduce AGE and TNF-α levels and improve markers of blood glucose control.637 However, in another trial in 41 patients with well-controlled type 2 diabetes or prediabetes, 2,000 mg of carnosine per day for 14 weeks did not reduce inflammatory biomarker levels relative to placebo.638 Carnosine is easily broken down in the digestive tract, which may account for mixed clinical results.639 Interestingly, supplementing with a carnosine metabolite called carcinine, at 60 mg daily for two months, was found to reduce AGE levels and improve markers of cardiometabolic health more than placebo in an observational study involving 100 volunteers with metabolic syndrome.640 Other research suggests topical carnosine may inhibit glycation reactions in the skin, reducing skin aging.641

Hesperidin

Reported dosage: approximately 300–1,000 mg of hesperidin or 500–750 mL (~16–25 oz.) orange juice daily

Hesperidin and its metabolite hesperetin are flavonoids found mainly in citrus fruits, particularly their peels. These compounds are powerful free radical scavengers and have demonstrated anti-inflammatory, insulin-sensitizing, lipid-lowering, neuroprotective, and anti-aging activities.642,643 Preclinical research has also indicated these compounds promote bone and cartilage repair through antioxidant and anti-inflammatory effects, which are partly due to downregulation of NF-kappaB.643 Furthermore, studies in animals suggest hesperidin can upregulate cellular metabolism by activating adenosine 5'-monophosphate protein kinase (AMPK), peroxisome proliferator-activated receptor (PPAR), and other signaling pathways.644

A growing body of research indicates hesperidin can promote insulin sensitivity and metabolic health by reducing chronic inflammation.645,646 In a randomized controlled trial, 49 patients with metabolic syndrome received 500 mg of hesperidin or placebo twice daily for 12 weeks. Compared with placebo, hesperidin reduced TNF-α (but not hs-CRP), fasting blood glucose, and triglyceride levels, as well as systolic blood pressure.647 A meta-analysis that pooled results from six randomized controlled trials with 296 participants found hesperidin supplementation at doses of 292–600 mg daily reduced levels of vascular cell adhesion molecule (VCAM)-1, an adhesion molecule involved in the inflammatory response. Although hesperidin did not decrease CRP levels in the overall analysis, it was found to have a significant CRP-lowering effect in parallel (rather than crossover) trials and trials lasting longer than four weeks.648

Orange juice is an important dietary source of hesperidin and contains an array of related flavonoids in smaller concentrations. These compounds appear to contribute to orange juice’s immune-modulating and anti-inflammatory effects.649 A systematic review of 20 controlled clinical trials found 500–750 mL of orange juice daily lowered CRP, IL-6, and TNF-α levels, and the effects increased as the amount and duration of use increased.650

2025

  • Nov: Substantially updated Inflammation: Lab Tests, Biomarkers & Mediators and Nutrients sections

2023

  • May: In Drug Strategies to Combat Chronic Inflammation, updated discussion about pentoxifylline and added discussion about carbergoline

2021

  • Oct: Added section on bromelain and vocal cord inflammation to Nutrients
  • Oct: Updated section on bromelain in Nutrients

2012

  • Feb: Comprehensive update & review

Disclaimer and Safety Information

This information (and any accompanying material) is not intended to replace the attention or advice of a physician or other qualified health care professional. Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle change intended to prevent or treat a specific disease or condition should first consult with and seek clearance from a physician or other qualified health care professional. Pregnant women in particular should seek the advice of a physician before using any protocol listed on this website. The protocols described on this website are for adults only, unless otherwise specified. Product labels may contain important safety information and the most recent product information provided by the product manufacturers should be carefully reviewed prior to use to verify the dose, administration, and contraindications. National, state, and local laws may vary regarding the use and application of many of the therapies discussed. The reader assumes the risk of any injuries. The authors and publishers, their affiliates and assigns are not liable for any injury and/or damage to persons arising from this protocol and expressly disclaim responsibility for any adverse effects resulting from the use of the information contained herein.

The protocols raise many issues that are subject to change as new data emerge. None of our suggested protocol regimens can guarantee health benefits. Life Extension has not performed independent verification of the data contained in the referenced materials, and expressly disclaims responsibility for any error in the literature.

  1. Centers for Disease Control and Prevention. FASTSTATS - Leading Causes of Death. cdc.gov. 2011; Available at: http://www.cdc.gov/NCHS/fastats/Default.htm [Accessed December 23, 2011].
  2. Bastard, J.-P., Maachi, M., Lagathu, C., et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur. Cytokine Netw. 2006;17(1):4–12
  3. Cao, J. J. Effects of obesity on bone metabolism. J Orthop Surg Res. 2011;6:30
  4. Jha, R. K., Ma, Q., Sha, H., and Palikhe, M. Acute pancreatitis: a literature review. Med Sci Monit. 2009;15(7):RA147–56
  5. Ferrucci, L., Semba, R. D., Guralnik, J. M., et al. Proinflammatory state, hepcidin, and anemia in older persons. Blood. 2010;115(18):3810–3816
  6. Glorieux, G., Cohen, G., Jankowski, J., and Vanholder, R. Platelet/Leukocyte activation, inflammation, and uremia. Semin Dial. 2009;22(4):423–427
  7. Kundu, J. K., and Surh, Y.-J. Inflammation: gearing the journey to cancer. Mutat Res. 2008;659(1-2):15–30
  8. Murphy SL. et al. Deaths: Preliminary Data for 2010. National Vital Statistics Report 60:4; 1/11/2012.
  9. Singh, T., and Newman, A. B. Inflammatory markers in population studies of aging. Ageing Res Rev. 2011;10(3):319–329
  10. Karin, M., Lawrence, T., and Nizet, V. Innate immunity gone awry: linking microbial infections to chronic inflammation and cancer. Cell. 2006;124(4):823–835
  11. Medzhitov, R. Origin and physiological roles of inflammation. Nature. 2008;454(7203):428–435
  12. Green DR et al. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science. 2011 Aug 26;333(6046):1109-12.
  13. Dinarello CA et al. A clinical perspective of IL-1β as the gatekeeper of inflammation. Eur J Immunol. 2011 May;41(5):1203-17. doi: 10.1002/eji.201141550.
  14. Tschopp J. Mitochondria: Sovereign of inflammation? Eur J Immunol. 2011 May;41(5):1196-202. doi: 10.1002/eji.201141436.
  15. Mosquera JA. [Role of the receptor for advanced glycation end products (RAGE) in inflammation]. Invest Clin. 2010 Jun;51(2):257-68.
  16. Witko-Sarsat, V., Friedlander, M., Nguyen Khoa, T., et al. Advanced oxidation protein products as novel mediators of inflammation and monocyte activation in chronic renal failure. J Immunol. 1998;161(5):2524–2532
  17. Vlassara, H., Cai, W., Crandall, J., et al. Inflammatory mediators are induced by dietary glycotoxins, a major risk factor for diabetic angiopathy. Proc Natl Acad Sci USA. 2002;99(24):15596–15601
  18. Martinon, F., Pétrilli, V., Mayor, A., Tardivel, A., and Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–241
  19. Alvarez-Lario, B., and Macarrón-Vicente, J. Is there anything good in uric acid? QJM. 2011;
  20. Nguyen Khoa, T., Massy, Z. A., Witko-Sarsat, V., et al. Oxidized low-density lipoprotein induces macrophage respiratory burst via its protein moiety: A novel pathway in atherogenesis? Biochem Biophys Res Commun. 1999;263(3):804–809
  21. Au-Yeung, K. K. W., Yip, J. C. W., Siow, Y. L., and O, K. Folic acid inhibits homocysteine-induced superoxide anion production and nuclear factor kappa B activation in macrophages. Can. J. Physiol. Pharmacol. 2006;84(1):141–147
  22. Emerging Risk Factors Collaboration, Kaptoge, S., Di Angelantonio, E., et al. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375(9709):132–140
  23. Trayhurn, P., and Wood, I. S. Signalling role of adipose tissue: adipokines and inflammation in obesity. Biochem. Soc. Trans. 2005;33(Pt 5):1078–1081
  24. Schrager, M. A., Metter, E. J., Simonsick, E., et al. Sarcopenic obesity and inflammation in the InCHIANTI study. J Appl Physiol. 2007;102(3):919–925
  25. Fried, S. K., Bunkin, D. A., and Greenberg, A. S. Omental and subcutaneous adipose tissues of obese subjects release interleukin-6: depot difference and regulation by glucocorticoid. J Clin Endocrinol Metab. 1998;83(3):847–850
  26. Mohamed-Ali, V., Goodrick, S., Rawesh, A., et al. Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-alpha, in vivo. J Clin Endocrinol Metab. 1997;82(12):4196–4200
  27. Ortega Martinez de Victoria, E., Xu, X., Koska, J., et al. Macrophage content in subcutaneous adipose tissue: associations with adiposity, age, inflammatory markers, and whole-body insulin action in healthy Pima Indians. Diabetes. 2009;58(2):385–393
  28. Weisberg, S. P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R. L., and Ferrante, A. W. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–1808
  29. Nappo, F., Esposito, K., Cioffi, M., et al. Postprandial endothelial activation in healthy subjects and in type 2 diabetic patients: role of fat and carbohydrate meals. J Am Coll Cardiol. 2002;39(7):1145–1150
  30. Peairs, A. D., Rankin, J. W., and Lee, Y. W. Effects of acute ingestion of different fats on oxidative stress and inflammation in overweight and obese adults. Nutr J. 2011;10:122
  31. Myhrstad, M. C. W., Narverud, I., Telle-Hansen, V. H., et al. Effect of the fat composition of a single high-fat meal on inflammatory markers in healthy young women. Br J Nutr. 2011;106(12):1826–1835
  32. Poppitt, S. D., Keogh, G. F., Lithander, F. E., et al. Postprandial response of adiponectin, interleukin-6, tumor necrosis factor-alpha, and C-reactive protein to a high-fat dietary load. Nutrition. 2008;24(4):322–329
  33. Payette, C., Blackburn, P., Lamarche, B., et al. Sex differences in postprandial plasma tumor necrosis factor-alpha, interleukin-6, and C-reactive protein concentrations. Metab. Clin. Exp. 2009;58(11):1593–1601
  34. Mozaffarian D, Pischon T, Hankinson SE, et al. Dietary intake of trans fatty acids and systemic inflammation in women. Am J Clin Nutr. 2004;79(4):606-12. doi: 10.1093/ajcn/79.4.606. https://pubmed.ncbi.nlm.nih.gov/15051604/
  35. Lopez-Garcia, E., Schulze, M. B., Meigs, J. B., et al. Consumption of trans fatty acids is related to plasma biomarkers of inflammation and endothelial dysfunction. J Nutr. 2005;135(3):562–566
  36. Nielsen, B. M., Nielsen, M. M., Jakobsen, M. U., et al. A cross-sectional study on trans-fatty acids and risk markers of CHD among middle-aged men representing a broad range of BMI. Br J Nutr. 2011;106(8):1245–1252
  37. Bendsen, N. T., Stender, S., Szecsi, P. B., et al. Effect of industrially produced trans fat on markers of systemic inflammation: evidence from a randomized trial in women. The Journal of Lipid Research. 2011;52(10):1821–1828
  38. Ahmadi N, Eshaghian S, Huizenga R, et al. Effects of intense exercise and moderate caloric restriction on cardiovascular risk factors and inflammation. Am J Med. 2011;124(10):978-82.
  39. González O, Tobia C, Ebersole J, et al. Caloric restriction and chronic inflammatory diseases. Oral Dis. 2012;18(1):16-31.
  40. Gilliver, S. C. Sex steroids as inflammatory regulators. J. Steroid Biochem. Mol. Biol. 2010;120(2-3):105–115
  41. Keller, E. T., Chang, C., and Ershler, W. B. Inhibition of NFkappaB activity through maintenance of IkappaBalpha levels contributes to dihydrotestosterone-mediated repression of the interleukin-6 promoter. J Biol Chem. 1996;271(42):26267–26275
  42. Ray, P., Ghosh, S. K., Zhang, D. H., and Ray, A. Repression of interleukin-6 gene expression by 17 beta-estradiol: inhibition of the DNA-binding activity of the transcription factors NF-IL6 and NF-kappa B by the estrogen receptor. FEBS letters. 1997;409(1):79–85
  43. Deshpande, R., Khalili, H., Pergolizzi, R. G., Michael, S. D., and Chang, M. D. Estradiol down-regulates LPS-induced cytokine production and NFkB activation in murine macrophages. Am. J. Reprod. Immunol. 1997;38(1):46–54
  44. Maggio, M., Basaria, S., Ble, A., et al. Correlation between testosterone and the inflammatory marker soluble interleukin-6 receptor in older men. J Clin Endocrinol Metab. 2006;91(1):345–347
  45. Khosla, S., Atkinson, E. J., Dunstan, C. R., and O'Fallon, W. M. Effect of estrogen versus testosterone on circulating osteoprotegerin and other cytokine levels in normal elderly men. J Clin Endocrinol Metab. 2002;87(4):1550–1554
  46. Gameiro, C., and Romao, F. Changes in the immune system during menopause and aging. Front Biosci (Elite Ed). 2010;2:1299–1303
  47. Kane, S. V., and Reddy, D. Hormonal replacement therapy after menopause is protective of disease activity in women with inflammatory bowel disease. Am J Gastroenterol. 2008;103(5):1193–1196
  48. Vural, P., Akgul, C., and Canbaz, M. Effects of hormone replacement therapy on plasma pro-inflammatory and anti-inflammatory cytokines and some bone turnover markers in postmenopausal women. Pharmacol. Res. 2006;54(4):298–302
  49. Anderson, G. L., Limacher, M., Assaf, A. R., et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA. 2004;291(14):1701–1712
  50. Arnson, Y., Shoenfeld, Y., and Amital, H. Effects of tobacco smoke on immunity, inflammation and autoimmunity. J. Autoimmun. 2010;34(3):J258–65
  51. Lee J, Taneja V, Vassallo R. Cigarette smoking and inflammation: cellular and molecular mechanisms. J Dent Res. 2012;91(2):142-9.
  52. Vgontzas, A. N., Papanicolaou, D. A., Bixler, E. O., Kales, A., Tyson, K., and Chrousos, G. P. Elevation of plasma cytokines in disorders of excessive daytime sleepiness: role of sleep disturbance and obesity. J Clin Endocrinol Metab. 1997;82(5):1313–1316
  53. Vgontzas, A. N., Papanicolaou, D. A., Bixler, E. O., et al. Sleep apnea and daytime sleepiness and fatigue: relation to visceral obesity, insulin resistance, and hypercytokinemia. J Clin Endocrinol Metab. 2000;85(3):1151–1158
  54. Vgontzas, A. N., Zoumakis, M., Bixler, E. O., et al. Impaired nighttime sleep in healthy old versus young adults is associated with elevated plasma interleukin-6 and cortisol levels: physiologic and therapeutic implications. J Clin Endocrinol Metab. 2003;88(5):2087–2095
  55. Trakada, G., Chrousos, G., Pejovic, S., and Vgontzas, A. Sleep Apnea and its association with the Stress System, Inflammation, Insulin Resistance and Visceral Obesity. Sleep Med Clin. 2007;2(2):251–261
  56. Slade, G. D., Ghezzi, E. M., Heiss, G., Beck, J. D., Riche, E., and Offenbacher, S. Relationship between periodontal disease and C-reactive protein among adults in the Atherosclerosis Risk in Communities study. Arch Intern Med. 2003;163(10):1172–1179
  57. Pradeep, A. R., Kathariya, R., Arjun Raju, P., Sushma Rani, R., Sharma, A., and Raghavendra, N. M. Risk factors for chronic kidney diseases may include periodontal diseases, as estimated by the correlations of plasma pentraxin-3 levels: a case-control study. Int Urol Nephrol. 2011;
  58. Vaishnava, P., Narayan, R., and Fuster, V. Understanding systemic inflammation, oral hygiene, and cardiovascular disease. Am. J. Med. 2011;124(11):997–999
  59. Pervanidou, P., and Chrousos, G. P. Metabolic consequences of stress during childhood and adolescence. Metab. Clin. Exp. 2011;
  60. van Westerloo, D. J. The vagal immune reflex: a blessing from above. Wien Med Wochenschr. 2010;160(5-6):112–117
  61. Pontet, J., Contreras, P., Curbelo, A., et al. Heart rate variability as early marker of multiple organ dysfunction syndrome in septic patients. J Crit Care. 2003;18(3):156–163
  62. Taylor, L., Loerbroks, A., Herr, R. M., Lane, R. D., Fischer, J. E., and Thayer, J. F. Depression and smoking: mediating role of vagal tone and inflammation. Ann Behav Med. 2011;42(3):334–340
  63. Basta G, Schmidt AM, De Caterina R. Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes. Cardiovasc Res. 2004 Sep 1;63(4):582-92.
  64. Uribarri J, Cai W, Sandu O, Peppa M, Goldberg T, Vlassara H. Diet-derived advanced glycation end products are major contributors to the body’s AGE pool and induce inflammation in healthy subjects. Ann N Y Acad Sci. 2005 Jun;1043:461-6.
  65. Toma L, Stancu CS, Botez GM, Sima AV, Simionescu M. Irreversibly glycated LDL induce oxidative and inflammatory state in human endothelial cells; added effect of high glucose. Biochem Biophys Res Commun. 2009 Dec 18;390(3):877-82.
  66. Henry-Vitrac C, Ibarra A, Roller M, Merillon JM, Vitrac X. Contribution of chlorogenic acids to the inhibition of human hepatic glucose-6-phosphatase activity in vitro by Svetol, a standardized decaffeinated green coffee extract. J Agric Food Chem. 2010 Apr 14;58(7):4141-4.
  67. Andrade-Cetto A, Vazquez RC. Gluconeogenesis inhibition and phytochemical composition of two Cecropia species. J Ethnopharmacol. 2010 Jul 6;130(1):93-7.
  68. Rodriguez de Sotillo DV, Hadley M, Sotillo JE. Insulin receptor exon 11+/- is expressed in Zucker (fa/fa) rats, and chlorogenic acid modifies their plasma insulin and liver protein and DNA. J Nutr Biochem. 2006 Jan;17(1):63-71.
  69. Nagendran MV. Effect of green coffee bean extract (GCE), High in Chlorogenic Acids, on Glucose Metabolism. Poster presentation number: 45-LB-P. Obesity 2011, the 29th Annual Scientific Meeting of the Obesity Society. Orlando, Florida. October 1-5, 2011.
  70. Aggarwal, B. B., Shishodia, S., Sandur, S. K., Pandey, M. K., and Sethi, G. Inflammation and cancer: how hot is the link? Biochem. Pharmacol. 2006;72(11):1605–1621
  71. Balkwill, F. Tumour necrosis factor and cancer. Nat. Rev. Cancer. 2009;9(5):361–371
  72. Pickup, J. C., Chusney, G. D., Thomas, S. M., and Burt, D. Plasma interleukin-6, tumour necrosis factor alpha and blood cytokine production in type 2 diabetes. Life Sci. 2000;67(3):291–300
  73. Hong, T., Tan, A. G., Mitchell, P., and Wang, J. J. A review and meta-analysis of the association between C-reactive protein and age-related macular degeneration. Surv Ophthalmol. 2011;56(3):184–194
  74. Dantzer, R. Depression and inflammation: an intricate relationship. Biol. Psychiatry. 2012;71(1):4–5
  75. Gimeno, D., Kivimäki, M., Brunner, E. J., et al. Associations of C-reactive protein and interleukin-6 with cognitive symptoms of depression: 12-year follow-up of the Whitehall II study. Psychol Med. 2009;39(3):413–423
  76. Copeland, W. E., Shanahan, L., Worthman, C., Angold, A., and Costello, E. J. Cumulative depression episodes predict later C-reactive protein levels: a prospective analysis. Biol. Psychiatry. 2012;71(1):15–21
  77. Yaffe, K., Lindquist, K., Penninx, B. W., et al. Inflammatory markers and cognition in well-functioning African-American and white elders. Neurology. 2003;61(1):76–80
  78. Kaser, A., and Tilg, H. “Metabolic aspects” in inflammatory bowel diseases. Curr Drug Deliv. 2011;
  79. Kadetoff, D., Lampa, J., Westman, M., Andersson, M., and Kosek, E. Evidence of central inflammation in fibromyalgia - Increased cerebrospinal fluid interleukin-8 levels. J. Neuroimmunol. 2011;
  80. Rolland, Y., Abellan van Kan, G., Gillette-Guyonnet, S., and Vellas, B. Cachexia versus sarcopenia. Current Opinion in Clinical Nutrition and Metabolic Care. 2011;14(1):15–21
  81. Groesdonk HV et al. [Anti-inflammatory effects of pentoxifylline: importance in cardiac surgery]. Anaesthesist. 2009 Nov;58(11):1136-43. Review. German.
  82. Li W et al. Systematic review on the treatment of pentoxifylline in patients with non-alcoholic fatty liver disease. Lipids Health Dis. 2011 Apr 8;10:49. Review.
  83. Lopes de Jesus CC et al. lline for diabetic retinopathy. Cochrane Database Syst Rev. 2008 Apr 16;(2):CD006693. Review.
  84. Lv D et al. Pentoxifylline versus medical therapies for subfertile women with endometriosis. Cochrane Database Syst Rev. 2009 Jul 8;(3):CD007677. Review.
  85. Hepgul G et al. Preventive effect of pentoxifylline on acute radiation damage via antioxidant and anti-inflammatory pathways. Dig Dis Sci. 2010 Mar;55(3):617-25. Epub 2009 Mar 18.
  86. Goicoechea M et al. Effects of pentoxifylline on inflammatory parameters in chronic kidney disease patients: a randomized trial. J Nephrol. 2012 Jan 11:0. doi: 10.5301/jn.5000077. [Epub ahead of print]
  87. Maiti R et al. Effect of Pentoxifylline on inflammatory burden, oxidative stress and platelet aggregability in hypertensive type 2 diabetes mellitus patients. Vascul Pharmacol. 2007 Aug-Sep;47(2-3):118-24. Epub 2007 Jun 2.
  88. Molavi, B., Rassouli, N., Bagwe, S., and Rasouli, N. A review of thiazolidinediones and metformin in the treatment of type 2 diabetes with focus on cardiovascular complications. Vasc Health Risk Manag. 2007;3(6):967–973
  89. Buler, M., Aatsinki, S.-M., Skoumal, R., et al. Energy-sensing Factors Coactivator Peroxisome Proliferator-activated Receptor γ Coactivator 1-α (PGC-1α) and AMP-activated Protein Kinase Control Expression of Inflammatory Mediators in Liver: INDUCTION OF INTERLEUKIN 1 RECEPTOR ANTAGONIST. J Biol Chem. 2012;287(3):1847–1860
  90. Sobel, B. E., Hardison, R. M., Genuth, S., et al. Profibrinolytic, antithrombotic, and antiinflammatory effects of an insulin-sensitizing strategy in patients in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial. Circulation. 2011;124(6):695–703
  91. Gómez-García, A., Martínez Torres, G., Ortega-Pierres, L. E., Rodríguez-Ayala, E., and Alvarez-Aguilar, C. [Rosuvastatin and metformin decrease inflammation and oxidative stress in patients with hypertension and dyslipidemia]. Rev Esp Cardiol. 2007;60(12):1242–1249
  92. Pruski, M., Krysiak, R., and Okopien, B. Pleiotropic action of short-term metformin and fenofibrate treatment, combined with lifestyle intervention, in type 2 diabetic patients with mixed dyslipidemia. Diabetes Care. 2009;32(8):1421–1424
  93. Patrono C, Rocca B. Aspirin: promise and resistance in the new millennium. Arterioscler Thromb Vasc Biol. 2008;28(3):s25-32.
  94. Rothwell PM, Fowkes FG, Belch JF, et al. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet. 2011;377(9759):31-41.
  95. Rothwell PM, Wilson M, Elwin CE, et al. Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet. 2010;376(9754):1741-50.
  96. Salinas CA, Kwon EM, FitzGerald LM, et al. Use of aspirin and other nonsteroidal antiinflammatory medications in relation to prostate cancer risk. Am J Epidemiol. 2010;172(5):578-90.
  97. Flossmann E, Rothwell PM, British Doctors Aspirin Trial and the UK-TIA Aspirin Trial. Effect of aspirin on long-term risk of colorectal cancer: consistent evidence from randomised and observational studies. Lancet. 2007;369(9573):1603-13.
  98. Sobolewski, C., Cerella, C., Dicato, M., Ghibelli, L., and Diederich, M. The role of cyclooxygenase-2 in cell proliferation and cell death in human malignancies. Int J Cell Biol. 2010;2010:215158
  99. Weber, C., Erl, W., Pietsch, A., and Weber, P. C. Aspirin inhibits nuclear factor-kappa B mobilization and monocyte adhesion in stimulated human endothelial cells. Circulation. 1995;91(7):1914–1917
  100. Ikonomidis, I., Andreotti, F., Economou, E., Stefanadis, C., Toutouzas, P., and Nihoyannopoulos, P. Increased proinflammatory cytokines in patients with chronic stable angina and their reduction by aspirin. Circulation. 1999;100(8):793–798
  101. Chen, Y.-G., Xu, F., Zhang, Y., et al. Effect of aspirin plus clopidogrel on inflammatory markers in patients with non-ST-segment elevation acute coronary syndrome. Chin. Med. J. 2006;119(1):32–36
  102. Solheim, S., Arnesen, H., Eikvar, L., Hurlen, M., and Seljeflot, I. Influence of aspirin on inflammatory markers in patients after acute myocardial infarction. Am J Cardiol. 2003;92(7):843–845
  103. Solheim, S., Pettersen, A. A., Arnesen, H., and Seljeflot, I. No difference in the effects of clopidogrel and aspirin on inflammatory markers in patients with coronary heart disease. Thromb. Haemost. 2006;96(5):660–664
  104. Serhan, C. N., Hong, S., Gronert, K., et al. Resolvins: A Family of Bioactive Products of Omega-3 Fatty Acid Transformation Circuits Initiated by Aspirin Treatment that Counter Proinflammation Signals. Journal of Experimental Medicine. 2002;196(8):1025–1037
  105. Stancu, C., and Sima, A. Statins: mechanism of action and effects. J. Cell. Mol. Med. 2001;5(4):378–387
  106. Bu, D.-X., Griffin, G., and Lichtman, A. H. Mechanisms for the anti-inflammatory effects of statins. Curr. Opin. Lipidol. 2011;22(3):165–170
  107. Ridker, P. M., Danielson, E., Fonseca, F. A. H., et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359(21):2195–2207
  108. Bulcão, C., Giuffrida, F. M. A., Ribeiro-Filho, F. F., and Ferreira, S. R. G. Are the beneficial cardiovascular effects of simvastatin and metformin also associated with a hormone-dependent mechanism improving insulin sensitivity? Braz. J. Med. Biol. Res. 2007;40(2):229–235
  109. Galland, L. Diet and inflammation. Nutr Clin Pract. 2010;25(6):634–640
  110. Levitan, E. B., Cook, N. R., Stampfer, M. J., et al. Dietary glycemic index, dietary glycemic load, blood lipids, and C-reactive protein. Metabolism. 2008;57(3):437–443
  111. Du, H., van der A, D. L., van Bakel, M. M. E., et al. Glycemic index and glycemic load in relation to food and nutrient intake and metabolic risk factors in a Dutch population. American Journal of Clinical Nutrition. 2008;87(3):655–661
  112. North, C. J., Venter, C. S., and Jerling, J. C. The effects of dietary fibre on C-reactive protein, an inflammation marker predicting cardiovascular disease. Eur J Clin Nutr. 2009;63(8):921–933
  113. Moschen, A. R., Molnar, C., Geiger, S., et al. Anti-inflammatory effects of excessive weight loss: potent suppression of adipose interleukin 6 and tumour necrosis factor alpha expression. Gut. 2010;59(9):1259–1264
  114. Cavicchia, P. P., Steck, S. E., Hurley, T. G., et al. A new dietary inflammatory index predicts interval changes in serum high-sensitivity C-reactive protein. Journal of Nutrition. 2009;139(12):2365–2372
  115. Bruunsgaard, H. Physical activity and modulation of systemic low-level inflammation. J Leukoc Biol. 2005;78(4):819–835
  116. Ma, Y., Hébert, J., Li, W., and Bertone-Johnson, E. Association between dietary fiber and markers of systemic inflammation in the Women's Health Initiative Observational Study. Nutrition. 2008;
  117. Pischon, T., Hankinson, S. E., Hotamisligil, G. S., Rifai, N., Willett, W. C., and Rimm, E. B. Habitual dietary intake of n-3 and n-6 fatty acids in relation to inflammatory markers among US men and women. Circulation. 2003;108(2):155–160
  118. Lopez-Garcia, E., Schulze, M. B., Manson, J. E., et al. Consumption of (n-3) fatty acids is related to plasma biomarkers of inflammation and endothelial activation in women. J Nutr. 2004;134(7):1806–1811
  119. Zampelas, A., Panagiotakos, D. B., Pitsavos, C., et al. Fish consumption among healthy adults is associated with decreased levels of inflammatory markers related to cardiovascular disease: the ATTICA study. J Am Coll Cardiol. 2005;46(1):120–124.
  120. He, K., Liu, K., Daviglus, M. L., et al. Associations of dietary long-chain n-3 polyunsaturated fatty acids and fish with biomarkers of inflammation and endothelial activation (from the Multi-Ethnic Study of Atherosclerosis [MESA]). Am J Cardiol. 2009;103(9):1238–1243
  121. Stracke H et al. Benfotiamine in diabetic polyneuropathy (BENDIP): results of a randomised, double blind, placebo-controlled clinical study. Exp Clin Endocrinol Diabetes. 2008 Nov;116(10):600-5. Epub 2008 May 13.
  122. Su J, Wu D, Lichtenstein AH, et al. EPA and DHA differentially modulate monocyte inflammatory response in subjects with chronic inflammation in part via plasma specialized pro-resolving lipid mediators: A randomized, double-blind, crossover study. Atherosclerosis. 2021;316:90-98. doi: 10.1016/j.atherosclerosis.2020.11.018. https://pubmed.ncbi.nlm.nih.gov/33303222/
  123. Chakraborty AJ, Mitra S, Tallei TE, et al. Bromelain a Potential Bioactive Compound: A Comprehensive Overview from a Pharmacological Perspective. Life (Basel). Apr 6 2021;11(4)doi:10.3390/life11040317
  124. Colletti A, Li S, Marengo M, Adinolfi S, Cravotto G. Recent Advances and Insights into Bromelain Processing, Pharmacokinetics and Therapeutic Uses. Applied Sciences. 2021;11(18):8428.
  125. de Souza GM, Fernandes IA, Dos Santos CRR, Falci SGM. Is bromelain effective in controlling the inflammatory parameters of pain, edema, and trismus after lower third molar surgery? A systematic review and meta-analysis. Phytother Res. Mar 2019;33(3):473-481. doi:10.1002/ptr.6244
  126. Walker AF, Bundy R, Hicks SM, Middleton RW. Bromelain reduces mild acute knee pain and improves well-being in a dose-dependent fashion in an open study of otherwise healthy adults. Phytomedicine. Dec 2002;9(8):681-6. doi:10.1078/094471102321621269
  127. Jayachandran S, Khobre P. Efficacy of Bromelain along with Trypsin, Rutoside Trihydrate Enzymes and Diclofenac Sodium Combination Therapy for the treatment of TMJ Osteoarthritis - A Randomised Clinical Trial. Journal of clinical and diagnostic research : JCDR. Jun 2017;11(6):ZC09-ZC11. doi:10.7860/JCDR/2017/25771.9964
  128. Italiano G, Raimondo M, Giannetti G, Gargiulo A. Benefits of a Food Supplement Containing Boswellia serrata and Bromelain for Improving the Quality of Life in Patients with Osteoarthritis: A Pilot Study. Journal of alternative and complementary medicine (New York, NY). Feb 2020;26(2):123-129. doi:10.1089/acm.2019.0258
  129. National Institute on Deafness and Other Communication Disorders. Taking Care of Your Voice. Updated 4/15/2021. Accessed 10/1/2021, https://www.nidcd.nih.gov/health/taking-care-your-voice
  130. Centre ICE. The voice and its disorders in teachers. INSERM Collective Expert Reports. Institut national de la santé et de la recherche médicale. Copyright © 2000-, Institut national de la santé et de la recherche médicale (INSERM).
  131. Baylor University. How to Prevent and Repair Vocal Damage. Updated 3/26/2020. Accessed 10/1/2021, https://onlinegrad.baylor.edu/resources/how-to-prevent-and-repair-vocal-damage-for-teachers-fitness-instructors-podcast-hosts-more/
  132. Zambito P, Bennici E. THE EFFICACY OF EVAN® IN POST-OPERATIVE TREATMENT OF BENIGN CORDAL INJURIES: AN OBSERVATIONAL PILOT STUDY. Accessed 09/28/2021, http://www.frontieraorl.it/en/772-2/
  133. Hancock RL, Stemple JC. Vocal fold hemorrhage: when vocal fold blood vessels are not up to the task! Voice and Speech Review. 2014/01/02 2014;8(1):36-39. doi:10.1080/23268263.2013.863048
  134. Al-Ali M, Anderson J. The role of steroid injection for vocal folds lesions in professional voice users. J Otolaryngol Head Neck Surg. Jul 20 2020;49(1):50. doi:10.1186/s40463-020-00434-5
  135. Insuan O, Janchai P, Thongchuai B, et al. Anti-Inflammatory Effect of Pineapple Rhizome Bromelain through Downregulation of the NF-kappaB- and MAPKs-Signaling Pathways in Lipopolysaccharide (LPS)-Stimulated RAW264.7 Cells. Current issues in molecular biology. May 7 2021;43(1):93-106. doi:10.3390/cimb43010008
  136. Paknezhad H, Ramadan O, Sataloff RT. Vocal Fold Hemorrhage. Ear, Nose & Throat Journal. 2021/07/01 2019;100(6):NP276-NP277. doi:10.1177/0145561319869914
  137. Annamaraju P, Baradhi KM. Pentoxifylline. StatPearls. StatPearls Publishing Copyright © 2023, StatPearls Publishing LLC.; 2023.
  138. Gonzalez-Espinoza L, Rojas-Campos E, Medina-Perez M, Pena-Quintero P, Gomez-Navarro B, Cueto-Manzano AM. Pentoxifylline decreases serum levels of tumor necrosis factor alpha, interleukin 6 and C-reactive protein in hemodialysis patients: results of a randomized double-blind, controlled clinical trial. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. May 2012;27(5):2023-8. doi:10.1093/ndt/gfr579. https://www.ncbi.nlm.nih.gov/pubmed/21968012
  139. Navarro-Gonzalez JF, Mora-Fernandez C, Muros de Fuentes M, et al. Effect of pentoxifylline on renal function and urinary albumin excretion in patients with diabetic kidney disease: the PREDIAN trial. Journal of the American Society of Nephrology : JASN. Jan 2015;26(1):220-9. doi:10.1681/ASN.2014010012. https://www.ncbi.nlm.nih.gov/pubmed/24970885
  140. Mansourian S, Bina P, Fehri A, Karimi AA, Boroumand MA, Abbasi K. Preoperative oral pentoxifylline in case of coronary artery bypass grafting with left ventricular dysfunction (ejection fraction equal to/less than 30%). Anatolian journal of cardiology. 2015;15(12):1014-9. doi:10.5152/akd.2014.5883. https://www.ncbi.nlm.nih.gov/pubmed/25880052
  141. Fernandes JL, de Oliveira RTD, Mamoni RL, et al. Pentoxifylline reduces pro-inflammatory and increases anti-inflammatory activity in patients with coronary artery disease--a randomized placebo-controlled study. Atherosclerosis. Jan 2008;196(1):434-442. doi:10.1016/j.atherosclerosis.2006.11.032. https://www.ncbi.nlm.nih.gov/pubmed/17196208
  142. Brie D, Sahebkar A, Penson PE, et al. Effects of pentoxifylline on inflammatory markers and blood pressure: a systematic review and meta-analysis of randomized controlled trials. J Hypertens. Dec 2016;34(12):2318-2329. doi:10.1097/HJH.0000000000001086. https://www.ncbi.nlm.nih.gov/pubmed/27512972
  143. Zeni F, Pain P, Vindimian M, et al. Effects of pentoxifylline on circulating cytokine concentrations and hemodynamics in patients with septic shock: results from a double-blind, randomized, placebo-controlled study. Critical care medicine. Feb 1996;24(2):207-14. doi:10.1097/00003246-199602000-00005. https://www.ncbi.nlm.nih.gov/pubmed/8605790
  144. Staubach KH, Schroder J, Stuber F, Gehrke K, Traumann E, Zabel P. Effect of pentoxifylline in severe sepsis: results of a randomized, double-blind, placebo-controlled study. Archives of surgery (Chicago, Ill : 1960). Jan 1998;133(1):94-100. doi:10.1001/archsurg.133.1.94. https://www.ncbi.nlm.nih.gov/pubmed/9438767
  145. Shabaan AE, Nasef N, Shouman B, Nour I, Mesbah A, Abdel-Hady H. Pentoxifylline therapy for late-onset sepsis in preterm infants: a randomized controlled trial. The Pediatric infectious disease journal. Jun 2015;34(6):e143-8. doi:10.1097/INF.0000000000000698. https://www.ncbi.nlm.nih.gov/pubmed/25970116
  146. Pammi M, Haque KN. Pentoxifylline for treatment of sepsis and necrotizing enterocolitis in neonates. Cochrane Database Syst Rev. Mar 9 2015;(3):CD004205. doi:10.1002/14651858.CD004205.pub3. https://www.ncbi.nlm.nih.gov/pubmed/25751631
  147. Lin S, Zhang A, Zhang X, Wu ZB. Treatment of Pituitary and Other Tumours with Cabergoline: New Mechanisms and Potential Broader Applications. Neuroendocrinology. 2020;110(6):477-488. doi:10.1159/000504000. https://www.ncbi.nlm.nih.gov/pubmed/31597135
  148. You L, Jiang H. Cabergoline possesses a beneficial effect on blood-brain barrier (BBB) integrity against lipopolysaccharide (LPS). Bioengineered. Dec 2021;12(1):8358-8369. doi:10.1080/21655979.2021.1987066. https://www.ncbi.nlm.nih.gov/pubmed/34592907
  149. Yamamoto K, Hanayama Y, Hasegawa K, et al. Clinical Relevance of Serum Prolactin Levels to Inflammatory Reaction in Male Patients. Acta Med Okayama. Oct 2020;74(5):381-389. doi:10.18926/AMO/60797. https://www.ncbi.nlm.nih.gov/pubmed/33106693
  150. Nguyen DN, Huyghens L, Schiettecatte J, Smitz J, Vincent JL. High prolactin levels are associated with more delirium in septic patients. Journal of critical care. Jun 2016;33:56-61. doi:10.1016/j.jcrc.2015.12.021. https://www.ncbi.nlm.nih.gov/pubmed/26852394
  151. El-Wakeel N, Hazzaa H, Gawish AS. Hypothesis: Rheumatoid arthritis and periodontitis: A new possible link via prolactin hormone. Med Hypotheses. Jan 2021;146:110350. doi:10.1016/j.mehy.2020.110350. https://www.ncbi.nlm.nih.gov/pubmed/33189451
  152. Serri O, Li L, Mamputu JC, Beauchamp MC, Maingrette F, Renier G. The influences of hyperprolactinemia and obesity on cardiovascular risk markers: effects of cabergoline therapy. Clin Endocrinol (Oxf). Apr 2006;64(4):366-70. doi:10.1111/j.1365-2265.2006.02469.x. https://www.ncbi.nlm.nih.gov/pubmed/16584506
  153. Inancli SS, Usluogullari A, Ustu Y, et al. Effect of cabergoline on insulin sensitivity, inflammation, and carotid intima media thickness in patients with prolactinoma. Endocrine. Aug 2013;44(1):193-9. doi:10.1007/s12020-012-9857-y. https://www.ncbi.nlm.nih.gov/pubmed/23233277
  154. Kany S, Vollrath JT, Relja B. Cytokines in Inflammatory Disease. International journal of molecular sciences. Nov 28 2019;20(23)doi:10.3390/ijms20236008. https://mdpi-res.com/d_attachment/ijms/ijms-20-06008/article_deploy/ijms-20-06008.pdf?version=1574957092
  155. Atalay Ekiner S, Gęgotek A, Skrzydlewska E. Inflammasome activity regulation by PUFA metabolites. Front Immunol. 2024;15:1452749. doi:10.3389/fimmu.2024.1452749. https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2024.1452749/pdf
  156. Soares CLR, Wilairatana P, Silva LR, et al. Biochemical aspects of the inflammatory process: A narrative review. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. Dec 2023;168:115764. doi:10.1016/j.biopha.2023.115764. https://www.sciencedirect.com/science/article/pii/S0753332223015627?via%3Dihub
  157. Mehta NN, Dey AK, Maddineni R, Kraus WE, Huffman KM. GlycA measured by NMR spectroscopy is associated with disease activity and cardiovascular disease risk in chronic inflammatory diseases. American Journal of Preventive Cardiology. 2020/12/01/ 2020;4:100120. doi:https://doi.org/10.1016/j.ajpc.2020.100120. https://www.sciencedirect.com/science/article/pii/S2666667720301203
  158. Ilari S, Proietti S, Milani F, et al. Dietary Patterns, Oxidative Stress, and Early Inflammation: A Systematic Review and Meta-Analysis Comparing Mediterranean, Vegan, and Vegetarian Diets. Nutrients. Jan 31 2025;17(3)doi:10.3390/nu17030548. https://mdpi-res.com/d_attachment/nutrients/nutrients-17-00548/article_deploy/nutrients-17-00548.pdf?version=1738340849
  159. Thompson G, Davison GW, Crawford J, Hughes CM. Exercise and inflammation in coronary artery disease: A systematic review and meta-analysis of randomised trials. Journal of sports sciences. Apr 2020;38(7):814-826. doi:10.1080/02640414.2020.1735684.
  160. Ceylan MN, Akdas S, Yazihan N. The Effects of Zinc Supplementation on C-Reactive Protein and Inflammatory Cytokines: A Meta-Analysis and Systematical Review. Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research. Mar 2021;41(3):81-101. doi:10.1089/jir.2020.0209. https://www.liebertpub.com/doi/10.1089/jir.2020.0209?url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org&rfr_dat=cr_pub++0pubmed
  161. Kumar NN, Ahmad Dit Al Hakim S, Grygiel-Górniak B. Antinuclear Antibodies in Non-Rheumatic Diseases. Arch Immunol Ther Exp (Warsz). Jan 1 2025;73(1)doi:10.2478/aite-2025-0004. https://intapi.sciendo.com:443/pdf/10.2478/aite-2025-0004
  162. Kraev K, Hristov B, Uchikov P, et al. Comprehensive Exploration of Antinuclear Antibodies (ANAs): Unveiling Clinical Significance, Associations with Cancer, and the Nuances of Differential Diagnosis in Positive ANA Patients. Diagnostics (Basel). Feb 1 2024;14(3)doi:10.3390/diagnostics14030320. https://pmc.ncbi.nlm.nih.gov/articles/PMC10855796/pdf/diagnostics-14-00320.pdf
  163. Li X, Liu X, Cui J, et al. Epidemiological survey of antinuclear antibodies in healthy population and analysis of clinical characteristics of positive population. Journal of Clinical Laboratory Analysis. 2019;33(8):e22965. doi:https://doi.org/10.1002/jcla.22965. https://onlinelibrary.wiley.com/doi/abs/10.1002/jcla.22965
  164. Ge Q, Gu X, Yu W, et al. Antinuclear antibodies in healthy population: Positive association with abnormal tissue metabolism, inflammation and immune dysfunction. International immunopharmacology. Dec 2022;113(Pt A):109292. doi:10.1016/j.intimp.2022.109292. https://www.sciencedirect.com/science/article/pii/S1567576922007767?via%3Dihub
  165. Gupta R, Vanlieshout E, Manion K, et al. Altered Balance of Pro-Inflammatory Immune Cells to T Regulatory Cells Differentiates Symptomatic From Asymptomatic Individuals With Anti-Nuclear Antibodies. Front Immunol. 2022;13:886442. doi:10.3389/fimmu.2022.886442. https://pmc.ncbi.nlm.nih.gov/articles/PMC9279569/pdf/fimmu-13-886442.pdf
  166. Dinse GE, Parks CG, Weinberg CR, et al. Increasing Prevalence of Antinuclear Antibodies in the United States. Arthritis Rheumatol. Dec 2022;74(12):2032-2041. doi:10.1002/art.42330. https://pmc.ncbi.nlm.nih.gov/articles/PMC10947520/pdf/nihms-1970346.pdf
  167. Singh U, Devaraj S, Jialal I. C-reactive protein decreases tissue plasminogen activator activity in human aortic endothelial cells: evidence that C-reactive protein is a procoagulant. Arteriosclerosis, thrombosis, and vascular biology. Oct 2005;25(10):2216-21. doi:10.1161/01.ATV.0000183718.62409.ea. https://www.ahajournals.org/doi/pdf/10.1161/01.ATV.0000183718.62409.ea?download=true
  168. Cirillo P, Golino P, Calabrò P, et al. C-reactive protein induces tissue factor expression and promotes smooth muscle and endothelial cell proliferation. Cardiovasc Res. Oct 1 2005;68(1):47-55. doi:10.1016/j.cardiores.2005.05.010. https://academic.oup.com/cardiovascres/article-abstract/68/1/47/288145?redirectedFrom=fulltext
  169. Melnikov IS, Kozlov SG, Saburova OS, Avtaeva YN, Prokofieva LV, Gabbasov ZA. Current Position on the Role of Monomeric C-reactive Protein in Vascular Pathology and Atherothrombosis. Curr Pharm Des. 2020;26(1):37-43. doi:10.2174/1381612825666191216144055. https://www.eurekaselect.com/article/103009
  170. Badimon L, Peña E, Arderiu G, et al. C-Reactive Protein in Atherothrombosis and Angiogenesis. Front Immunol. 2018;9:430. doi:10.3389/fimmu.2018.00430. https://pmc.ncbi.nlm.nih.gov/articles/PMC5840191/pdf/fimmu-09-00430.pdf
  171. Zhou HH, Tang YL, Xu TH, Cheng B. C-reactive protein: structure, function, regulation, and role in clinical diseases. Front Immunol. 2024;15:1425168. doi:10.3389/fimmu.2024.1425168. https://pmc.ncbi.nlm.nih.gov/articles/PMC11211361/pdf/fimmu-15-1425168.pdf
  172. Meier HCS, Mitchell C, Karadimas T, Faul JD. Systemic inflammation and biological aging in the Health and Retirement Study. GeroScience. Dec 2023;45(6):3257-3265. doi:10.1007/s11357-023-00880-9. https://pmc.ncbi.nlm.nih.gov/articles/PMC10643484/pdf/11357_2023_Article_880.pdf
  173. Lassale C, Batty GD, Steptoe A, et al. Association of 10-Year C-Reactive Protein Trajectories With Markers of Healthy Aging: Findings From the English Longitudinal Study of Aging. The journals of gerontology Series A, Biological sciences and medical sciences. Jan 16 2019;74(2):195-203. doi:10.1093/gerona/gly028. https://pmc.ncbi.nlm.nih.gov/articles/PMC6333942/pdf/gly028.pdf
  174. Ali S, Zehra A, Khalid MU, Hassan M, Shah SIA. Role of C-reactive protein in disease progression, diagnosis and management. Discoveries (Craiova). Oct-Dec 2023;11(4):e179. doi:10.15190/d.2023.18. https://pmc.ncbi.nlm.nih.gov/articles/PMC11569793/pdf/discoveries-11-179.pdf
  175. Harwood JL. Polyunsaturated Fatty Acids: Conversion to Lipid Mediators, Roles in Inflammatory Diseases and Dietary Sources. International journal of molecular sciences. May 16 2023;24(10)doi:10.3390/ijms24108838. https://mdpi-res.com/d_attachment/ijms/ijms-24-08838/article_deploy/ijms-24-08838.pdf?version=1684226470
  176. FDA. Food and Drug Adminstration. Highlights of Prescribing Information. Zyflo CR (zileuon) extended-release tablets. Updated 12/2018. Accessed 9/25/2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/022052s014lbl.pdf
  177. Kaur B, Singh P. Inflammation: Biochemistry, cellular targets, anti-inflammatory agents and challenges with special emphasis on cyclooxygenase-2. Bioorganic chemistry. Apr 2022;121:105663. doi:10.1016/j.bioorg.2022.105663. https://www.ncbi.nlm.nih.gov/pubmed/35180488
  178. Valentovic M. Zileuton☆. Reference Module in Biomedical Sciences. Elsevier; 2018.
  179. Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. May 2008;8(5):349-61. doi:10.1038/nri2294. https://pmc.ncbi.nlm.nih.gov/articles/PMC2744593/
  180. Grosser T, Fries S, FitzGerald GA. Biological basis for the cardiovascular consequences of COX-2 inhibition: therapeutic challenges and opportunities. J Clin Invest. Jan 2006;116(1):4-15. doi:10.1172/jci27291. https://dm5migu4zj3pb.cloudfront.net/manuscripts/27000/27291/JCI0627291.pdf
  181. Wallace JL. Prostaglandins, NSAIDs, and Gastric Mucosal Protection: Why Doesn't the Stomach Digest Itself? Physiological Reviews. 2008;88(4):1547-1565. doi:10.1152/physrev.00004.2008. https://journals.physiology.org/doi/abs/10.1152/physrev.00004.2008
  182. Chandrasekharan JA, Sharma-Walia N. Lipoxins: nature's way to resolve inflammation. J Inflamm Res. 2015;8:181-92. doi:10.2147/jir.S90380. https://www.dovepress.com/article/download/23904
  183. Afroz M, Zihad S, Uddin SJ, et al. A systematic review on antioxidant and antiinflammatory activity of Sesame (Sesamum indicum L.) oil and further confirmation of antiinflammatory activity by chemical profiling and molecular docking. Phytother Res. Oct 2019;33(10):2585-2608. doi:10.1002/ptr.6428.
  184. Hartung NM, Mainka M, Pfaff R, et al. Development of a quantitative proteomics approach for cyclooxygenases and lipoxygenases in parallel to quantitative oxylipin analysis allowing the comprehensive investigation of the arachidonic acid cascade. Anal Bioanal Chem. Feb 2023;415(5):913-933. doi:10.1007/s00216-022-04489-3. https://pmc.ncbi.nlm.nih.gov/articles/PMC9883352/pdf/216_2022_Article_4489.pdf
  185. Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang N, Gronert K. Novel functional sets of lipid-derived mediators with antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal antiinflammatory drugs and transcellular processing. J Exp Med. Oct 16 2000;192(8):1197-204. doi:10.1084/jem.192.8.1197. https://pmc.ncbi.nlm.nih.gov/articles/PMC2195872/pdf/000832.pdf
  186. Groeger AL, Cipollina C, Cole MP, et al. Cyclooxygenase-2 generates anti-inflammatory mediators from omega-3 fatty acids. Nat Chem Biol. Jun 2010;6(6):433-41. doi:10.1038/nchembio.367. https://pmc.ncbi.nlm.nih.gov/articles/PMC3077712/pdf/nihms283532.pdf
  187. Calder PC. Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem Soc Trans. Oct 15 2017;45(5):1105-1115. doi:10.1042/BST20160474. https://www.ncbi.nlm.nih.gov/pubmed/28900017
  188. Harris WS. Recent studies confirm the utility of the omega-3 index. Current opinion in clinical nutrition and metabolic care. Mar 1 2025;28(2):91-95. doi:10.1097/mco.0000000000001078. https://pmc.ncbi.nlm.nih.gov/articles/PMC11809719/pdf/cocnm-28-91.pdf
  189. Wolberg AS. Fibrinogen and fibrin: synthesis, structure, and function in health and disease. J Thromb Haemost. Nov 2023;21(11):3005-3015. doi:10.1016/j.jtha.2023.08.014. https://www.jthjournal.org/action/showPdf?pii=S1538783623006426
  190. Luyendyk JP, Schoenecker JG, Flick MJ. The multifaceted role of fibrinogen in tissue injury and inflammation. Blood. Feb 7 2019;133(6):511-520. doi:10.1182/blood-2018-07-818211. https://pubmed.ncbi.nlm.nih.gov/30523120/
  191. Lan H, Zhao S, Xiong Y, Yan XZ. The emerging role of fibrin(ogen) in cardiovascular disease. Inflammation research : official journal of the European Histamine Research Society [et al]. Sep 2024;73(9):1435-1444. doi:10.1007/s00011-024-01916-2. https://link.springer.com/article/10.1007/s00011-024-01916-2
  192. Liang D, Liang D, Liu J, et al. Impact of lipoprotein(a) and fibrinogen on prognosis in patients with coronary artery disease: A retrospective cohort study. Technol Health Care. 2024;32(5):3317-3328. doi:10.3233/THC-240005. https://www.ncbi.nlm.nih.gov/pubmed/38848204
  193. Ndrepepa G, Braun S, King L, et al. Relation of fibrinogen level with cardiovascular events in patients with coronary artery disease. The American journal of cardiology. Mar 15 2013;111(6):804-10. doi:10.1016/j.amjcard.2012.11.060. https://www.ncbi.nlm.nih.gov/pubmed/23291088
  194. Su H, Cao Y, Chen Q, et al. The association between fibrinogen levels and severity of coronary artery disease and long-term prognosis following percutaneous coronary intervention in patients with type 2 diabetes mellitus. Frontiers in endocrinology. 2023;14:1287855. doi:10.3389/fendo.2023.1287855. https://www.ncbi.nlm.nih.gov/pubmed/38093962
  195. Kryczka KE, Kruk M, Demkow M, Lubiszewska B. Fibrinogen and a Triad of Thrombosis, Inflammation, and the Renin-Angiotensin System in Premature Coronary Artery Disease in Women: A New Insight into Sex-Related Differences in the Pathogenesis of the Disease. Biomolecules. Jul 15 2021;11(7)doi:10.3390/biom11071036. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-11-01036/article_deploy/biomolecules-11-01036-v2.pdf?version=1626423946
  196. Hu J, Xu D, Zeng H, Chen Y, Zhu X. Preoperative plasma fibrinogen level is a risk factor for the long-term survival of postmenopausal women after surgery for breast cancer. Maturitas. Nov 2024;189:108108. doi:10.1016/j.maturitas.2024.108108. https://www.ncbi.nlm.nih.gov/pubmed/39241485
  197. Guven B, Can M, Tekin A. Comparison of Fibrinogen Concentrations Determined by the Clauss Method with Prothrombin-Derived Measurements on an Automated Coagulometer. J Appl Lab Med. Oct 29 2022;7(6):1337-1345. doi:10.1093/jalm/jfac066. https://pubmed.ncbi.nlm.nih.gov/35993826/
  198. Mackie I, Casini A, Pieters M, Pruthi R, Reilly-Stitt C, Suzuki A. International council for standardisation in haematology recommendations on fibrinogen assays, thrombin clotting time and related tests in the investigation of bleeding disorders. International Journal of Laboratory Hematology. 2024;46(1):20-32. doi:https://doi.org/10.1111/ijlh.14201. https://onlinelibrary.wiley.com/doi/abs/10.1111/ijlh.14201
  199. Wang Z, Yan X, Fang L, Tang J, Zhang J. Association between lipoprotein(a), fibrinogen and their combination with all-cause, cardiovascular disease and cancer-related mortality: findings from the NHANES. BMC Public Health. Jul 18 2024;24(1):1927. doi:10.1186/s12889-024-19443-4. https://www.ncbi.nlm.nih.gov/pubmed/39026192
  200. Wu K, Wang J, Li X, et al. Association between fibrinogen and cognitive impairment in patients with ischemic cerebrovascular disease. J Stroke Cerebrovasc Dis. May 2025;34(5):108227. doi:10.1016/j.jstrokecerebrovasdis.2025.108227. https://www.ncbi.nlm.nih.gov/pubmed/39952449
  201. Yousefi O, Farrokhi A, Taheri R, et al. Effect of low fibrinogen level on in-hospital mortality and 6-month functional outcome of TBI patients, a single center experience. Neurosurg Rev. Feb 28 2024;47(1):95. doi:10.1007/s10143-024-02326-y. https://www.ncbi.nlm.nih.gov/pubmed/38413402
  202. Bouffette S, Botez I, De Ceuninck F. Targeting galectin-3 in inflammatory and fibrotic diseases. Trends Pharmacol Sci. Aug 2023;44(8):519-531. doi:10.1016/j.tips.2023.06.001. https://www.cell.com/action/showPdf?pii=S0165614723001268
  203. Mahalleh M, Behnoush AH, Khalaji A, et al. Serum Galectin-3 Level in Patients with Rheumatoid Arthritis: A Systematic Review and Meta-analysis. Iranian journal of allergy, asthma, and immunology. Oct 6 2024;23(5):476-488. doi:10.18502/ijaai.v23i5.16777. https://ijaai.tums.ac.ir/index.php/ijaai/article/download/4160/2114
  204. Li Y, Li T, Zhou Z, Xiao Y. Emerging roles of Galectin-3 in diabetes and diabetes complications: A snapshot. Reviews in endocrine & metabolic disorders. Jun 2022;23(3):569-577. doi:10.1007/s11154-021-09704-7. https://pmc.ncbi.nlm.nih.gov/articles/PMC9156459/pdf/11154_2021_Article_9704.pdf
  205. Bellos I, Marinaki S, Lagiou P, Benetou V. Association of serum galectin-3 levels with mortality and cardiovascular disease outcomes in hemodialysis patients: a systematic review and dose-response meta-analysis. International urology and nephrology. Aug 2024;56(8):2755-2767. doi:10.1007/s11255-024-04026-4. https://pmc.ncbi.nlm.nih.gov/articles/PMC11266241/pdf/11255_2024_Article_4026.pdf
  206. Jeethy Ram T, Lekshmi A, Somanathan T, Sujathan K. Galectin-3: A factotum in carcinogenesis bestowing an archery for prevention. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2021;43(1):77-96. doi:10.3233/tub-200051. https://content.iospress.com:443/download/tumor-biology/tub200051?id=tumor-biology%2Ftub200051
  207. Capone E, Iacobelli S, Sala G. Role of galectin 3 binding protein in cancer progression: a potential novel therapeutic target. Journal of translational medicine. Sep 26 2021;19(1):405. doi:10.1186/s12967-021-03085-w. https://translational-medicine.biomedcentral.com/counter/pdf/10.1186/s12967-021-03085-w.pdf
  208. Zaborska B, Sikora-Frąc M, Smarż K, et al. The Role of Galectin-3 in Heart Failure-The Diagnostic, Prognostic and Therapeutic Potential-Where Do We Stand? International journal of molecular sciences. Aug 23 2023;24(17)doi:10.3390/ijms241713111. https://mdpi-res.com/d_attachment/ijms/ijms-24-13111/article_deploy/ijms-24-13111.pdf?version=1692786300
  209. Procyk G, Czapla A, Jałocha K, Tymińska A, Grabowski M, Gąsecka A. The role of galectin-3 in atrial fibrillation. Journal of molecular medicine (Berlin, Germany). Dec 2023;101(12):1481-1492. doi:10.1007/s00109-023-02378-5. https://pmc.ncbi.nlm.nih.gov/articles/PMC10698102/pdf/109_2023_Article_2378.pdf
  210. Mohtasham Kia Y, Cannavo A, Bahiraie P, et al. Insights into the Role of Galectin-3 as a Diagnostic and Prognostic Biomarker of Atrial Fibrillation. Disease markers. 2023;2023:2097012. doi:10.1155/2023/2097012. https://onlinelibrary.wiley.com/doi/pdfdirect/10.1155/2023/2097012?download=true
  211. Baccouche BM, Mahmoud MA, Nief C, Patel K, Natterson-Horowitz B. Galectin-3 is Associated with Heart Failure Incidence: A Meta-Analysis. Current cardiology reviews. 2023;19(3):e171122211004. doi:10.2174/1573403x19666221117122012. https://pmc.ncbi.nlm.nih.gov/articles/PMC10280995/pdf/CCR-19-E171122211004.pdf
  212. Ahmed R, Anam K, Ahmed H. Development of Galectin-3 Targeting Drugs for Therapeutic Applications in Various Diseases. International journal of molecular sciences. May 1 2023;24(9)doi:10.3390/ijms24098116. https://mdpi-res.com/d_attachment/ijms/ijms-24-08116/article_deploy/ijms-24-08116-v3.pdf?version=1683602804
  213. Shah A, Ding Y, Walji D, Rabinovich GA, Pelletier M, El-Diasty M. The prognostic utility of galectin-3 in patients undergoing cardiac surgery: a scoping review. Biomarkers. Nov 2024;29(7):485-493. doi:10.1080/1354750x.2024.2415073.
  214. Seropian IM, El-Diasty M, El-Sherbini AH, González GE, Rabinovich GA. Central role of Galectin-3 at the cross-roads of cardiac inflammation and fibrosis: Implications for heart failure and transplantation. Cytokine & growth factor reviews. Dec 2024;80:47-58. doi:10.1016/j.cytogfr.2024.10.002. https://www.sciencedirect.com/science/article/abs/pii/S1359610124000819?via%3Dihub
  215. Soares LC, Al-Dalahmah O, Hillis J, et al. Novel Galectin-3 Roles in Neurogenesis, Inflammation and Neurological Diseases. Cells. Nov 5 2021;10(11)doi:10.3390/cells10113047. https://mdpi-res.com/d_attachment/cells/cells-10-03047/article_deploy/cells-10-03047-v2.pdf?version=1636381422
  216. Chiesa ST, Charakida M, Georgiopoulos G, et al. Glycoprotein Acetyls: A Novel Inflammatory Biomarker of Early Cardiovascular Risk in the Young. J Am Heart Assoc. Feb 15 2022;11(4):e024380. doi:10.1161/jaha.121.024380. https://www.ahajournals.org/doi/pdf/10.1161/JAHA.121.024380?download=true
  217. Kasher M, Freidin MB, Williams FMK, Cherny SS, Ashkenazi S, Livshits G. Glycoprotein Acetyls Is a Novel Biomarker Predicting Cardiovascular Complications in Rheumatoid Arthritis. International journal of molecular sciences. May 30 2024;25(11)doi:10.3390/ijms25115981. https://mdpi-res.com/d_attachment/ijms/ijms-25-05981/article_deploy/ijms-25-05981.pdf?version=1717048832
  218. Mansell T, Bekkering S, Longmore D, et al. Change in adiposity is associated with change in glycoprotein acetyls but not hsCRP in adolescents with severe obesity. Obes Res Clin Pract. Jul-Aug 2023;17(4):343-348. doi:10.1016/j.orcp.2023.08.003. https://www.sciencedirect.com/science/article/pii/S1871403X23000686?via%3Dihub
  219. Crick DCP, Khandaker GM, Halligan SL, Burgner D, Mansell T, Fraser A. Comparison of the stability of glycoprotein acetyls and high sensitivity C-reactive protein as markers of chronic inflammation. Immunology. Apr 2024;171(4):497-512. doi:10.1111/imm.13739. https://pmc.ncbi.nlm.nih.gov/articles/PMC7616614/pdf/EMS198608.pdf
  220. Kasher M, Freidin MB, Williams FMK, Livshits G, Charge Inflammation Working G. GlycA and CRP Are Genetically Correlated: Insight into the Genetic Architecture of Inflammageing. Biomolecules. May 8 2024;14(5)doi:10.3390/biom14050563. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-14-00563/article_deploy/biomolecules-14-00563-v2.pdf?version=1715344050
  221. Filho RCF, Generoso G, Cardoso R, et al. GlycA Levels Independently Predict Coronary Artery Calcium Incidence and Progression in the ELSA-Brasil Cohort (Brazilian Longitudinal Study of Adult Health). The American journal of cardiology. Jun 15 2024;221:29-36. doi:10.1016/j.amjcard.2024.03.041. https://www.ajconline.org/article/S0002-9149(24)00265-0/abstract
  222. Tebar WR, Meneghini V, Goulart AC, et al. Combined Association of Novel and Traditional Inflammatory Biomarkers With Carotid Artery Plaque: GlycA Versus C-Reactive Protein (ELSA-Brasil). The American journal of cardiology. Oct 1 2023;204:140-150. doi:10.1016/j.amjcard.2023.07.034. https://www.ajconline.org/article/S0002-9149(23)00560-X/abstract
  223. Liang N, Nho K, Newman JW, et al. Peripheral inflammation is associated with brain atrophy and cognitive decline linked to mild cognitive impairment and Alzheimer's disease. Sci Rep. Jul 29 2024;14(1):17423. doi:10.1038/s41598-024-67177-5. https://www.nature.com/articles/s41598-024-67177-5.pdf
  224. Calice-Silva V, Suemoto CK, Brunoni AR, Bensenor IM, Lotufo PA. Association Between GlycA and Cognitive Function: Cross-Sectional Results From the ELSA-Brasil Study. Alzheimer Dis Assoc Disord. Apr-Jun 01 2021;35(2):128-134. doi:10.1097/wad.0000000000000431.
  225. Choi Y, Jacobs DR, Jr., Kramer HJ, Shroff GR, Chang AR, Duprez DA. Nontraditional Risk Factors for Progression Through Chronic Kidney Disease Risk Categories: The Coronary Artery Risk Development in Young Adults Study. Am J Med. Apr 2023;136(4):380-389.e10. doi:10.1016/j.amjmed.2022.12.001.
  226. Huckvale S, Reyes S, Kulikova A, Rohatgi A, Riggs KA, Brown ES. An Association Between the Inflammatory Biomarker GlycA and Depressive Symptom Severity. The Journal of clinical psychiatry. Nov 17 2020;82(1)doi:10.4088/JCP.20m13245.
  227. Llop D, Paredes S, Rosales R, et al. Comprehensive analysis of glycoprotein profiles and their association with cardiovascular disease-related microRNAs in rheumatoid arthritis, metabolic disorders, and controls. Sci Rep. Nov 2 2024;14(1):26471. doi:10.1038/s41598-024-77772-1. https://www.nature.com/articles/s41598-024-77772-1.pdf
  228. Guo Y, Liu Q, Zheng Z, et al. Genetic association of inflammatory marker GlycA with lung function and respiratory diseases. Nature communications. May 4 2024;15(1):3751. doi:10.1038/s41467-024-47845-w. https://www.nature.com/articles/s41467-024-47845-w.pdf
  229. Seeling KS, Hehl L, Vell MS, et al. Comorbidities, biomarkers and cause specific mortality in patients with irritable bowel syndrome: A phenome-wide association study. United European Gastroenterol J. Jun 2023;11(5):458-470. doi:10.1002/ueg2.12397. https://pmc.ncbi.nlm.nih.gov/articles/PMC10256994/pdf/UEG2-11-458.pdf
  230. Moreno-Vedia J, Rosales R, Ozcariz E, et al. Triglyceride-Rich Lipoproteins and Glycoprotein A and B Assessed by 1H-NMR in Metabolic-Associated Fatty Liver Disease. Frontiers in endocrinology. 2021;12:775677. doi:10.3389/fendo.2021.775677.
  231. Connelly MA, Gruppen EG, Wolak-Dinsmore J, et al. GlycA, a marker of acute phase glycoproteins, and the risk of incident type 2 diabetes mellitus: PREVEND study. Clin Chim Acta. Jan 15 2016;452:10-7. doi:10.1016/j.cca.2015.11.001.
  232. Bourgonje AR, van der Vaart A, Gruppen EG, et al. Plasma levels of GlycA, a pro-inflammatory glycoprotein biomarker, associate with an increased risk of microvascular complications in patients with type 2 diabetes (Zodiac-62). Endocrine. May 2023;80(2):312-316. doi:10.1007/s12020-023-03319-5. https://link.springer.com/article/10.1007/s12020-023-03319-5
  233. Gruppen EG, Kunutsor SK, Kieneker LM, et al. GlycA, a novel pro-inflammatory glycoprotein biomarker is associated with mortality: results from the PREVEND study and meta-analysis. J Intern Med. Nov 2019;286(5):596-609. doi:10.1111/joim.12953. https://pmc.ncbi.nlm.nih.gov/articles/PMC6851697/pdf/JOIM-286-596.pdf
  234. Chen Y, Ye X, Escames G, et al. The NLRP3 inflammasome: contributions to inflammation-related diseases. Cellular & molecular biology letters. Jun 27 2023;28(1):51. doi:10.1186/s11658-023-00462-9. https://cmbl.biomedcentral.com/counter/pdf/10.1186/s11658-023-00462-9.pdf
  235. Napodano C, Carnazzo V, Basile V, et al. NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases-A Lesson from Cytokine Storm Syndrome. International journal of molecular sciences. Nov 21 2023;24(23)doi:10.3390/ijms242316556. https://mdpi-res.com/d_attachment/ijms/ijms-24-16556/article_deploy/ijms-24-16556.pdf?version=1700547821
  236. Cheng R, Wu Z, Li M, Shao M, Hu T. Interleukin-1β is a potential therapeutic target for periodontitis: a narrative review. International journal of oral science. 2020/01/02 2020;12(1):2. doi:10.1038/s41368-019-0068-8. https://doi.org/10.1038/s41368-019-0068-8
  237. Forcina L, Franceschi C, Musarò A. The hormetic and hermetic role of IL-6. Ageing Res Rev. Sep 2022;80:101697. doi:10.1016/j.arr.2022.101697. https://www.sciencedirect.com/science/article/pii/S1568163722001398?via%3Dihub
  238. Byrne T, Cooke J, Bambrick P, McNeela E, Harrison M. Circulating inflammatory biomarker responses in intervention trials in frail and sarcopenic older adults: A systematic review and meta-analysis. Exp Gerontol. Jun 15 2023;177:112199. doi:10.1016/j.exger.2023.112199. https://www.sciencedirect.com/science/article/pii/S0531556523001201?via%3Dihub
  239. Custodero C, Ciavarella A, Panza F, et al. Role of inflammatory markers in the diagnosis of vascular contributions to cognitive impairment and dementia: a systematic review and meta-analysis. GeroScience. Jun 2022;44(3):1373-1392. doi:10.1007/s11357-022-00556-w. https://pmc.ncbi.nlm.nih.gov/articles/PMC9213626/pdf/11357_2022_Article_556.pdf
  240. Fard MT, Savage KM, Stough CK. Peripheral inflammation marker relationships to cognition in healthy older adults - A systematic review. Psychoneuroendocrinology. Oct 2022;144:105870. doi:10.1016/j.psyneuen.2022.105870. https://www.sciencedirect.com/science/article/abs/pii/S0306453022002116?via%3Dihub
  241. Tylutka A, Walas Ł, Zembron-Lacny A. Level of IL-6, TNF, and IL-1β and age-related diseases: a systematic review and meta-analysis. Front Immunol. 2024;15:1330386. doi:10.3389/fimmu.2024.1330386. https://pmc.ncbi.nlm.nih.gov/articles/PMC10943692/pdf/fimmu-15-1330386.pdf
  242. Chung HY, Kim DH, Lee EK, et al. Redefining Chronic Inflammation in Aging and Age-Related Diseases: Proposal of the Senoinflammation Concept. Aging Dis. Apr 2019;10(2):367-382. doi:10.14336/ad.2018.0324. https://pmc.ncbi.nlm.nih.gov/articles/PMC6457053/pdf/ad-10-2-367.pdf
  243. Xu S, Wang D, Tan L, Lu J. The role of NLRP3 inflammasome in type 2 inflammation related diseases. Autoimmunity. Dec 2024;57(1):2310269. doi:10.1080/08916934.2024.2310269. https://www.tandfonline.com/doi/pdf/10.1080/08916934.2024.2310269
  244. Yao J, Sterling K, Wang Z, Zhang Y, Song W. The role of inflammasomes in human diseases and their potential as therapeutic targets. Signal transduction and targeted therapy. 2024/01/05 2024;9(1):10. doi:10.1038/s41392-023-01687-y. https://doi.org/10.1038/s41392-023-01687-y
  245. den Hartigh LJ, May KS, Zhang XS, Chait A, Blaser MJ. Serum amyloid A and metabolic disease: evidence for a critical role in chronic inflammatory conditions. Front Cardiovasc Med. 2023;10:1197432. doi:10.3389/fcvm.2023.1197432.
  246. Eklund KK, Niemi K, Kovanen PT. Immune functions of serum amyloid A. Crit Rev Immunol. 2012;32(4):335-48. doi:10.1615/critrevimmunol.v32.i4.40.
  247. Webb NR. High-Density Lipoproteins and Serum Amyloid A (SAA). Curr Atheroscler Rep. Jan 15 2021;23(2):7. doi:10.1007/s11883-020-00901-4. https://www.ncbi.nlm.nih.gov/pubmed/33447953
  248. Stute M, Kreysing M, Zorn M, Michl P, Gauss A. Serum Amyloid A as a Potential Biomarker in Inflammatory Bowel Diseases, Especially in Patients with Low C-Reactive Protein. International journal of molecular sciences. Jan 18 2024;25(2)doi:10.3390/ijms25021177.
  249. Targońska-Stępniak B, Majdan M. Serum amyloid A as a marker of persistent inflammation and an indicator of cardiovascular and renal involvement in patients with rheumatoid arthritis. Mediators of inflammation. 2014;2014:793628-793628. doi:10.1155/2014/793628. https://www.ncbi.nlm.nih.gov/pubmed/25525305
  250. Yarur AJ, Quintero MA, Jain A, Czul F, Barkin JS, Abreu MT. Serum Amyloid A as a Surrogate Marker for Mucosal and Histologic Inflammation in Patients with Crohn's Disease. Inflammatory bowel diseases. Jan 2017;23(1):158-164. doi:10.1097/mib.0000000000000991.
  251. Getz GS, Krishack PA, Reardon CA. Serum amyloid A and atherosclerosis. Curr Opin Lipidol. Oct 2016;27(5):531-5. doi:10.1097/MOL.0000000000000331. https://www.ncbi.nlm.nih.gov/pubmed/27579547
  252. Shridas P, Patrick AC, Tannock LR. Role of Serum Amyloid A in Abdominal Aortic Aneurysm and Related Cardiovascular Diseases. Biomolecules. Dec 15 2021;11(12)doi:10.3390/biom11121883. https://www.ncbi.nlm.nih.gov/pubmed/34944527
  253. Zhou J, Lu Y, Wang S, Chen K. Association between serum amyloid A levels and coronary heart disease: a systematic review and meta-analysis of 26 studies. Inflammation research: official journal of the European Histamine Research Society [et al]. Apr 2020;69(4):331-345. doi:10.1007/s00011-020-01325-1.
  254. Holbrook J, Lara-Reyna S, Jarosz-Griffiths H, McDermott M. Tumour necrosis factor signalling in health and disease. F1000Research. 2019;8doi:10.12688/f1000research.17023.1. https://pubmed.ncbi.nlm.nih.gov/30755793/
  255. Tate AR, Rao GHR. Inflammation: Is It a Healer, Confounder, or a Promoter of Cardiometabolic Risks? Biomolecules. Aug 6 2024;14(8)doi:10.3390/biom14080948. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-14-00948/article_deploy/biomolecules-14-00948-v2.pdf?version=1722996799
  256. Gu L, Du Y, Liang F. Meta-Analysis and Network Analysis Differentially Detect Various Pro-Inflammatory Mediators and Risk Factors for Type 2 Diabetes in the Elderly. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme. Oct 2024;56(10):727-736. doi:10.1055/a-2241-5281. https://www.thieme-connect.com/products/ejournals/abstract/10.1055/a-2241-5281
  257. Su Z, Efremov L, Mikolajczyk R. Differences in the levels of inflammatory markers between metabolically healthy obese and other obesity phenotypes in adults: A systematic review and meta-analysis. Nutr Metab Cardiovasc Dis. Feb 2024;34(2):251-269. doi:10.1016/j.numecd.2023.09.002. https://www.nmcd-journal.com/action/showPdf?pii=S0939475323003484
  258. Yuan S, Carter P, Bruzelius M, et al. Effects of tumour necrosis factor on cardiovascular disease and cancer: A two-sample Mendelian randomization study. EBioMedicine. Sep 2020;59:102956. doi:10.1016/j.ebiom.2020.102956.
  259. Freeman ML, Hossain MB, Burrowes SAB, et al. Association of Soluble Markers of Inflammation With Peri-coronary Artery Inflammation in People With and Without HIV Infection and Without Cardiovascular Disease. Open forum infectious diseases. Aug 2023;10(8):ofad328. doi:10.1093/ofid/ofad328.
  260. Mattisson IY, Björkbacka H, Wigren M, et al. Elevated Markers of Death Receptor-Activated Apoptosis are Associated with Increased Risk for Development of Diabetes and Cardiovascular Disease. EBioMedicine. Dec 2017;26:187-197. doi:10.1016/j.ebiom.2017.11.023.
  261. Bhatraju PK, Zelnick LR, Shlipak M, Katz R, Kestenbaum B. Association of Soluble TNFR-1 Concentrations with Long-Term Decline in Kidney Function: The Multi-Ethnic Study of Atherosclerosis. Journal of the American Society of Nephrology : JASN. Nov 2018;29(11):2713-2721. doi:10.1681/asn.2018070719.
  262. Lu WH, Guyonnet S, Martinez LO, et al. Association between aging-related biomarkers and longitudinal trajectories of intrinsic capacity in older adults. GeroScience. Dec 2023;45(6):3409-3418. doi:10.1007/s11357-023-00906-2. https://pubmed.ncbi.nlm.nih.gov/37620614/
  263. Kartikasari AER, Cassar E, Razqan MAM, et al. Elevation of circulating TNF receptor 2 in cancer: A systematic meta-analysis for its potential as a diagnostic cancer biomarker. Front Immunol. 2022;13:918254. doi:10.3389/fimmu.2022.918254. https://pmc.ncbi.nlm.nih.gov/articles/PMC9708892/pdf/fimmu-13-918254.pdf
  264. Biswas M, Suvarna R, Krishnan SV, Devasia T, Shenoy Belle V, Prabhu K. The mechanistic role of neutrophil lymphocyte ratio perturbations in the leading non communicable lifestyle diseases. F1000Research. 2022;11:960. doi:10.12688/f1000research.123245.1. https://pmc.ncbi.nlm.nih.gov/articles/PMC9780608/pdf/f1000research-11-135331.pdf
  265. García-Escobar A, Vera-Vera S, Tébar-Márquez D, et al. Neutrophil-to-lymphocyte ratio an inflammatory biomarker, and prognostic marker in heart failure, cardiovascular disease and chronic inflammatory diseases: New insights for a potential predictor of anti-cytokine therapy responsiveness. Microvascular research. Nov 2023;150:104598. doi:10.1016/j.mvr.2023.104598. https://www.sciencedirect.com/science/article/abs/pii/S0026286223001243?via%3Dihub
  266. Firment J, Hulin I. Zahorec index or Neutrophil-to-lymphocyte ratio, valid biomarker of inflammation and immune response to infection, cancer and surgery. Bratislavske lekarske listy. 2024;125(2):75-83. doi:10.4149/bll_2024_012.
  267. Mahmood A, Haider H, Samad S, et al. Association of white blood cell parameters with metabolic syndrome: A systematic review and meta-analysis of 168,000 patients. Medicine. Mar 8 2024;103(10):e37331. doi:10.1097/md.0000000000037331.
  268. Uribe-Querol E, Rosales C. Neutrophils Actively Contribute to Obesity-Associated Inflammation and Pathological Complications. Cells. Jun 10 2022;11(12)doi:10.3390/cells11121883. https://mdpi-res.com/d_attachment/cells/cells-11-01883/article_deploy/cells-11-01883-v2.pdf?version=1655114533
  269. Olsson A, Gustavsen S, Gisselø Lauridsen K, et al. Neutrophil-to-lymphocyte ratio and CRP as biomarkers in multiple sclerosis: A systematic review. Acta Neurol Scand. Jun 2021;143(6):577-586. doi:10.1111/ane.13401. https://onlinelibrary.wiley.com/doi/10.1111/ane.13401
  270. Ye JH, Zhang Y, Naidoo K, Ye S. Neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in psoriasis: a systematic review and meta-analysis. Archives of dermatological research. Feb 8 2024;316(3):85. doi:10.1007/s00403-024-02823-6.
  271. Walther KA, Gröger S, Vogler JAH, Wöstmann B, Meyle J. Inflammation indices in association with periodontitis and cancer. Periodontology 2000. Oct 2024;96(1):281-315. doi:10.1111/prd.12612. https://pmc.ncbi.nlm.nih.gov/articles/PMC11579835/pdf/PRD-96-281.pdf
  272. Khorrampazhouh N, Omranzadeh A, Fazeli B, et al. A Systematic Review and Meta-analysis of Clinical Studies on Ankylosing Spondylitis and Neutrophil to Lymphocyte Ratio. Current rheumatology reviews. 2022;18(2):160-167. doi:10.2174/1573397117666210921114431. https://www.eurekaselect.com/article/118086
  273. Mosca M, Nigro MC, Pagani R, De Giglio A, Di Federico A. Neutrophil-to-Lymphocyte Ratio (NLR) in NSCLC, Gastrointestinal, and Other Solid Tumors: Immunotherapy and Beyond. Biomolecules. Dec 18 2023;13(12)doi:10.3390/biom13121803. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-13-01803/article_deploy/biomolecules-13-01803.pdf?version=1702873327
  274. Chen H, Yang Q, Zheng H, et al. Planetary health diet index and mortality among US cancer survivors: mediating roles of systemic immune-inflammation index and neutrophil-to-lymphocyte ratio. Nutr J. Feb 22 2025;24(1):28. doi:10.1186/s12937-025-01097-6. https://nutritionj.biomedcentral.com/counter/pdf/10.1186/s12937-025-01097-6.pdf
  275. Chen G, Che L, Lai M, et al. Association of neutrophil-lymphocyte ratio with all-cause and cardiovascular mortality in US adults with diabetes and prediabetes: a prospective cohort study. BMC endocrine disorders. May 10 2024;24(1):64. doi:10.1186/s12902-024-01592-7. https://bmcendocrdisord.biomedcentral.com/counter/pdf/10.1186/s12902-024-01592-7.pdf
  276. Zhou E, Wu J, Zhou X, Yin Y. The neutrophil-lymphocyte ratio predicts all-cause and cardiovascular mortality among U.S. adults with rheumatoid arthritis: results from NHANES 1999-2020. Front Immunol. 2023;14:1309835. doi:10.3389/fimmu.2023.1309835. https://pmc.ncbi.nlm.nih.gov/articles/PMC10690944/pdf/fimmu-14-1309835.pdf
  277. Buonacera A, Stancanelli B, Colaci M, Malatino L. Neutrophil to Lymphocyte Ratio: An Emerging Marker of the Relationships between the Immune System and Diseases. International journal of molecular sciences. Mar 26 2022;23(7)doi:10.3390/ijms23073636. https://mdpi-res.com/d_attachment/ijms/ijms-23-03636/article_deploy/ijms-23-03636-v2.pdf?version=1648458498
  278. Gasparyan AY, Ayvazyan L, Mukanova U, Yessirkepov M, Kitas GD. The Platelet-to-Lymphocyte Ratio as an Inflammatory Marker in Rheumatic Diseases. Ann Lab Med. Jul 2019;39(4):345-357. doi:10.3343/alm.2019.39.4.345. https://pmc.ncbi.nlm.nih.gov/articles/PMC6400713/pdf/alm-39-345.pdf
  279. Xu R, Chen L, Yan C, Xu H, Cao G. Elevated Platelet-to-Lymphocyte Ratio as a Predictor of All-Cause and Cardiovascular Mortality in Hypertensive Individuals. Journal of clinical hypertension (Greenwich, Conn). Jan 2025;27(1):e14980. doi:10.1111/jch.14980.
  280. Mangoni AA, Zinellu A. Diagnostic accuracy of the neutrophil-to-lymphocyte ratio and the platelet-to-lymphocyte ratio in rheumatoid arthritis: a systematic review and meta-analysis. Clinical and experimental medicine. Sep 4 2024;24(1):207. doi:10.1007/s10238-024-01478-x.
  281. Qiu Z, Jiang Y, Jiang X, et al. Relationship Between Platelet to Lymphocyte Ratio and Stable Coronary Artery Disease: Meta-Analysis of Observational Studies. Angiology. Nov 2020;71(10):909-915. doi:10.1177/0003319720943810.
  282. Ye Z, Hu T, Wang J, et al. Systemic immune-inflammation index as a potential biomarker of cardiovascular diseases: A systematic review and meta-analysis. Front Cardiovasc Med. 2022;9:933913. doi:10.3389/fcvm.2022.933913. https://pmc.ncbi.nlm.nih.gov/articles/PMC9393310/pdf/fcvm-09-933913.pdf
  283. Hannan MA, Rahman MA, Sohag AAM, et al. Black Cumin (Nigella sativa L.): A Comprehensive Review on Phytochemistry, Health Benefits, Molecular Pharmacology, and Safety. Nutrients. May 24 2021;13(6)doi:10.3390/nu13061784. https://mdpi-res.com/d_attachment/nutrients/nutrients-13-01784/article_deploy/nutrients-13-01784-v2.pdf?version=1621930269
  284. Huseini HF, Mohtashami R, Sadeghzadeh E, Shadmanfar S, Hashem-Dabaghian F, Kianbakht S. Efficacy and safety of oral Nigella sativa oil for symptomatic treatment of knee osteoarthritis: A double-blind, randomized, placebo-controlled clinical trial. Complementary therapies in clinical practice. Nov 2022;49:101666. doi:10.1016/j.ctcp.2022.101666. https://www.sciencedirect.com/science/article/abs/pii/S1744388122001347?via%3Dihub
  285. Tavakoli-Rouzbehani OM, Abbasnezhad M, Kheirouri S, Alizadeh M. Efficacy of nigella sativa oil on endothelial function and atherogenic indices in patients with coronary artery diseases: A randomized, double-blind, placebo-control clinical trial. Phytother Res. Dec 2022;36(12):4516-4526. doi:10.1002/ptr.7568.
  286. Kavyani Z, Musazadeh V, Golpour-Hamedani S, Moridpour AH, Vajdi M, Askari G. The effect of Nigella sativa (black seed) on biomarkers of inflammation and oxidative stress: an updated systematic review and meta-analysis of randomized controlled trials. Inflammopharmacol. Jun 2023;31(3):1149-1165. doi:10.1007/s10787-023-01213-0. https://link.springer.com/article/10.1007/s10787-023-01213-0
  287. Hosseini H, Ghavidel F, Aliyari M, Hashemy SI, Jamialahmadi T, Sahebkar A. Effect of Nigella sativa Intake on Oxidative Stress and Inflammation in Patients with Metabolic Syndrome and Related Disorders: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Current pharmaceutical biotechnology. 2024;25(7):896-907. doi:10.2174/0113892010266109230928000134. https://www.eurekaselect.com/article/134821
  288. Lan X, Xia Y. Alleviating effects of Nigella sativa supplements on biomarkers of inflammation and oxidative stress: Results from an umbrella meta-analysis. Prostaglandins & other lipid mediators. Jan 2025;176:106945. doi:10.1016/j.prostaglandins.2024.106945.
  289. Hallajzadeh J, Milajerdi A, Mobini M, et al. Effects of Nigella sativa on glycemic control, lipid profiles, and biomarkers of inflammatory and oxidative stress: A systematic review and meta-analysis of randomized controlled clinical trials. Phytother Res. Oct 2020;34(10):2586-2608. doi:10.1002/ptr.6708. https://onlinelibrary.wiley.com/doi/10.1002/ptr.6708
  290. Chen L, Zhang L, Hua H, Liu L, Mao Y, Wang R. Interactions between toll-like receptors signaling pathway and gut microbiota in host homeostasis. Immun Inflamm Dis. Jul 2024;12(7):e1356. doi:10.1002/iid3.1356.
  291. Patel D, Evanchuk J, Wang R, Dunbar CL, Munhoz J, Field CJ. Regulation of immune function in healthy adults: one-stop guide on the role of dietary fatty acids, gut microbiota-derived short chain fatty acids, and select micronutrients in combination with physical activity. Applied physiology, nutrition, and metabolism = Physiologie appliquee, nutrition et metabolisme. Aug 1 2023;48(8):554-568. doi:10.1139/apnm-2022-0456. https://cdnsciencepub.com/doi/pdf/10.1139/apnm-2022-0456
  292. Mahdavi-Roshan M, Salari A, Kheirkhah J, Ghorbani Z. The Effects of Probiotics on Inflammation, Endothelial Dysfunction, and Atherosclerosis Progression: A Mechanistic Overview. Heart Lung Circ. May 2022;31(5):e45-e71. doi:10.1016/j.hlc.2021.09.006. https://www.heartlungcirc.org/article/S1443-9506(21)01242-7/abstract
  293. Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct. Jul 18 2022;13(14):7423-7447. doi:10.1039/d2fo00911k. https://pubs.rsc.org/en/content/articlelanding/2022/fo/d2fo00911k
  294. Faghfouri AH, Afrakoti L, Kavyani Z, et al. The role of probiotic supplementation in inflammatory biomarkers in adults: an umbrella meta-analysis of randomized controlled trials. Inflammopharmacol. Oct 2023;31(5):2253-2268. doi:10.1007/s10787-023-01332-8. https://link.springer.com/article/10.1007/s10787-023-01332-8
  295. Ruggiero M, Motti ML, Meccariello R, Mazzeo F. Resveratrol and Physical Activity: A Successful Combination for the Maintenance of Health and Wellbeing? Nutrients. Feb 28 2025;17(5)doi:10.3390/nu17050837. https://mdpi-res.com/d_attachment/nutrients/nutrients-17-00837/article_deploy/nutrients-17-00837.pdf?version=1740716028
  296. Liu Y, You Y, Lu J, Chen X, Yang Z. Recent Advances in Synthesis, Bioactivity, and Pharmacokinetics of Pterostilbene, an Important Analog of Resveratrol. Molecules (Basel, Switzerland). Nov 6 2020;25(21)doi:10.3390/molecules25215166. https://mdpi-res.com/d_attachment/molecules/molecules-25-05166/article_deploy/molecules-25-05166.pdf?version=1604645998
  297. Chen X, Song X, Zhao X, et al. Insights into the Anti-inflammatory and Antiviral Mechanisms of Resveratrol. Mediators Inflamm. 2022;2022:7138756. doi:10.1155/2022/7138756. https://onlinelibrary.wiley.com/doi/pdfdirect/10.1155/2022/7138756?download=true
  298. Prakash V, Bose C, Sunilkumar D, Cherian RM, Thomas SS, Nair BG. Resveratrol as a Promising Nutraceutical: Implications in Gut Microbiota Modulation, Inflammatory Disorders, and Colorectal Cancer. International journal of molecular sciences. Mar 16 2024;25(6)doi:10.3390/ijms25063370. https://mdpi-res.com/d_attachment/ijms/ijms-25-03370/article_deploy/ijms-25-03370.pdf?version=1710570648
  299. Gorabi AM, Aslani S, Imani D, Razi B, Sathyapalan T, Sahebkar A. Effect of resveratrol on C-reactive protein: An updated meta-analysis of randomized controlled trials. Phytother Res. Dec 2021;35(12):6754-6767. doi:10.1002/ptr.7262. https://onlinelibrary.wiley.com/doi/10.1002/ptr.7262
  300. Molani-Gol R, Rafraf M. Effects of resveratrol on the anthropometric indices and inflammatory markers: an umbrella meta-analysis. European journal of nutrition. Jun 2024;63(4):1023-1040. doi:10.1007/s00394-024-03335-9. https://link.springer.com/article/10.1007/s00394-024-03335-9
  301. Teimouri M, Homayouni-Tabrizi M, Rajabian A, Amiri H, Hosseini H. Anti-inflammatory effects of resveratrol in patients with cardiovascular disease: A systematic review and meta-analysis of randomized controlled trials. Complementary therapies in medicine. Nov 2022;70:102863. doi:10.1016/j.ctim.2022.102863. https://www.sciencedirect.com/science/article/pii/S0965229922000656?via%3Dihub
  302. Rafiee S, Mohammadi H, Ghavami A, Sadeghi E, Safari Z, Askari G. Efficacy of resveratrol supplementation in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis of clinical trials. Complementary therapies in clinical practice. Feb 2021;42:101281. doi:10.1016/j.ctcp.2020.101281. https://www.sciencedirect.com/science/article/abs/pii/S1744388120311567?via%3Dihub
  303. Tabrizi R, Tamtaji OR, Lankarani KB, et al. The effects of resveratrol supplementation on biomarkers of inflammation and oxidative stress among patients with metabolic syndrome and related disorders: a systematic review and meta-analysis of randomized controlled trials. Food Funct. Dec 13 2018;9(12):6116-6128. doi:10.1039/c8fo01259h. https://pubs.rsc.org/en/content/articlelanding/2018/fo/c8fo01259h
  304. Zhu P, Jin Y, Sun J, Zhou X. The efficacy of resveratrol supplementation on inflammation and oxidative stress in type-2 diabetes mellitus patients: randomized double-blind placebo meta-analysis. Frontiers in endocrinology. 2024;15:1463027. doi:10.3389/fendo.2024.1463027. https://pmc.ncbi.nlm.nih.gov/articles/PMC11771208/pdf/fendo-15-1463027.pdf
  305. Esmaealzadeh N, Miri MS, Mavaddat H, et al. The regulating effect of curcumin on NF-κB pathway in neurodegenerative diseases: a review of the underlying mechanisms. Inflammopharmacol. Aug 2024;32(4):2125-2151. doi:10.1007/s10787-024-01492-1. https://link.springer.com/article/10.1007/s10787-024-01492-1
  306. Panknin TM, Howe CL, Hauer M, Bucchireddigari B, Rossi AM, Funk JL. Curcumin Supplementation and Human Disease: A Scoping Review of Clinical Trials. International journal of molecular sciences. Feb 24 2023;24(5)doi:10.3390/ijms24054476. https://mdpi-res.com/d_attachment/ijms/ijms-24-04476/article_deploy/ijms-24-04476-v2.pdf?version=1677564959
  307. Dehzad MJ, Ghalandari H, Nouri M, Askarpour M. Antioxidant and anti-inflammatory effects of curcumin/turmeric supplementation in adults: A GRADE-assessed systematic review and dose-response meta-analysis of randomized controlled trials. Cytokine. Apr 2023;164:156144. doi:10.1016/j.cyto.2023.156144. https://www.sciencedirect.com/science/article/abs/pii/S1043466623000224?via%3Dihub
  308. Wilar G, Suhandi C, Fukunaga K, et al. Effects of nanocurcumin supplementation on metabolic syndrome: A systematic review and meta-analysis of randomized controlled trials. Pharmacological research: the official journal of the Italian Pharmacological Society. Mar 2025;213:107641. doi:10.1016/j.phrs.2025.107641. https://www.sciencedirect.com/science/article/pii/S1043661825000660?via%3Dihub
  309. Futuhi F, Naghibzadeh Tahami A, Azmandian J, Saber A. The effects of curcumin-containing supplementations on inflammatory markers and lipid profiles in patients with chronic kidney diseases: a systematic review and meta-analysis of randomized controlled trials. Journal of complementary & integrative medicine. Sep 1 2022;19(3):531-541. doi:10.1515/jcim-2022-0082.
  310. Arabi SM, Bahari H, Hamidipor S, et al. The effects of curcumin-containing supplements on inflammatory biomarkers in hemodialysis patients: A systematic review and meta-analysis. Phytother Res. Dec 2022;36(12):4361-4370. doi:10.1002/ptr.7642. https://onlinelibrary.wiley.com/doi/10.1002/ptr.7642
  311. Ebrahimzadeh A, Abbasi F, Ebrahimzadeh A, Jibril AT, Milajerdi A. Effects of curcumin supplementation on inflammatory biomarkers in patients with Rheumatoid Arthritis and Ulcerative colitis: A systematic review and meta-analysis. Complementary therapies in medicine. Sep 2021;61:102773. doi:10.1016/j.ctim.2021.102773. https://www.sciencedirect.com/science/article/pii/S096522992100114X?via%3Dihub
  312. Sedighi S, Faramarzipalangar Z, Mohammadi E, Aghamohammadi V, Bahnemiri MG, Mohammadi K. The effects of curcumin supplementation on inflammatory markers in systemic lupus erythematosus patients: a randomized placebo-controlled trial. European journal of nutrition. Nov 15 2024;64(1):8. doi:10.1007/s00394-024-03515-7. https://link.springer.com/article/10.1007/s00394-024-03515-7
  313. Dicks LMT. How important are fatty acids in human health and can they be used in treating diseases? Gut Microbes. Jan-Dec 2024;16(1):2420765. doi:10.1080/19490976.2024.2420765. https://www.tandfonline.com/doi/pdf/10.1080/19490976.2024.2420765
  314. Chiang N, Serhan CN. Specialized pro-resolving mediator network: an update on production and actions. Essays in biochemistry. Sep 23 2020;64(3):443-462. doi:10.1042/ebc20200018. https://pmc.ncbi.nlm.nih.gov/articles/PMC7682745/pdf/nihms-1617799.pdf
  315. Banaszak M, Dobrzyńska M, Kawka A, et al. Role of Omega-3 fatty acids eicosapentaenoic (EPA) and docosahexaenoic (DHA) as modulatory and anti-inflammatory agents in noncommunicable diet-related diseases - Reports from the last 10 years. Clinical nutrition ESPEN. Oct 2024;63:240-258. doi:10.1016/j.clnesp.2024.06.053. https://www.clinicalnutritionespen.com/action/showPdf?pii=S2405457724001992
  316. Hong K, Hun M, Wu F, et al. Association between Omega-3 fatty acids and autoimmune disease: Evidence from the umbrella review and Mendelian randomization analysis. Autoimmun Rev. Nov 2024;23(11):103651. doi:10.1016/j.autrev.2024.103651. https://www.sciencedirect.com/science/article/pii/S1568997224001423?via%3Dihub
  317. Calderon Martinez E, Zachariah Saji S, Salazar Ore JV, et al. The effects of omega-3, DHA, EPA, Souvenaid® in Alzheimer's disease: A systematic review and meta-analysis. Neuropsychopharmacol Rep. Sep 2024;44(3):545-556. doi:10.1002/npr2.12455.
  318. Ticinesi A, Meschi T, Lauretani F, et al. Nutrition and Inflammation in Older Individuals: Focus on Vitamin D, n-3 Polyunsaturated Fatty Acids and Whey Proteins. Nutrients. Mar 29 2016;8(4):186. doi:10.3390/nu8040186. https://mdpi-res.com/d_attachment/nutrients/nutrients-08-00186/article_deploy/nutrients-08-00186.pdf?version=1459239559
  319. Wijayabahu AT, Mickle AM, Mai V, et al. Associations between Vitamin D, Omega 6:Omega 3 Ratio, and Biomarkers of Aging in Individuals Living with and without Chronic Pain. Nutrients. Jan 9 2022;14(2)doi:10.3390/nu14020266. https://www.ncbi.nlm.nih.gov/pubmed/35057447
  320. Szymanska P, Rozalski M, Wilczynski M, Golanski J. Systemic immune-inflammation index (SII) and neutrophil to lymphocyte ratio (NLR) are useful markers for assessing effects of anti-inflammatory diet in patients before coronary artery bypass grafting. Roczniki Panstwowego Zakladu Higieny. 2021;72(3):327-335. doi:10.32394/rpzh.2021.0170.
  321. Sut A, Chiżyński K, Różalski M, Golański J. Dietary intake of omega fatty acids and polyphenols and its relationship with the levels of inflammatory markers in men with chronic coronary syndrome after percutaneous coronary intervention. Kardiologia polska. Feb 25 2020;78(2):117-123. doi:10.33963/kp.15078. https://journals.viamedica.pl/polish_heart_journal/article/download/82463/61798
  322. Ahmadi M, Askari VR, Shahri B, Mousavi Noghab SM, Jarahi L, Baradaran Rahimi V. Omega-3 fatty acids effectively mitigate high-sensitivity C-reactive protein (hs-CRP) biomarker of inflammation in acute myocardial infarction patients: a randomized, double-blind, placebo-controlled clinical trial. Naunyn-Schmiedeberg's archives of pharmacology. Jan 2025;398(1):881-890. doi:10.1007/s00210-024-03330-1. https://link.springer.com/article/10.1007/s00210-024-03330-1
  323. Wang Y, Wang Y, Shehzad Q, et al. Does omega-3 PUFAs supplementation improve metabolic syndrome and related cardiovascular diseases? A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. Sep 2024;64(26):9455-9482. doi:10.1080/10408398.2023.2212817.
  324. Ibrahim Mohialdeen Gubari M. Effect of omega-3 fatty acid supplementation on markers of inflammation and endothelial function in patients with chronic heart disease: A systematic review and meta-analysis. Cellular and molecular biology (Noisy-le-Grand, France). Jun 5 2024;70(6):171-177. doi:10.14715/cmb/2024.70.6.26.
  325. Prokopidis K, Therdyothin A, Giannos P, et al. Does omega-3 supplementation improve the inflammatory profile of patients with heart failure? a systematic review and meta-analysis. Heart failure reviews. Nov 2023;28(6):1417-1425. doi:10.1007/s10741-023-10327-0. https://pmc.ncbi.nlm.nih.gov/articles/PMC10575807/pdf/10741_2023_Article_10327.pdf
  326. Serhan CN, Bäck M, Chiurchiù V, et al. Expert consensus report on lipid mediators: Role in resolution of inflammation and muscle preservation. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. May 31 2024;38(10):e23699. doi:10.1096/fj.202400619R. https://faseb.onlinelibrary.wiley.com/doi/pdfdirect/10.1096/fj.202400619R?download=true
  327. Parolini C. The Role of Marine n-3 Polyunsaturated Fatty Acids in Inflammatory-Based Disease: The Case of Rheumatoid Arthritis. Mar Drugs. Dec 27 2023;22(1)doi:10.3390/md22010017. https://mdpi-res.com/d_attachment/marinedrugs/marinedrugs-22-00017/article_deploy/marinedrugs-22-00017.pdf?version=1703657819
  328. Elajami TK, Colas RA, Dalli J, Chiang N, Serhan CN, Welty FK. Specialized proresolving lipid mediators in patients with coronary artery disease and their potential for clot remodeling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. Aug 2016;30(8):2792-801. doi:10.1096/fj.201500155R. https://faseb.onlinelibrary.wiley.com/doi/abs/10.1096/fj.201500155R
  329. Al-Shaer AE, Regan J, Buddenbaum N, et al. Enriched Marine Oil Supplement Increases Specific Plasma Specialized Pro-Resolving Mediators in Adults with Obesity. J Nutr. Jul 6 2022;152(7):1783-1791. doi:10.1093/jn/nxac075. https://pmc.ncbi.nlm.nih.gov/articles/PMC9258560/pdf/nxac075.pdf
  330. Lamon-Fava S, So J, Mischoulon D, et al. Dose- and time-dependent increase in circulating anti-inflammatory and pro-resolving lipid mediators following eicosapentaenoic acid supplementation in patients with major depressive disorder and chronic inflammation. Prostaglandins Leukot Essent Fatty Acids. Jan 2021;164:102219. doi:10.1016/j.plefa.2020.102219. https://pmc.ncbi.nlm.nih.gov/articles/PMC7855824/pdf/nihms-1654304.pdf
  331. Marchand NE, Choi MY, Oakes EG, et al. Over-the-counter fish oil supplementation and pro-resolving and pro-inflammatory lipid mediators in rheumatoid arthritis. Prostaglandins Leukot Essent Fatty Acids. Mar 2023;190:102542. doi:10.1016/j.plefa.2023.102542. https://pmc.ncbi.nlm.nih.gov/articles/PMC10027850/pdf/nihms-1873081.pdf
  332. Barden AE, Mas E, Croft KD, Phillips M, Mori TA. Specialized proresolving lipid mediators in humans with the metabolic syndrome after n-3 fatty acids and aspirin. Am J Clin Nutr. Dec 2015;102(6):1357-64. doi:10.3945/ajcn.115.116384. https://www.sciencedirect.com/science/article/pii/S0002916523272081?via%3Dihub
  333. So J, Yao JH, Magadmi R, Matthan NR, Lamon-Fava S. Sex differences in lipid mediators derived from omega-3 fatty acids in older individuals with low-grade chronic inflammation. Prostaglandins Leukot Essent Fatty Acids. Apr 2024;203:102655. doi:10.1016/j.plefa.2024.102655. https://www.plefa.com/article/S0952-3278(24)00049-8/abstract
  334. Callan N, Hanes D, Bradley R. Early evidence of efficacy for orally administered SPM-enriched marine lipid fraction on quality of life and pain in a sample of adults with chronic pain. Journal of translational medicine. Oct 21 2020;18(1):401. doi:10.1186/s12967-020-02569-5. https://translational-medicine.biomedcentral.com/counter/pdf/10.1186/s12967-020-02569-5.pdf
  335. Ramirez JL, Gasper WJ, Khetani SA, et al. Fish Oil Increases Specialized Pro-resolving Lipid Mediators in PAD (The OMEGA-PAD II Trial). The Journal of surgical research. Jun 2019;238:164-174. doi:10.1016/j.jss.2019.01.038. https://pmc.ncbi.nlm.nih.gov/articles/PMC6462350/pdf/nihms-1008290.pdf
  336. Lamon-Fava S. Associations between omega-3 fatty acid-derived lipid mediators and markers of inflammation in older subjects with low-grade chronic inflammation. Prostaglandins & other lipid mediators. Jan 2025;176:106948. doi:10.1016/j.prostaglandins.2025.106948. https://www.sciencedirect.com/science/article/abs/pii/S1098882325000012?via%3Dihub
  337. Lin Z, Li W. The Roles of Vitamin D and Its Analogs in Inflammatory Diseases. Current topics in medicinal chemistry. 2016;16(11):1242-61. doi:10.2174/1568026615666150915111557.
  338. Zhou Z, Liu J, Zhang H, Prabahar K, Hernández-Wolters B, Yuan Y. The effect of vitamin D2 on lipid profile, anthropometric indices, blood pressure, and inflammatory and glycemic biomarkers in humans: A systematic review and meta-analysis of randomized controlled trials. Prostaglandins & other lipid mediators. Oct 2024;174:106883. doi:10.1016/j.prostaglandins.2024.106883. https://www.sciencedirect.com/science/article/abs/pii/S1098882324000777?via%3Dihub
  339. Moslemi E, Musazadeh V, Kavyani Z, Naghsh N, Shoura SMS, Dehghan P. Efficacy of vitamin D supplementation as an adjunct therapy for improving inflammatory and oxidative stress biomarkers: An umbrella meta-analysis. Pharmacological research : the official journal of the Italian Pharmacological Society. Dec 2022;186:106484. doi:10.1016/j.phrs.2022.106484. https://www.sciencedirect.com/science/article/abs/pii/S1043661822004303?via%3Dihub
  340. MacGirlley R, Phoswa WN, Mokgalaboni K. Modulatory Properties of Vitamin D in Type 2 Diabetic Patients: A Focus on Inflammation and Dyslipidemia. Nutrients. Oct 27 2023;15(21)doi:10.3390/nu15214575. https://mdpi-res.com/d_attachment/nutrients/nutrients-15-04575/article_deploy/nutrients-15-04575-v3.pdf?version=1698721625
  341. Gwenzi T, Zhu A, Schrotz-King P, Schöttker B, Hoffmeister M, Brenner H. Effects of vitamin D supplementation on inflammatory response in patients with cancer and precancerous lesions: Systematic review and meta-analysis of randomized trials. Clin Nutr. Jul 2023;42(7):1142-1150. doi:10.1016/j.clnu.2023.05.009. https://www.clinicalnutritionjournal.com/article/S0261-5614(23)00151-6/abstract
  342. Jiang Q, Prabahar K, Saleh SAK, et al. The Effects of Vitamin D Supplementation on C-Reactive Protein and Systolic and Diastolic Blood Pressure in Postmenopausal Women: A Meta-Analysis and Systematic Review of Randomized Controlled Trials. Journal of the Academy of Nutrition and Dietetics. Mar 2024;124(3):387-396.e5. doi:10.1016/j.jand.2023.10.013. https://www.jandonline.org/article/S2212-2672(23)01630-1/abstract
  343. Al-Saoodi H, Kolahdooz F, Andersen JR, Jalili M. Effect of vitamin D on inflammatory and clinical outcomes in patients with rheumatoid arthritis: a systematic review and dose-response meta-analysis of randomized controlled trials. Nutrition reviews. Apr 12 2024;82(5):600-611. doi:10.1093/nutrit/nuad083. https://academic.oup.com/nutritionreviews/article-abstract/82/5/600/7223423?redirectedFrom=fulltext
  344. Kerlikowsky F, Kruger K, Hahn A, Schuchardt JP. Multimicronutrient and omega-3 fatty acid supplementation reduces low-grade inflammation in older participants: An exploratory study. Nutr Res. Aug 2025;140:46-58. doi:10.1016/j.nutres.2025.06.005. https://www.ncbi.nlm.nih.gov/pubmed/40644954
  345. Mohd Zaffarin AS, Ng SF, Ng MH, Hassan H, Alias E. Pharmacology and Pharmacokinetics of Vitamin E: Nanoformulations to Enhance Bioavailability. International journal of nanomedicine. 2020;15:9961-9974. doi:10.2147/ijn.S276355. https://www.tandfonline.com/doi/pdf/10.2147/IJN.S276355
  346. Asbaghi O, Sadeghian M, Nazarian B, et al. The effect of vitamin E supplementation on selected inflammatory biomarkers in adults: a systematic review and meta-analysis of randomized clinical trials. Sci Rep. Oct 14 2020;10(1):17234. doi:10.1038/s41598-020-73741-6. https://www.nature.com/articles/s41598-020-73741-6.pdf
  347. Moosavian SP, Arab A, Mehrabani S, Moradi S, Nasirian M. The effect of omega-3 and vitamin E on oxidative stress and inflammation: Systematic review and meta-analysis of randomized controlled trials. International journal for vitamin and nutrition research Internationale Zeitschrift fur Vitamin- und Ernahrungsforschung Journal international de vitaminologie et de nutrition. Oct 2020;90(5-6):553-563. doi:10.1024/0300-9831/a000599. https://econtent.hogrefe.com/doi/pdf/10.1024/0300-9831/a000599?download=true
  348. Rafique S, Khan DA, Farhat K, Khan MA, Noor M, Sharif M. Comparative efficacy of tocotrienol and tocopherol (vitamin E) on atherosclerotic cardiovascular diseases in humans. J Pak Med Assoc. Jun 2024;74(6):1124-1129. doi:10.47391/jpma.9227.
  349. Lin YT, Wang LK, Hung KC, et al. Prevalence and Predictors of Insufficient Plasma Vitamin C in a Subtropical Region and Its Associations with Risk Factors of Cardiovascular Diseases: A Retrospective Cross-Sectional Study. Nutrients. Mar 6 2022;14(5)doi:10.3390/nu14051108. https://mdpi-res.com/d_attachment/nutrients/nutrients-14-01108/article_deploy/nutrients-14-01108.pdf?version=1646554551
  350. Crook JM, Horgas AL, Yoon SL, Grundmann O, Johnson-Mallard V. Vitamin C Plasma Levels Associated with Inflammatory Biomarkers, CRP and RDW: Results from the NHANES 2003-2006 Surveys. Nutrients. Mar 16 2022;14(6)doi:10.3390/nu14061254. https://mdpi-res.com/d_attachment/nutrients/nutrients-14-01254/article_deploy/nutrients-14-01254.pdf?version=1647420808
  351. Munday MR, Rodricks R, Fitzpatrick M, Flood VM, Gunton JE. A Pilot Study Examining Vitamin C Levels in Periodontal Patients. Nutrients. Jul 28 2020;12(8)doi:10.3390/nu12082255. https://mdpi-res.com/d_attachment/nutrients/nutrients-12-02255/article_deploy/nutrients-12-02255.pdf?version=1595934275
  352. Isola G, Polizzi A, Muraglie S, Leonardi R, Lo Giudice A. Assessment of Vitamin C and Antioxidant Profiles in Saliva and Serum in Patients with Periodontitis and Ischemic Heart Disease. Nutrients. Dec 4 2019;11(12)doi:10.3390/nu11122956. https://mdpi-res.com/d_attachment/nutrients/nutrients-11-02956/article_deploy/nutrients-11-02956.pdf?version=1575462141
  353. Jiang N, Li N, Huang J, et al. Impact of Vitamin C on Inflammatory Response and Myocardial Injury in Sepsis Patients. Alternative therapies in health and medicine. Oct 2024;30(10):427-431.
  354. Jafarnejad S, Boccardi V, Hosseini B, Taghizadeh M, Hamedifard Z. A Meta-analysis of Randomized Control Trials: The Impact of Vitamin C Supplementation on Serum CRP and Serum hs-CRP Concentrations. Curr Pharm Des. 2018;24(30):3520-3528. doi:10.2174/1381612824666181017101810. https://www.eurekaselect.com/article/93744
  355. Ren J, Liang J, Wang J, et al. Vascular benefits of vitamin C supplementation against fine particulate air pollution in healthy adults: A double-blind randomised crossover trial. Ecotoxicology and environmental safety. Aug 2022;241:113735. doi:10.1016/j.ecoenv.2022.113735. https://www.sciencedirect.com/science/article/pii/S0147651322005759?via%3Dihub
  356. Żychowska M, Grzybkowska A, Zasada M, et al. Effect of six weeks 1000 mg/day vitamin C supplementation and healthy training in elderly women on genes expression associated with the immune response - a randomized controlled trial. Journal of the International Society of Sports Nutrition. Mar 2 2021;18(1):19. doi:10.1186/s12970-021-00416-6. https://www.tandfonline.com/doi/pdf/10.1186/s12970-021-00416-6
  357. Righi NC, Schuch FB, De Nardi AT, et al. Effects of vitamin C on oxidative stress, inflammation, muscle soreness, and strength following acute exercise: meta-analyses of randomized clinical trials. European journal of nutrition. Oct 2020;59(7):2827-2839. doi:10.1007/s00394-020-02215-2. https://link.springer.com/article/10.1007/s00394-020-02215-2
  358. Maywald M, Rink L. Zinc in Human Health and Infectious Diseases. Biomolecules. Nov 24 2022;12(12)doi:10.3390/biom12121748. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-12-01748/article_deploy/biomolecules-12-01748-v2.pdf?version=1669364056
  359. Nira NH, Hoque MR, Khan SR, Ferdausee M, Momo FR, Ara R. Status of Serum Magnesium, Serum Zinc and C-reactive Protein in Hospitalized Patients with Chronic Kidney Disease in Mymensingh Locality. Mymensingh medical journal : MMJ. Apr 2024;33(2):327-333.
  360. Nira NH, Hoque MR, Khan SR, Ferdausee M, Momo FR. Status of C-reactive protein, Serum Albumin and Serum Zinc in Hospital Admitted Patients with Chronic Kidney Disease. Mymensingh medical journal : MMJ. Jan 2024;33(1):1-8.
  361. Jafari A, Noormohammadi Z, Askari M, Daneshzad E. Zinc supplementation and immune factors in adults: a systematic review and meta-analysis of randomized clinical trials. Crit Rev Food Sci Nutr. 2022;62(11):3023-3041. doi:10.1080/10408398.2020.1862048.
  362. Nazari M, Nikbaf-Shandiz M, Pashayee-Khamene F, et al. Zinc Supplementation in Individuals with Prediabetes and type 2 Diabetes: a GRADE-Assessed Systematic Review and Dose-Response Meta-analysis. Biol Trace Elem Res. Jul 2024;202(7):2966-2990. doi:10.1007/s12011-023-03895-7. https://link.springer.com/article/10.1007/s12011-023-03895-7
  363. Ghaedi K, Ghasempour D, Jowshan M, Zheng M, Ghobadi S, Jafari A. Effect of zinc supplementation in the management of type 2 diabetes: A grading of recommendations assessment, development, and evaluation-assessed, dose-response meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. 2024;64(25):9228-9239. doi:10.1080/10408398.2023.2209802.
  364. Nazari M, Ashtary-Larky D, Nikbaf-Shandiz M, et al. Zinc supplementation and cardiovascular disease risk factors: A GRADE-assessed systematic review and dose-response meta-analysis. Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements (GMS). Sep 2023;79:127244. doi:10.1016/j.jtemb.2023.127244. https://www.sciencedirect.com/science/article/abs/pii/S0946672X23001207?via%3Dihub
  365. Bakac ER, Percin E, Gunes-Bayir A, Dadak A. A Narrative Review: The Effect and Importance of Carotenoids on Aging and Aging-Related Diseases. International journal of molecular sciences. Oct 15 2023;24(20)doi:10.3390/ijms242015199. https://mdpi-res.com/d_attachment/ijms/ijms-24-15199/article_deploy/ijms-24-15199.pdf?version=1697351992
  366. Bohn T, Balbuena E, Ulus H, et al. Carotenoids in Health as Studied by Omics-Related Endpoints. Adv Nutr. Nov 2023;14(6):1538-1578. doi:10.1016/j.advnut.2023.09.002. https://pmc.ncbi.nlm.nih.gov/articles/PMC10721521/pdf/main.pdf
  367. Eroglu A, Al'Abri IS, Kopec RE, Crook N, Bohn T. Carotenoids and Their Health Benefits as Derived via Their Interactions with Gut Microbiota. Adv Nutr. Mar 2023;14(2):238-255. doi:10.1016/j.advnut.2022.10.007. https://pmc.ncbi.nlm.nih.gov/articles/PMC10229386/pdf/main.pdf
  368. Sumalla-Cano S, Eguren-García I, Lasarte-García Á, Prola TA, Martínez-Díaz R, Elío I. Carotenoids Intake and Cardiovascular Prevention: A Systematic Review. Nutrients. Nov 12 2024;16(22)doi:10.3390/nu16223859. https://mdpi-res.com/d_attachment/nutrients/nutrients-16-03859/article_deploy/nutrients-16-03859.pdf?version=1731401887
  369. Hajizadeh-Sharafabad F, Zahabi ES, Malekahmadi M, Zarrin R, Alizadeh M. Carotenoids supplementation and inflammation: a systematic review and meta-analysis of randomized clinical trials. Crit Rev Food Sci Nutr. 2022;62(29):8161-8177. doi:10.1080/10408398.2021.1925870.
  370. Cheng HM, Koutsidis G, Lodge JK, Ashor A, Siervo M, Lara J. Tomato and lycopene supplementation and cardiovascular risk factors: A systematic review and meta-analysis. Atherosclerosis. Feb 2017;257:100-108. doi:10.1016/j.atherosclerosis.2017.01.009. https://core.ac.uk/download/pdf/191422137.pdf
  371. Ma B, Lu J, Kang T, Zhu M, Xiong K, Wang J. Astaxanthin supplementation mildly reduced oxidative stress and inflammation biomarkers: a systematic review and meta-analysis of randomized controlled trials. Nutr Res. Mar 2022;99:40-50. doi:10.1016/j.nutres.2021.09.005. https://www.sciencedirect.com/science/article/abs/pii/S0271531721000725?via%3Dihub
  372. Bahari H, Shahraki Jazinaki M, Aghakhani L, et al. Crocin Supplementation on Inflammation and Oxidative Stress: A Systematic Review and Meta-Analysis. Phytother Res. Jan 2025;39(1):465-479. doi:10.1002/ptr.8380.
  373. Aslani MR, Abdollahi N, Matin S, Zakeri A, Ghobadi H. Effect of crocin of Crocus sativus L. on serum inflammatory markers (IL-6 and TNF-α) in chronic obstructive pulmonary disease patients: a randomised, double-blind, placebo-controlled trial. The British journal of nutrition. Aug 14 2023;130(3):446-453. doi:10.1017/s0007114522003397. https://pubmed.ncbi.nlm.nih.gov/36628554/
  374. Rahimi G, Shams S, Aslani MR. Effects of crocin supplementation on inflammatory markers, lipid profiles, insulin and cardioprotective indices in women with PCOS: A randomized, double-blind, placebo-controlled trial. Phytother Res. Jun 2022;36(6):2605-2615. doi:10.1002/ptr.7474.
  375. Behrouz V, Sohrab G, Hedayati M, Sedaghat M. Inflammatory markers response to crocin supplementation in patients with type 2 diabetes mellitus: A randomized controlled trial. Phytother Res. Jul 2021;35(7):4022-4031. doi:10.1002/ptr.7124.
  376. Hsieh CT, Wang J, Chien KL. Association between dietary flavonoid intakes and C-reactive protein levels: a cross-sectional study in Taiwan. Journal of nutritional science. 2021;10:e15. doi:10.1017/jns.2021.8. https://pmc.ncbi.nlm.nih.gov/articles/PMC8057367/pdf/S2048679021000082a.pdf
  377. Cornelis MC, van Dam RM. Habitual Coffee and Tea Consumption and Cardiometabolic Biomarkers in the UK Biobank: The Role of Beverage Types and Genetic Variation. J Nutr. Oct 12 2020;150(10):2772-2788. doi:10.1093/jn/nxaa212. https://pmc.ncbi.nlm.nih.gov/articles/PMC7549305/pdf/nxaa212.pdf
  378. Ilesanmi-Oyelere BL, Kruger MC. Associations between dietary patterns and an array of inflammation biomarkers and plasma lipid profile in postmenopausal women. BMC Womens Health. May 12 2023;23(1):256. doi:10.1186/s12905-023-02417-w. https://bmcwomenshealth.biomedcentral.com/counter/pdf/10.1186/s12905-023-02417-w.pdf
  379. Mozos I, Flangea C, Vlad DC, et al. Effects of Anthocyanins on Vascular Health. Biomolecules. May 30 2021;11(6)doi:10.3390/biom11060811. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-11-00811/article_deploy/biomolecules-11-00811-v2.pdf?version=1622528728
  380. Nocella C, Cavarretta E, Fossati C, et al. Dark Chocolate Intake Positively Modulates Gut Permeability in Elite Football Athletes: A Randomized Controlled Study. Nutrients. Sep 28 2023;15(19)doi:10.3390/nu15194203. https://mdpi-res.com/d_attachment/nutrients/nutrients-15-04203/article_deploy/nutrients-15-04203-v2.pdf?version=1696236179
  381. Dryer-Beers ER, Griffin J, Matthews PM, Frost GS. Higher Dietary Polyphenol Intake Is Associated With Lower Blood Inflammatory Markers. J Nutr. Aug 2024;154(8):2470-2480. doi:10.1016/j.tjnut.2024.05.005.
  382. Panza VP, Brunetta HS, de Oliveira MV, Nunes EA, da Silva EL. Effect of mate tea (Ilex paraguariensis) on the expression of the leukocyte NADPH oxidase subunit p47(phox) and on circulating inflammatory cytokines in healthy men: a pilot study. International journal of food sciences and nutrition. Mar 2019;70(2):212-221. doi:10.1080/09637486.2018.1486393.
  383. Behzadi M, Bideshki MV, Ahmadi-Khorram M, Zarezadeh M, Hatami A. Effect of dark chocolate/ cocoa consumption on oxidative stress and inflammation in adults: A GRADE-assessed systematic review and dose-response meta-analysis of controlled trials. Complementary therapies in medicine. Sep 2024;84:103061. doi:10.1016/j.ctim.2024.103061.
  384. Rondanelli M, Gasparri C, Perna S, et al. A 60-Day Green Tea Extract Supplementation Counteracts the Dysfunction of Adipose Tissue in Overweight Post-Menopausal and Class I Obese Women. Nutrients. Dec 7 2022;14(24)doi:10.3390/nu14245209. https://mdpi-res.com/d_attachment/nutrients/nutrients-14-05209/article_deploy/nutrients-14-05209.pdf?version=1670410487
  385. Asbaghi O, Fouladvand F, Gonzalez MJ, Aghamohammadi V, Choghakhori R, Abbasnezhad A. The effect of green tea on C-reactive protein and biomarkers of oxidative stress in patients with type 2 diabetes mellitus: A systematic review and meta-analysis. Complementary therapies in medicine. Oct 2019;46:210-216. doi:10.1016/j.ctim.2019.08.019.
  386. Serban C, Sahebkar A, Antal D, Ursoniu S, Banach M. Effects of supplementation with green tea catechins on plasma C-reactive protein concentrations: A systematic review and meta-analysis of randomized controlled trials. Nutrition (Burbank, Los Angeles County, Calif). Sep 2015;31(9):1061-71. doi:10.1016/j.nut.2015.02.004. https://www.sciencedirect.com/science/article/abs/pii/S0899900715000726?via%3Dihub
  387. Börner F, Pace S, Jordan PM, et al. Allosteric Activation of 15-Lipoxygenase-1 by Boswellic Acid Induces the Lipid Mediator Class Switch to Promote Resolution of Inflammation. Adv Sci (Weinh). Feb 2023;10(6):e2205604. doi:10.1002/advs.202205604. https://pmc.ncbi.nlm.nih.gov/articles/PMC9951388/pdf/ADVS-10-2205604.pdf
  388. Karlapudi V, Sunkara KB, Konda PR, Sarma KV, Rokkam MP. Efficacy and Safety of Aflapin®, a Novel Boswellia Serrata Extract, in the Treatment of Osteoarthritis of the Knee: A Short-Term 30-Day Randomized, Double-Blind, Placebo-Controlled Clinical Study. J Am Nutr Assoc. Feb 2023;42(2):159-168. doi:10.1080/07315724.2021.2014370.
  389. Barzin Tond S, Balenci L, Khajavirad N, et al. Inflawell(®) improves neutrophil-to-lymphocyte ratio and shortens hospitalization in patients with moderate COVID-19, in a randomized double-blind placebo-controlled clinical trial. Inflammopharmacol. Apr 2022;30(2):465-475. doi:10.1007/s10787-022-00928-w. https://pmc.ncbi.nlm.nih.gov/articles/PMC8867130/pdf/10787_2022_Article_928.pdf
  390. Baram SM, Karima S, Shateri S, et al. Functional improvement and immune-inflammatory cytokines profile of ischaemic stroke patients after treatment with boswellic acids: a randomized, double-blind, placebo-controlled, pilot trial. Inflammopharmacol. Dec 2019;27(6):1101-1112. doi:10.1007/s10787-019-00627-z. https://link.springer.com/article/10.1007/s10787-019-00627-z
  391. Majumdar A, Prasad M, Gandavarapu SR, et al. Efficacy and safety evaluation of Boswellia serrata and Curcuma longa extract combination in the management of chronic lower back pain: A randomised, double-blind, placebo-controlled clinical study. Explore (New York, NY). Jan-Feb 2025;21(1):103099. doi:10.1016/j.explore.2024.103099. https://www.sciencedirect.com/science/article/abs/pii/S1550830724002064?via%3Dihub
  392. Vaidya N, Agarwal R, Dipankar DG, et al. Efficacy and Safety of Boswellia serrata and Apium graveolens L. Extract Against Knee Osteoarthritis and Cartilage Degeneration: A Randomized, Double-blind, Multicenter, Placebo-Controlled Clinical Trial. Pharmaceutical research. Feb 2025;42(2):249-269. doi:10.1007/s11095-025-03818-2. https://pmc.ncbi.nlm.nih.gov/articles/PMC11880083/pdf/11095_2025_Article_3818.pdf
  393. Medina-Vera I, Avila-Nava A, León-López L, Gutiérrez-Solis AL, Talamantes-Gómez JM, Márquez-Mota CC. Plant-based proteins: clinical and technological importance. Food Sci Biotechnol. Aug 2024;33(11):2461-2475. doi:10.1007/s10068-024-01600-5. https://link.springer.com/content/pdf/10.1007/s10068-024-01600-5.pdf
  394. Prokopidis K, Mazidi M, Sankaranarayanan R, Tajik B, McArdle A, Isanejad M. Effects of whey and soy protein supplementation on inflammatory cytokines in older adults: a systematic review and meta-analysis. The British journal of nutrition. Mar 14 2023;129(5):759-770. doi:10.1017/s0007114522001787. https://pmc.ncbi.nlm.nih.gov/articles/PMC9975787/pdf/S0007114522001787a.pdf
  395. Jamshidi S, Mohsenpour MA, Masoumi SJ, et al. Effect of whey protein consumption on IL-6 and TNF-α: A systematic review and meta-analysis of randomized controlled trials. Diabetes Metab Syndr. Jan 2022;16(1):102372. doi:10.1016/j.dsx.2021.102372. https://www.sciencedirect.com/science/article/abs/pii/S1871402121003921?via%3Dihub
  396. Nielsen FH. Dietary Magnesium and Chronic Disease. Adv Chronic Kidney Dis. May 2018;25(3):230-235. doi:10.1053/j.ackd.2017.11.005. https://www.akdh.org/article/S1548-5595(17)30206-9/abstract
  397. Dong Y, Chen L, Gutin B, Huang Y, Dong Y, Zhu H. Magnesium Intake, C-Reactive Protein, and Muscle Mass in Adolescents. Nutrients. Jul 14 2022;14(14)doi:10.3390/nu14142882. https://mdpi-res.com/d_attachment/nutrients/nutrients-14-02882/article_deploy/nutrients-14-02882.pdf?version=1657782500
  398. Kemp JA, Britto IK, Ribeiro M, et al. Serum Magnesium Levels in Patients with Chronic Kidney Disease: Is There a Relationship with Inflammation Status? Biol Trace Elem Res. May 2024;202(5):1983-1990. doi:10.1007/s12011-023-03829-3. https://link.springer.com/article/10.1007/s12011-023-03829-3
  399. Alateeq K, Walsh EI, Ambikairajah A, Cherbuin N. Association between dietary magnesium intake, inflammation, and neurodegeneration. European journal of nutrition. Aug 2024;63(5):1807-1818. doi:10.1007/s00394-024-03383-1. https://pmc.ncbi.nlm.nih.gov/articles/PMC11329609/pdf/394_2024_Article_3383.pdf
  400. Rao ND, Lemaitre RN, Sitlani CM, et al. Dietary magnesium, C-reactive protein and interleukin-6: The Strong Heart Family Study. PLoS One. 2023;18(12):e0296238. doi:10.1371/journal.pone.0296238. https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0296238&type=printable
  401. Mohebi F, Ostadhadi S, Vaziri MS, et al. The effect of magnesium sulfate on gene expression and serum level of inflammatory cytokines in coronary artery disease patients. Inflammopharmacol. Oct 2023;31(5):2421-2430. doi:10.1007/s10787-023-01328-4.
  402. Veronese N, Pizzol D, Smith L, Dominguez LJ, Barbagallo M. Effect of Magnesium Supplementation on Inflammatory Parameters: A Meta-Analysis of Randomized Controlled Trials. Nutrients. Feb 5 2022;14(3)doi:10.3390/nu14030679. https://mdpi-res.com/d_attachment/nutrients/nutrients-14-00679/article_deploy/nutrients-14-00679-v2.pdf?version=1644302571
  403. Talebi S, Miraghajani M, Hosseini R, Mohammadi H. The Effect of Oral Magnesium Supplementation on Inflammatory Biomarkers in Adults: A Comprehensive Systematic Review and Dose-response Meta-analysis of Randomized Clinical Trials. Biol Trace Elem Res. Apr 2022;200(4):1538-1550. doi:10.1007/s12011-021-02783-2. https://link.springer.com/article/10.1007/s12011-021-02783-2
  404. Simental-Mendia LE, Sahebkar A, Rodriguez-Moran M, Zambrano-Galvan G, Guerrero-Romero F. Effect of Magnesium Supplementation on Plasma C-reactive Protein Concentrations: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr Pharm Des. 2017;23(31):4678-4686. doi:10.2174/1381612823666170525153605. https://www.eurekaselect.com/article/83674
  405. Handy DE, Joseph J, Loscalzo J. Selenium, a Micronutrient That Modulates Cardiovascular Health via Redox Enzymology. Nutrients. Sep 17 2021;13(9)doi:10.3390/nu13093238. https://mdpi-res.com/d_attachment/nutrients/nutrients-13-03238/article_deploy/nutrients-13-03238.pdf?version=1631874780
  406. Zhang F, Li X, Wei Y. Selenium and Selenoproteins in Health. Biomolecules. May 8 2023;13(5)doi:10.3390/biom13050799. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-13-00799/article_deploy/biomolecules-13-00799.pdf?version=1683536149
  407. Sahebari M, Rezaieyazdi Z, Khodashahi M. Selenium and Autoimmune Diseases: A Review Article. Current rheumatology reviews. 2019;15(2):123-134. doi:10.2174/1573397114666181016112342. https://www.eurekaselect.com/article/93711
  408. Giacconi R, Chiodi L, Boccoli G, et al. Reduced levels of plasma selenium are associated with increased inflammation and cardiovascular disease in an Italian elderly population. Exp Gerontol. Mar 2021;145:111219. doi:10.1016/j.exger.2020.111219. https://www.sciencedirect.com/science/article/abs/pii/S0531556520305672?via%3Dihub
  409. Ju W, Li X, Li Z, et al. The effect of selenium supplementation on coronary heart disease: A systematic review and meta-analysis of randomized controlled trials. Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements (GMS). Dec 2017;44:8-16. doi:10.1016/j.jtemb.2017.04.009. https://www.sciencedirect.com/science/article/abs/pii/S0946672X17300263?via%3Dihub
  410. Gholami A, Amirkalali B, Baradaran HR, Hariri M. A systematic review and dose-response meta-analysis of the effect of selenium supplementation on serum concentration of C-reactive protein. Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements (GMS). Dec 2023;80:127273. doi:10.1016/j.jtemb.2023.127273. https://www.sciencedirect.com/science/article/abs/pii/S0946672X23001499?via%3Dihub
  411. Gholizadeh M, Khalili A, Roodi PB, et al. Selenium supplementation decreases CRP and IL-6 and increases TNF-alpha: A systematic review and meta-analysis of randomized controlled trials. Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements (GMS). Sep 2023;79:127199. doi:10.1016/j.jtemb.2023.127199. https://www.sciencedirect.com/science/article/abs/pii/S0946672X23000755?via%3Dihub
  412. Sadowski M, Zawieja E, Chmurzynska A. The impact of N-acetylcysteine on lactate, biomarkers of oxidative stress, immune response, and muscle damage: A systematic review and meta-analysis. Journal of cellular and molecular medicine. Dec 2024;28(23):e70198. doi:10.1111/jcmm.70198. https://pmc.ncbi.nlm.nih.gov/articles/PMC11617117/pdf/JCMM-28-e70198.pdf
  413. Wasyanto T, Yasa A, Jalaludinsyah A. Effect of Oral N-Acetylcysteine Supplementation on the Immunity System in Patients with Acute Myocardial Infarction. Acta medica Indonesiana. Oct 2019;51(4):311-317. https://www.actamedindones.org/index.php/ijim/article/download/1000/pdf
  414. Askari M, Faryabi R, Mozaffari H, Darooghegi Mofrad M. The effects of N-Acetylcysteine on serum level of inflammatory biomarkers in adults. Findings from a systematic review and meta-analysis of randomized clinical trials. Cytokine. Nov 2020;135:155239. doi:10.1016/j.cyto.2020.155239.
  415. Faghfouri AH, Zarezadeh M, Tavakoli-Rouzbehani OM, et al. The effects of N-acetylcysteine on inflammatory and oxidative stress biomarkers: A systematic review and meta-analysis of controlled clinical trials. European journal of pharmacology. Oct 5 2020;884:173368. doi:10.1016/j.ejphar.2020.173368.
  416. Hikisz P, Bernasinska-Slomczewska J. Beneficial Properties of Bromelain. Nutrients. Nov 29 2021;13(12)doi:10.3390/nu13124313. https://mdpi-res.com/d_attachment/nutrients/nutrients-13-04313/article_deploy/nutrients-13-04313-v3.pdf?version=1638321549
  417. Kansakar U, Trimarco V, Manzi MV, Cervi E, Mone P, Santulli G. Exploring the Therapeutic Potential of Bromelain: Applications, Benefits, and Mechanisms. Nutrients. Jun 28 2024;16(13)doi:10.3390/nu16132060. https://mdpi-res.com/d_attachment/nutrients/nutrients-16-02060/article_deploy/nutrients-16-02060.pdf?version=1719565439
  418. Alves Nobre T, de Sousa AA, Pereira IC, et al. Bromelain as a natural anti-inflammatory drug: a systematic review. Natural product research. Mar 2025;39(5):1258-1271. doi:10.1080/14786419.2024.2342553.
  419. Klein G, Kullich W, Schnitker J, Schwann H. Efficacy and tolerance of an oral enzyme combination in painful osteoarthritis of the hip. A double-blind, randomised study comparing oral enzymes with non-steroidal anti-inflammatory drugs. Clin Exp Rheumatol. Jan-Feb 2006;24(1):25-30.
  420. Pereira IC, Sátiro Vieira EE, de Oliveira Torres LR, Carneiro da Silva FC, de Castro ESJM, Torres-Leal FL. Bromelain supplementation and inflammatory markers: A systematic review of clinical trials. Clinical nutrition ESPEN. Jun 2023;55:116-127. doi:10.1016/j.clnesp.2023.02.028. https://www.clinicalnutritionespen.com/article/S2405-4577(23)00059-1/abstract
  421. Majdalawieh AF, Ahari SH, Yousef SM, Nasrallah GK. Sesamol: A lignan in sesame seeds with potent anti-inflammatory and immunomodulatory properties. European journal of pharmacology. Dec 5 2023;960:176163. doi:10.1016/j.ejphar.2023.176163.
  422. Majdalawieh AF, Yousef SM, Abu-Yousef IA, Nasrallah GK. Immunomodulatory and anti-inflammatory effects of sesamin: mechanisms of action and future directions. Crit Rev Food Sci Nutr. 2022;62(18):5081-5112. doi:10.1080/10408398.2021.1881438.
  423. Helli B, Shahi MM, Mowla K, Jalali MT, Haghighian HK. A randomized, triple-blind, placebo-controlled clinical trial, evaluating the sesamin supplement effects on proteolytic enzymes, inflammatory markers, and clinical indices in women with rheumatoid arthritis. Phytother Res. Sep 2019;33(9):2421-2428. doi:10.1002/ptr.6433. https://onlinelibrary.wiley.com/doi/10.1002/ptr.6433
  424. Khadem Haghighian M, Alipoor B, Malek Mahdavi A, Eftekhar Sadat B, Asghari Jafarabadi M, Moghaddam A. Effects of sesame seed supplementation on inflammatory factors and oxidative stress biomarkers in patients with knee osteoarthritis. Acta medica Iranica. 2015;53(4):207-13. https://acta.tums.ac.ir/index.php/acta/article/download/4905/4400
  425. Wu JH, Hodgson JM, Puddey IB, Belski R, Burke V, Croft KD. Sesame supplementation does not improve cardiovascular disease risk markers in overweight men and women. Nutr Metab Cardiovasc Dis. Dec 2009;19(11):774-80. doi:10.1016/j.numecd.2009.01.003. https://www.nmcd-journal.com/article/S0939-4753(09)00011-8/abstract
  426. Eliaz I, Raz A. Pleiotropic Effects of Modified Citrus Pectin. Nutrients. Nov 1 2019;11(11)doi:10.3390/nu11112619. https://mdpi-res.com/d_attachment/nutrients/nutrients-11-02619/article_deploy/nutrients-11-02619.pdf?version=1572604783
  427. An L, Chang G, Zhang L, Wang P, Gao W, Li X. Pectin: Health-promoting properties as a natural galectin-3 inhibitor. Glycoconjugate journal. Apr 2024;41(2):93-118. doi:10.1007/s10719-024-10152-z. https://www.ncbi.nlm.nih.gov/pubmed/38630380
  428. Xu GR, Zhang C, Yang HX, et al. Modified citrus pectin ameliorates myocardial fibrosis and inflammation via suppressing galectin-3 and TLR4/MyD88/NF-κB signaling pathway. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. Jun 2020;126:110071. doi:10.1016/j.biopha.2020.110071. https://www.sciencedirect.com/science/article/pii/S0753332220302626?via%3Dihub
  429. Hu H, Zhang S, Pan S. Characterization of Citrus Pectin Oligosaccharides and Their Microbial Metabolites as Modulators of Immunometabolism on Macrophages. J Agric Food Chem. Aug 4 2021;69(30):8403-8414. doi:10.1021/acs.jafc.1c01445. https://pubs.acs.org/doi/10.1021/acs.jafc.1c01445
  430. Martínez-Martínez E, López-Ándres N, Jurado-López R, et al. Galectin-3 Participates in Cardiovascular Remodeling Associated With Obesity. Hypertension. Nov 2015;66(5):961-9. doi:10.1161/hypertensionaha.115.06032. https://www.ahajournals.org/doi/pdf/10.1161/HYPERTENSIONAHA.115.06032?download=true
  431. Ibarrola J, Matilla L, Martínez-Martínez E, et al. Myocardial Injury After Ischemia/Reperfusion Is Attenuated By Pharmacological Galectin-3 Inhibition. Sci Rep. Jul 3 2019;9(1):9607. doi:10.1038/s41598-019-46119-6. https://www.nature.com/articles/s41598-019-46119-6.pdf
  432. Zhang Y, Su D, Wang Y, et al. Locally delivered modified citrus pectin - a galectin-3 inhibitor shows expected anti-inflammatory and unexpected regeneration-promoting effects on repair of articular cartilage defect. Biomaterials. Dec 2022;291:121870. doi:10.1016/j.biomaterials.2022.121870. https://www.sciencedirect.com/science/article/abs/pii/S0142961222005105?via%3Dihub
  433. Lu Y, Zhang M, Zhao P, et al. Modified citrus pectin inhibits galectin-3 function to reduce atherosclerotic lesions in apoE-deficient mice. Molecular medicine reports. Jul 2017;16(1):647-653. doi:10.3892/mmr.2017.6646.
  434. Chen Y, Su D, Zheng J, et al. Intra-articular injection of modified citrus pectin and hyaluronate gel induces synergistic effects in treating osteoarthritis. International journal of biological macromolecules. Sep 2024;276(Pt 1):133840. doi:10.1016/j.ijbiomac.2024.133840. https://www.sciencedirect.com/science/article/abs/pii/S0141813024046452?via%3Dihub
  435. Vijay A, Kelly A, Miller S, et al. Supplementation with Citrus Low-Methoxy Pectin Reduces Levels of Inflammation and Anxiety in Healthy Volunteers: A Pilot Controlled Dietary Intervention Study. Nutrients. Sep 30 2024;16(19)doi:10.3390/nu16193326. https://mdpi-res.com/d_attachment/nutrients/nutrients-16-03326/article_deploy/nutrients-16-03326-v2.pdf?version=1728201210
  436. Chen S, Li Q, Shi H, Li F, Duan Y, Guo Q. New insights into the role of mitochondrial dynamics in oxidative stress-induced diseases. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. Sep 2024;178:117084. doi:10.1016/j.biopha.2024.117084. https://www.sciencedirect.com/science/article/pii/S0753332224009685?via%3Dihub
  437. Nesci S, Spagnoletta A, Oppedisano F. Inflammation, Mitochondria and Natural Compounds Together in the Circle of Trust. International journal of molecular sciences. Mar 24 2023;24(7)doi:10.3390/ijms24076106. https://mdpi-res.com/d_attachment/ijms/ijms-24-06106/article_deploy/ijms-24-06106.pdf?version=1679628859
  438. Qin X, Li H, Zhao H, Fang L, Wang X. Enhancing healthy aging with small molecules: A mitochondrial perspective. Medicinal research reviews. Jul 2024;44(4):1904-1922. doi:10.1002/med.22034. https://onlinelibrary.wiley.com/doi/10.1002/med.22034
  439. Mantle D, Dewsbury M, Hargreaves IP. The Ubiquinone-Ubiquinol Redox Cycle and Its Clinical Consequences: An Overview. International journal of molecular sciences. Jun 20 2024;25(12)doi:10.3390/ijms25126765. https://mdpi-res.com/d_attachment/ijms/ijms-25-06765/article_deploy/ijms-25-06765.pdf?version=1718864950
  440. Jonscher KR, Chowanadisai W, Rucker RB. Pyrroloquinoline-Quinone Is More Than an Antioxidant: A Vitamin-like Accessory Factor Important in Health and Disease Prevention. Biomolecules. Sep 30 2021;11(10)doi:10.3390/biom11101441. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-11-01441/article_deploy/biomolecules-11-01441-v2.pdf?version=1634022163
  441. Liu Z, Sun C, Tao R, et al. Pyrroloquinoline Quinone Decelerates Rheumatoid Arthritis Progression by Inhibiting Inflammatory Responses and Joint Destruction via Modulating NF-κB and MAPK Pathways. Inflammation. Feb 2016;39(1):248-256. doi:10.1007/s10753-015-0245-7.
  442. Han GT, Cai WS, Zhang YB, Zhou SQ, He B, Li HH. Protective Effect of Pyrroloquinoline Quinone on TNF-α-induced Mitochondrial Injury in Chondrocytes. Current medical science. Feb 2021;41(1):100-107. doi:10.1007/s11596-020-2248-3.
  443. Harris CB, Chowanadisai W, Mishchuk DO, Satre MA, Slupsky CM, Rucker RB. Dietary pyrroloquinoline quinone (PQQ) alters indicators of inflammation and mitochondrial-related metabolism in human subjects. J Nutr Biochem. Dec 2013;24(12):2076-84. doi:10.1016/j.jnutbio.2013.07.008.
  444. Nakano M, Murayama Y, Hu L, Ikemoto K, Uetake T, Sakatani K. Effects of Antioxidant Supplements (BioPQQ™) on Cerebral Blood Flow and Oxygen Metabolism in the Prefrontal Cortex. Springer International Publishing; 2016:215-222.
  445. Baltic S, Nedeljkovic D, Todorovic N, et al. The impact of six-week dihydrogen-pyrroloquinoline quinone supplementation on mitochondrial biomarkers, brain metabolism, and cognition in elderly individuals with mild cognitive impairment: a randomized controlled trial. The journal of nutrition, health & aging. Aug 2024;28(8):100287. doi:10.1016/j.jnha.2024.100287. https://www.sciencedirect.com/science/article/pii/S1279770724003749?via%3Dihub
  446. Shiojima Y, Megumi T, Ryohei T, et al. Effect of Dietary Pyrroloquinoline Quinone Disodium Salt on Cognitive Function in Healthy Volunteers: A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study. Journal of the American Nutrition Association. 2022/11/08 2022;41(8):796-809. doi:10.1080/07315724.2021.1962770. https://doi.org/10.1080/07315724.2021.1962770
  447. Hwang PS, Machek SB, Cardaci TD, et al. Effects of Pyrroloquinoline Quinone (PQQ) Supplementation on Aerobic Exercise Performance and Indices of Mitochondrial Biogenesis in Untrained Men. J Am Coll Nutr. Aug 2020;39(6):547-556. doi:10.1080/07315724.2019.1705203.
  448. Mantle D, Turton N, Hargreaves IP. Depletion and Supplementation of Coenzyme Q10 in Secondary Deficiency Disorders. Frontiers in bioscience (Landmark edition). Dec 19 2022;27(12):322. doi:10.31083/j.fbl2712322.
  449. Shimizu M, Miyazaki T, Takagi A, et al. Low coenzyme Q10 levels in patients with acute cardiovascular disease are associated with long-term mortality. Heart Vessels. Mar 2021;36(3):401-407. doi:10.1007/s00380-020-01698-7.
  450. Freire de Carvalho J, Skare T. Coenzyme Q10 supplementation in rheumatic diseases: A systematic review. Clinical nutrition ESPEN. Feb 2024;59:63-69. doi:10.1016/j.clnesp.2023.11.016. https://www.clinicalnutritionespen.com/article/S2405-4577(23)02215-5/abstract
  451. Hou S, Tian Z, Zhao D, et al. Efficacy and Optimal Dose of Coenzyme Q10 Supplementation on Inflammation-Related Biomarkers: A GRADE-Assessed Systematic Review and Updated Meta-Analysis of Randomized Controlled Trials. Mol Nutr Food Res. Jul 2023;67(13):e2200800. doi:10.1002/mnfr.202200800. https://onlinelibrary.wiley.com/doi/10.1002/mnfr.202200800
  452. Dludla PV, Orlando P, Silvestri S, et al. Coenzyme Q(10) Supplementation Improves Adipokine Levels and Alleviates Inflammation and Lipid Peroxidation in Conditions of Metabolic Syndrome: A Meta-Analysis of Randomized Controlled Trials. International journal of molecular sciences. May 4 2020;21(9)doi:10.3390/ijms21093247. https://mdpi-res.com/d_attachment/ijms/ijms-21-03247/article_deploy/ijms-21-03247.pdf?version=1588583157
  453. Jorat MV, Tabrizi R, Kolahdooz F, et al. The effects of coenzyme Q10 supplementation on biomarkers of inflammation and oxidative stress in among coronary artery disease: a systematic review and meta-analysis of randomized controlled trials. Inflammopharmacol. Apr 2019;27(2):233-248. doi:10.1007/s10787-019-00572-x. https://link.springer.com/article/10.1007/s10787-019-00572-x
  454. Zhou M, Zhang Y, Shi L, et al. Activation and modulation of the AGEs-RAGE axis: Implications for inflammatory pathologies and therapeutic interventions - A review. Pharmacological research : the official journal of the Italian Pharmacological Society. Aug 2024;206:107282. doi:10.1016/j.phrs.2024.107282. https://www.sciencedirect.com/science/article/pii/S1043661824002275?via%3Dihub
  455. Rojas A, Lindner C, Schneider I, Gonzalez I, Uribarri J. The RAGE Axis: A Relevant Inflammatory Hub in Human Diseases. Biomolecules. Mar 28 2024;14(4)doi:10.3390/biom14040412. https://mdpi-res.com/d_attachment/biomolecules/biomolecules-14-00412/article_deploy/biomolecules-14-00412-v2.pdf?version=1711974677
  456. Raj V, Ojha S, Howarth FC, Belur PD, Subramanya SB. Therapeutic potential of benfotiamine and its molecular targets. European review for medical and pharmacological sciences. May 2018;22(10):3261-3273. doi:10.26355/eurrev_201805_15089.
  457. Allowitz KV, Yoo JJ, Taylor JR, Baloch OA, Harames K, Ramana KV. Therapeutic potential of vitamin B(1) derivative benfotiamine from diabetes to COVID-19. Future medicinal chemistry. Jun 2022;14(11):809-826. doi:10.4155/fmc-2022-0040.
  458. Nwadiugwu MC. Inflammatory Activities in Type 2 Diabetes Patients With Co-morbid Angiopathies and Exploring Beneficial Interventions: A Systematic Review. Frontiers in public health. 2020;8:600427. doi:10.3389/fpubh.2020.600427.
  459. Shoeb M, Ramana KV. Anti-inflammatory effects of benfotiamine are mediated through the regulation of the arachidonic acid pathway in macrophages. Free radical biology & medicine. Jan 1 2012;52(1):182-90. doi:10.1016/j.freeradbiomed.2011.10.444. https://www.ncbi.nlm.nih.gov/pubmed/22067901
  460. Gibson GE, Luchsinger JA, Cirio R, et al. Benfotiamine and Cognitive Decline in Alzheimer's Disease: Results of a Randomized Placebo-Controlled Phase IIa Clinical Trial. J Alzheimers Dis. 2020;78(3):989-1010. doi:10.3233/jad-200896. https://pmc.ncbi.nlm.nih.gov/articles/PMC7880246/pdf/nihms-1666597.pdf
  461. Garg S, Syngle A, Vohra K. Efficacy and tolerability of advanced glycation end-products inhibitor in osteoarthritis: a randomized, double-blind, placebo-controlled study. Clin J Pain. Aug 2013;29(8):717-24. doi:10.1097/AJP.0b013e318272ebec.
  462. Syngle A, Vohra K, Garg N, Kaur L, Chand P. Advanced glycation end-products inhibition improves endothelial dysfunction in rheumatoid arthritis. International journal of rheumatic diseases. Feb 2012;15(1):45-55. doi:10.1111/j.1756-185X.2011.01679.x.
  463. Fraser DA, Diep LM, Hovden IA, et al. The effects of long-term oral benfotiamine supplementation on peripheral nerve function and inflammatory markers in patients with type 1 diabetes: a 24-month, double-blind, randomized, placebo-controlled trial. Diabetes Care. May 2012;35(5):1095-7. doi:10.2337/dc11-1895.
  464. Alkhalaf A, Kleefstra N, Groenier KH, et al. Effect of benfotiamine on advanced glycation endproducts and markers of endothelial dysfunction and inflammation in diabetic nephropathy. PLoS One. 2012;7(7):e40427. doi:10.1371/journal.pone.0040427.
  465. Aldini G, de Courten B, Regazzoni L, et al. Understanding the antioxidant and carbonyl sequestering activity of carnosine: direct and indirect mechanisms. Free Radic Res. Apr 2021;55(4):321-330. doi:10.1080/10715762.2020.1856830.
  466. Ghodsi R, Kheirouri S. Carnosine and advanced glycation end products: a systematic review. Amino Acids. Sep 2018;50(9):1177-1186. doi:10.1007/s00726-018-2592-9. https://link.springer.com/article/10.1007/s00726-018-2592-9
  467. Saadati S, Kabthymer RH, Aldini G, Mousa A, Feehan J, de Courten B. Effects of carnosine and histidine-containing dipeptides on biomarkers of inflammation and oxidative stress: a systematic review and meta-analysis. Nutrition reviews. Dec 1 2024;82(12):1696-1709. doi:10.1093/nutrit/nuad150. https://pmc.ncbi.nlm.nih.gov/articles/PMC11551452/pdf/nuad150.pdf
  468. Houjeghani S, Kheirouri S, Faraji E, Jafarabadi MA. l-Carnosine supplementation attenuated fasting glucose, triglycerides, advanced glycation end products, and tumor necrosis factor-α levels in patients with type 2 diabetes: a double-blind placebo-controlled randomized clinical trial. Nutr Res. Jan 2018;49:96-106. doi:10.1016/j.nutres.2017.11.003. https://www.sciencedirect.com/science/article/abs/pii/S0271531717303652?via%3Dihub
  469. Saadati S, de Courten M, Deceneux C, et al. Carnosine Supplementation Has No Effect on Inflammatory Markers in Adults with Prediabetes and Type 2 Diabetes: A Randomised Controlled Trial. Nutrients. Nov 15 2024;16(22)doi:10.3390/nu16223900. https://mdpi-res.com/d_attachment/nutrients/nutrients-16-03900/article_deploy/nutrients-16-03900.pdf?version=1731664788
  470. Menini S, Iacobini C, Fantauzzi CB, Pugliese G. L-carnosine and its Derivatives as New Therapeutic Agents for the Prevention and Treatment of Vascular Complications of Diabetes. Curr Med Chem. 2020;27(11):1744-1763. doi:10.2174/0929867326666190711102718. https://www.eurekaselect.com/article/99558
  471. Palmieri B, Vadalà M. The role of Carcinine treatment on glico-lipidic imbalance of patients with altered blood glucose pattern. Clin Ter. Mar-Apr 2023;174(2):195-202. doi:10.7417/ct.2023.2519.
  472. Narda M, Peno-Mazzarino L, Krutmann J, Trullas C, Granger C. Novel Facial Cream Containing Carnosine Inhibits Formation of Advanced Glycation End-Products in Human Skin. Skin Pharmacol Physiol. 2018;31(6):324-331. doi:10.1159/000492276. https://karger.com/spp/article-pdf/31/6/324/3547324/000492276.pdf
  473. Song B, Hao M, Zhang S, et al. Comprehensive review of Hesperetin: Advancements in pharmacokinetics, pharmacological effects, and novel formulations. Fitoterapia. Dec 2024;179:106206. doi:10.1016/j.fitote.2024.106206.
  474. Mbara KC, Devnarain N, Owira PMO. Potential Role of Polyphenolic Flavonoids as Senotherapeutic Agents in Degenerative Diseases and Geroprotection. Pharmaceut Med. Dec 2022;36(6):331-352. doi:10.1007/s40290-022-00444-w. https://pmc.ncbi.nlm.nih.gov/articles/PMC9470070/pdf/40290_2022_Article_444.pdf
  475. Xiong H, Wang J, Ran Q, et al. Hesperidin: A Therapeutic Agent For Obesity. Drug design, development and therapy. 2019;13:3855-3866. doi:10.2147/dddt.S227499. https://www.tandfonline.com/doi/pdf/10.2147/DDDT.S227499
  476. Yang H, Wang Y, Xu S, et al. Hesperetin, a Promising Treatment Option for Diabetes and Related Complications: A Literature Review. J Agric Food Chem. Jul 20 2022;70(28):8582-8592. doi:10.1021/acs.jafc.2c03257. https://pubs.acs.org/doi/10.1021/acs.jafc.2c03257
  477. Rehman K, Munawar SM, Akash MSH, et al. Hesperidin improves insulin resistance via down-regulation of inflammatory responses: Biochemical analysis and in silico validation. PLoS One. 2020;15(1):e0227637. doi:10.1371/journal.pone.0227637. https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0227637&type=printable
  478. Yari Z, Movahedian M, Imani H, Alavian SM, Hedayati M, Hekmatdoost A. The effect of hesperidin supplementation on metabolic profiles in patients with metabolic syndrome: a randomized, double-blind, placebo-controlled clinical trial. European journal of nutrition. Sep 2020;59(6):2569-2577. doi:10.1007/s00394-019-02105-2. https://link.springer.com/article/10.1007/s00394-019-02105-2
  479. Lorzadeh E, Ramezani-Jolfaie N, Mohammadi M, Khoshbakht Y, Salehi-Abargouei A. The effect of hesperidin supplementation on inflammatory markers in human adults: A systematic review and meta-analysis of randomized controlled clinical trials. Chem Biol Interact. Jul 1 2019;307:8-15. doi:10.1016/j.cbi.2019.04.016. https://www.sciencedirect.com/science/article/abs/pii/S0009279719301449?via%3Dihub
  480. Miles EA, Calder PC. Effects of Citrus Fruit Juices and Their Bioactive Components on Inflammation and Immunity: A Narrative Review. Front Immunol. 2021;12:712608. doi:10.3389/fimmu.2021.712608. https://pmc.ncbi.nlm.nih.gov/articles/PMC8264544/pdf/fimmu-12-712608.pdf
  481. Tadros FJ, Andrade JM. Impact of hesperidin in 100% orange juice on chronic disease biomarkers: A narrative systematic review and gap analysis. Crit Rev Food Sci Nutr. 2022;62(30):8335-8354. doi:10.1080/10408398.2021.1927976. https://www.tandfonline.com/doi/pdf/10.1080/10408398.2021.1927976
  482. Irún P, Carrera-Lasfuentes P, Sánchez-Luengo M, et al. Pharmacokinetics and Changes in Lipid Mediator Profiling after Consumption of Specialized Pro-Resolving Lipid-Mediator-Enriched Marine Oil in Healthy Subjects. Int J Mol Sci. Nov 9 2023;24(22)doi:10.3390/ijms242216143. https://pubmed.ncbi.nlm.nih.gov/38003333/

  483. Kolobarić N, Drenjančević I, Matić A, Šušnjara P, Mihaljević Z, Mihalj M. Dietary Intake of n-3 PUFA-Enriched Hen Eggs Changes Inflammatory Markers' Concentration and Treg/Th17 Cells Distribution in Blood of Young Healthy Adults-A Randomised Study. Nutrients. May 28 2021;13(6)doi:10.3390/nu13061851. https://pubmed.ncbi.nlm.nih.gov/34071714/