Saturday, April 24, 2021

Inflammatory Bowel Disease—Knowing the Basics

Inflammatory bowel disease (IBD) is a complicated chronic inflammatory disease that involves various genetic and environmental driving factors.[1]

Video 1.  The Best Diet for Crohn’s Disease Treatment (YouTube link)

Types of IBD?


Inflammatory bowel disease (IBD) is an umbrella term used to describe disorders that involve chronic inflammation of your digestive tract. Types of IBD include:
  • Ulcerative colitis
    • Which involves inflammation and sores (ulcers) along the superficial lining of your large intestine (colon) and rectum
  • Crohn's disease
    • Which is characterized by inflammation of the lining of your digestive tract, which often can involve the deeper layers of the digestive tract
Both ulcerative colitis and Crohn's disease usually are characterized by diarrhea, rectal bleeding, abdominal pain, fatigue and weight loss.

5 Proven Health Benefits of Walnuts

Image via: AHealthBlog

Diets and IBD


IBD can be debilitating and sometimes leads to life-threatening complications.  Although its pathogenesis is poorly understood, an increasing number of studies have highlighted that dietary intake plays a key role in disease occurrence due to its underlying effects on gut microbiota, barrier function, and mucosal immunity.[2] For instance, 
  • Westernized diet (WD)
    • In [7], it shows the implications of the WD in the onset and progression of IBD
    • WD is very different from the traditional diet of previous generations when the prevalence of IBD was considerably lower
      • The most radical change has been the switch from a plant-based to an animal-sourced diet
      • Another important change brought about by the Western diet is an overall higher calorie intake, especially from sugar, refined carbohydrates, animal proteins and ultra-processed foods.
  • High-salt diet
  • High-fat diet
    • Can contribute toward IBD progression by activating proinflammatory signaling and disrupting barrier systems[4]
  • Ketogenic diet (KD)
    • KD is characterized by high-fat and low-carbohydrate, is a dietary therapy that was initially used to treat drug-resistant epilepsy.[5]
    • In a study, it has shown that KD significantly reduced inflammatory responses, protected intestinal barrier function, and reduced ILC3 production and the expression of related inflammatory cytokines (IL-17α, IL-18, IL-22, Ccl4), whereas the opposite effects were observed for the low-carbohydrate diet (LCD).[6]

More Readings


References

  1. Digby-Bell, J. L., Atreya, R., Monteleone, G. & Powell, N. Interrogating host immunity to predict treatment response in inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 17, 9–20 (2020).
  2. Keshteli, A. H., Madsen, K. L. & Dieleman, L. A. Diet in the pathogenesis and management of ulcerative colitis; a review of randomized controlled dietary interventions. Nutrients 11, 1498 (2019).
  3. Miranda, P. M. et al. High salt diet exacerbates colitis in mice by decreasing Lactobacillus levels and butyrate production. Microbiome 6, 57 (2018).
  4. Rohr, M. W., Narasimhulu, C. A., Rudeski-Rohr, T. A. & Parthasarathy, S. Negative effects of a high-fat diet on intestinal permeability: a review. Adv. Nutr. 11, 77–91 (2020).
  5. Ulamek-Koziol, M., Czuczwar, S. J., Januszewski, S. & Pluta, R. Ketogenic diet and epilepsy. Nutrients 11, 2510 (2019).
  6. Ketogenic diet alleviates colitis by reduction of colonic group 3 innate lymphoid cells through altering gut microbiome
  7. Implications of the Westernized Diet in the Onset and Progression of IBD
  8. Diet and nutrition in the management of inflammatory bowel disease
  9. Walnuts may help protect against ulcerative colitis

Sunday, April 18, 2021

Vitiligo—Knowing the Basics

Figure 1.  Blaschko's lines (source: ScienceDirect.com)

Vitiligo is a disorder that causes the presence of pale patchy areas of depigmented skin on the face, hands and wrists; these patches are initially small, but often grow and change in shape.[1]  Genetic and environmental factors are important in the pathogenesis of vitiligo. There are many therapies for vitiligo, the most used are local steroids and ultraviolet light.


Segmental Vitiligo (SV) vs Nonsegmental Vitiligo 


Vitiligo results from autoimmune destruction of melanocytes,[21]  with elevated prevalence of other autoimmune diseases in patients with vitiligo and their relatives.[22] Vitiligo pathogenesis involves innate immune responses triggered by damage-associated molecular patterns (DAMPs) released by damaged melanocytes, causing downstream adaptive immune activation that drives melanocyte destruction by autoreactive cytotoxic T lymphocytes.[23] 

Vitiligo have the following characteristics:
  • Melanocytes in vitiligo patients are more susceptible to oxidative damage than melanocytes from unaffected individuals 
    • Which is due to an inherited inability to manage stressors from normal cellular processes, or exposure to environmental chemicals
    • Chemicals such as monobenzone and other phenols commonly found in commercial products (including rubber, leather products, cosmetic dyes, etc.) are well known to both induce and exacerbate vitiligo 
    • In addition to the very specific effect of phenols in melanocytes through interaction with tyrosinase, cellular stress results from oxygen and/or nutrient imbalances, chemical or physical agents, infection, inflammation, and misfolded proteins.
  • Innate immunity plays an important role too (melanocyte stress → innate immunity → adaptive immunity)[31]
    • While cellular stress in melanocytes provides a reasonable explanation for how vitiligo is induced, it cannot completely account for the disease, since stressed melanocytes remain viable. 
  • Tyrosinase is an enzyme that catalyzes melanin biosynthesis, and it is a major autoantigen that is implicated in generalized vitiligo.  
  • Critical cytokines in pathogenesis
    • Cytokine interleukin-1beta (IL-1β) is expressed at high levels in patients with vitiligo
    • IFN-γ is identified as a critical cytokine in pathogenesis[24,25]
  • Tregs are reported to be dysregulated in patients with vitiligo[26-29]
    • Disease is exacerbated in their absence, suggesting a possible role for T regulatory (Treg) suppression
    • Although there is no clear consensus on specific abnormalities (i.e. decreased numbers, skin homing defects, or functional defects)[24]
  • The deficiency of TSLP gene expression produces the dominance of the Th1 immune response, which is involved in vitiligo development.
    • TSLP gene encodes thymic stromal lymphopoietin that induces naive CD4+ T cells to produce cytokines that induce a T helper (Th)2 response [IL-4, IL-5, IL-13, tumor necrosis factor (TNF)-α] and inhibit production of Th1cytokines [IL-10, interferon (IFN)-γ].[7]

The disorder is classified in two categories:
  • Nonsegmental vitiligo
    • Bilateral and symmetrical distribution of depigmented macules.
    • Genetic studies concern mainly patients with NSV.  
      • Up to now, about 36 NSV susceptibility loci have been identified: 90% encode immunoregulatory proteins; 10% encode melanocyte proteins
  • Segmental vitiligo
Figure 2.  Interferon-mediated antiviral responses and diseases (Source: [18])


CXCL10


It has been recently reported that the serum and/or the tissue CXCL10 expressions are increased in organ specific autoimmune diseases, which include vitiligo. Determination of high level of CXCL10 in peripheral liquids is a marker of host immune response, especially of a Th1 orientated immune response.
A critical role for CXCL10 in both the progression and maintenance of vitiligo was identified and thereby support inhibiting CXCL10 (e.g. by administering CXCL10 neutralizing antibodies) as a targeted treatment strategy.[8-10]
More importantly, a recent study reports that both human vitiligo and a mouse model of vitiligo reflect an IFN-γ-specific Th1 immune response in the skin that involves the IFN-γ-dependent chemokines CXCL9, 10, and 11:[8]
Melanocyte-specific CD8+ T cells expressed CXCR3 (the common receptor for CXCL9, CXCL10 and CXCL11, unlike healthy controls.
In a mice study, autoreactive T cells express CXCR3, similarly to what observed in humans with disease.
In inflamed tissues recruited Th1 lymphocytes are responsible for enhanced IFN-γ and TNF-α production, which in turn stimulate CXCL10 secretion from a variety of cells, therefore creating an amplification feedback loop.[11]
CXCL10 is secreted by several cell types in response to IFN-γ. These cell types include monocytes, endothelial cells and fibroblasts.[12] CXCL10 has been attributed to several roles, such as chemoattraction for monocytes/macrophages, T cells, NK cells, and dendritic cells, promotion of T cell adhesion to endothelial cells, antitumor activity, and inhibition of bone marrow colony formation and angiogenesis.[13-14]

Simvastatin—a Potential Treatment Option


On the base of the results obtained from an important study,[8] it was evaluated the effect of inhibition of IFN-γ in vitiligo. STAT1 activation is required for IFN-γ signaling and recent studies revealed that simvastatin, an FDA-approved cholesterol-lowering medication, inhibited STAT1 activation in vitro.[15] Therefore, it was hypothesized that simvastatin may be an effective treatment for vitiligo.

In fact, simvastatin both prevented and reversed depigmentation in the mouse model of vitiligo, and reduced the number of infiltrating autoreactive CD8+ T cells in the skin. Treatment of melanocyte-specific, CD8+ T cells in vitro decreased proliferation and IFN-γ production, suggesting additional effects of simvastatin directly on T cells. Based on these data, it has been suggested that simvastatin may be a safe, targeted treatment option for patients with vitiligo.[15] However, the clinical trial finished in 2016 and the result was not ideal as the author explained in his review article:
Vitiligo Simvastatin Clinical Trial Results

References

  1. Taïeb A, Picardo M. Clinical practice. Vitiligo. N Engl J Med 2009;360:160-9.
  2. The role of the Th1 chemokine CXCL10 in vitiligo
  3. World Vitiligo Day: 10 Effective Home Remedies To Treat Vitiligo
  4. Are You Th1 or Th2 Dominant?
  5. Blaschko lines and other patterns of cutaneous mosaicism
  6. Jin Y, Birlea SA, Fain PR, et al. Variant of TYR and autoimmunity susceptibility loci in generalized vitiligo. N Engl J Med 2010;362:1686-97.
  7. Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol 2002;3:673-80.
  8. Rashighi M, Agarwal P, Richmond JM, et al. CXCL10 is critical for the progression and maintenance of depigmentation in a mouse model of vitiligo. Sci Transl Med 2014;6:223ra23.
  9. Zhang S, Liu S, Yu N, et al. RNA released from necrotic keratinocytes upregulates intercellular adhesion molecule-1 expression in melanocytes. Arch Dermatol Res 2011;303:771-6.
  10. Harris JE, Harris TH, Weninger W, et al. A mouse model of vitiligo with focused epidermal depigmentation requires IFN-γ for autoreactive CD8+ T-cell accumulation in the skin. J Invest Dermatol 2012;132:1869-76.
  11. Antonelli A, Ferrari SM, Giuggioli D, et al. Chemokine (C-X-C motif) ligand (CXCL)10 in autoimmune diseases. Autoimmun Rev 2014;13:272-80.
  12. Luster AD, Unkeless JC, Ravetch JV (1985). "Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins". Nature. 315 (6021): 672–6.
  13. Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD (April 2002). "IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking". Journal of Immunology. 168 (7): 3195–204.
  14. Angiolillo AL, Sgadari C, Taub DD, Liao F, Farber JM, Maheshwari S, et al. (July 1995). "Human interferon-inducible protein 10 is a potent inhibitor of angiogenesis in vivo". The Journal of Experimental Medicine. 182 (1): 155–62.
  15. Agarwal P, Rashighi M, Essien KI, et al. Simvastatin Prevents and Reverses Depigmentation in a Mouse Model of Vitiligo. J Invest Dermatol 2015;135:1080-8.
  16. N-Acetylglucosamine Inhibits T-helper 1 (Th1)/T-helper 17 (Th17) Cell Responses and Treats Experimental Autoimmune Encephalomyelitis
  17. Association of a Marker of N-Acetylglucosamine With Progressive Multiple Sclerosis and Neurodegeneration
  18. Regulation of RIG-I-like receptor-mediated signaling: interaction between host and viral factors
  19. Jin, Y. et al. Genome-wide association analyses identify 13 new susceptibility loci for generalized vitiligo. Nat. Genet. 44, 676–680 (2012).
  20. Jin, Y., Andersen, G. H. L., Santorico, S. A. & Spritz, R. A. Multiple Functional Variants of IFIH1, a Gene Involved in Triggering Innate Immune Responses, Protect against Vitiligo. J. Invest. Dermatol 137, 522–524 (2017).
  21. Picardo M. Taïeb A. Vitiligo. Springer, Heidelberg & New York2010
  22. Alkhateeb A., Fain P.R., Thody A., Bennett D.C., Spritz R.A. Epidemiology of vitiligo and associated autoimmune diseases in Caucasian probands and their families.Pigment Cell Res. 2003; 16: 208-214.
  23. Richmond J.M., Frisoli M.L., Harris J.E. Innate immune mechanisms in vitiligo: danger from within.Curr Opin Immunol. 2013; 25: 676-682.
  24. Gregg RK, Nichols L, Chen Y, Lu B, Engelhard VH. Mechanisms of spatial and temporal development of autoimmune vitiligo in tyrosinase-specific TCR transgenic mice. J Immunol. 2010;184:1909–1917.
  25. Harris JE, Harris TH, Weninger W, Wherry EJ, Hunter CA, Turka LA. A mouse model of vitiligo with focused epidermal depigmentation requires IFN-gamma for autoreactive CD8(+) T-cell accumulation in the skin. J Invest Dermatol. 2012;132:1869–1876.
  26. Klarquist J, Denman CJ, Hernandez C, Wainwright DA, Strickland FM, Overbeck A, Mehrotra S, Nishimura MI, Le Poole IC. Reduced skin homing by functional Treg in vitiligo. Pigment Cell Melanoma Res. 2010;23:276–286.
  27. Tu CX, Jin WW, Lin M, Wang ZH, Man MQ. Levels of TGF-beta(1) in serum and culture supernatants of CD4(+)CD25 (+) T cells from patients with non-segmental vitiligo. Arch Dermatol Res. 2011;303:685–689. 
  28. Lili Y, Yi W, Ji Y, Yue S, Weimin S, Ming L. Global activation of CD8+ cytotoxic T lymphocytes correlates with an impairment in regulatory T cells in patients with generalized vitiligo. PLoS One. 2012;7:e37513. 
  29. Zhou L, Li K, Shi YL, Hamzavi I, Gao TW, Henderson M, Huggins RH, Agbai O, Mahmoud B, Mi X, et al. Systemic analyses of immunophenotypes of peripheral T cells in non-segmental vitiligo: implication of defective natural killer T cells. Pigment Cell Melanoma Res. 2012;25:602–611.
  30. N-ACETYL GLUCOSAMINE
  31. Shi F, Kong BW, Song JJ, Lee JY, Dienglewicz RL, Erf GF. Understanding mechanisms of vitiligo development in Smyth line of chickens by transcriptomic microarray analysis of evolving autoimmune lesions. BMC Immunol. 2012;13:18.
  32. Phenol
    • Phenol derivatives have been used in the preparation of cosmetics including sunscreens,hair colorings, skin lightening preparations, as well as in skin toners/exfoliators. However, due to safety concerns, phenol is banned from use in cosmetic products in the European Union and Canada.
  33. Damage-Associated Molecular Patterns in Inflammatory Diseases
  34. Oral Gliadin-Protected Superoxide Dismutase in Addition to Phototherapy for Treating Non-segmental Vitiligo: A 24-Week Prospective Randomized Placebo-Controlled Study

Sunday, April 11, 2021

Tregs and Autoimmune Diseases

Figure 1. Schematic representation of the host immune response against microbial pathogens (Source: 18). 

Allergy and hypersensitivity reactions in general are traditionally thought of as misguided or excessive reactions by the immune system, possibly due to broken or underdeveloped mechanisms of peripheral tolerance.
Hypersensitivity reactions are immune mediated responses that occur in pre-sensitized hosts.[41] 
These reactions can be classified into four categories: 
  1. Type I, anaphylactic, mediated by IgE; 
  2. Type II, cytotoxic, mediated by antibodies recognizing self-antigens; 
  3. Type III, immune complex, caused by deposition of antigen-antibody complexes in tissues, leading to the tissue-damaging effects of complement and leukocytes (e.g. Arthus type reaction); and 
  4. Type IV, delayed-type hypersensitivity, which is not mediated by antibodies but by T cells.
Naive CD4 T helper cells differentiate into different CD4 T effector lineages after antigen encounter, including Th1, Th2, Th17, Tfh and Treg cells (see Figure 1).

It's important to note that the numeral allocation of hypersensitivity "types" does not correlate (and is completely unrelated) to the "response" in the Th model (i.e., Th1, Th2).  

CD4 T CellsKey
Transcription
Factor
Signature 
Cytokines
Effects of Overactivation
against Antigen
Th1T-bet
STAT4
Triggered by the polarizing cytokine IL-12 and their effector cytokines are IFN-γ and IL-2Type IV
hypersensitivity which include Type 1 diabetes
Th2GATA3
STAT6
Triggered by the polarizing cytokines IL-4 and IL-2, and their effector cytokines are IL-4, IL-5, IL-9, IL-10, IL-13 and IL-25Type I hypersensitivity which is an allergic reaction mediated by IgE

Are associated with autoimmune disease such as:
• allergic rhinitis
• atopic dermatitis
• asthma

Th17Stat3
RORγ
RORα
Triggered by the polarizing cytokines TGF-β, IL-6, IL-21 and IL-23[31]Type 3 immune complex and complement-mediated hypersensitivity.

Are associated with autoimmune disease such as:
• multiple sclerosis[45]
• rheumatoid arthritis
• psoriasis
TfhBcl-6Triggered by CD278 or ICOS and their effector cytokines are IL-21 and IL-4Are associated with antibody-mediated autoimmune diseases, which include SLE and Sjögren syndrome
       Table 1.  Helper T Cells and associated hypersensitivity

Key Transcription Factor

Transcription factors are proteins that help turn specific genes "on" or "off" by binding to nearby DNA. Transcription factors that are activators boost a gene's transcription. Groups of transcription factor binding sites called enhancers and silencers can turn a gene on/off in specific parts of the body.

Lineage-Specific Transcription Factors

When the environment is rich in interleukin (IL)-12 and/or interferon (IFN)-γ, naive CD4 T cells differentiate into IFN-γ-producing Th1 cells, driven by the transcription factor T-bet.

Similarly, IL-4 induces IL-4-producing Th2 cells mediated by the transcription factor GATA binding protein 3 (GATA3).

Traditional Concept—Master Transcription Factor and Signature Cytokines

The traditional concept of CD4 T cell differentiation is that each CD4 T lineage has its own master transcription factor and signature cytokines, such as T-bet for IFN-γ-producing Th1, GATA3 for IL-4-producing Th2 and RORγt for IL-17-producing Th17 cells. However, recent studies have challenged this concept:[35,36] 
Several lineage specific transcription factors are expressed in more than one lineage: 
For example, T-bet is the master transcription factor for Th1 differentiation and IFN-γ production in Th1 cells, but it is also expressed in encephalitogenic Th17 cells and contributes to the encephalitogenicity of Th17 cells. Similarly, Bcl-6, the key transcription factor for T follicular helper cells (Tfh), is also expressed in early-stage Th1 cells, although the detailed function in Th1 cells have not been well characterized. These data suggest that lineage-defining transcription factors have functions beyond driving lineage-specific cytokine production.


 ForeignAutoimmune
Type I/allergy/atopy
(IgE)
Atopic eczema, Allergic urticaria, Allergic rhinitis (Hay fever), Allergic asthma, Anaphylaxis, Food allergy

Common allergies include: Milk, Egg, Peanut, Tree nut, Seafood, Soy, Wheat, Penicillin allergy
Eosinophilic esophagitis
Type II/ADCC
IgM IgG 
Hemolytic disease of the newborn
Cytotoxic
Autoimmune hemolytic anemia, Immune thrombocytopenic purpura, Bullous pemphigoid, Pemphigus vulgaris, Rheumatic fever, Goodpasture syndrome, Guillain–Barré syndrome

"Type V"/receptor
Graves' disease, Myasthenia gravis, Pernicious anemia
Type III
(Immune complex)
Henoch–Schönlein purpura, Hypersensitivity vasculitis, Reactive arthritis, Farmer's lung, Post-streptococcal glomerulonephritis, Serum sickness, Arthus reaction
Systemic lupus erythematosus, Subacute bacterial endocarditis, Rheumatoid arthritis
Type IV/cell-mediated
(T cells)
Allergic contact dermatitis, Mantoux test
Diabetes mellitus type 1, Hashimoto's thyroiditis, Multiple sclerosis, Coeliac disease, Giant-cell arteritis, Postorgasmic illness syndrome, Reactive arthritis 
Unknown/
multiple
Hypersensitivity pneumonitis, Allergic bronchopulmonary aspergillosis, Transplant rejection,  Latex allergy (I+IV) 
Sjögren syndrome, Autoimmune hepatitis, Autoimmune polyendocrine syndrome,
APS1, APS2 Autoimmune adrenalitis, Systemic autoimmune disease 
Table 2.  Source: [39]

Tregs


The regulatory T cells (Tregs) are a subpopulation of T cells that modulate the immune system, maintain tolerance to self-antigens, and prevent autoimmune disease

Tregs are immunosuppressive and generally suppress or downregulate induction and proliferation of effector T cells.[36] 
Tregs express the biomarkers CD4, FOXP3, and CD25 and are thought to be derived from the same lineage as naïve CD4 cells.[37] Because effector T cells also express CD4 and CD25, Tregs are very difficult to effectively discern from effector CD4+, making them difficult to study. 
Recent research has found that the cytokine TGFβ is essential for Tregs to differentiate from naïve CD4+ cells and is important in maintaining Treg homeostasis.[9]

Tregs and Autoimmune Diseases


The autoimmune diseases are attributable to loss of immune tolerance within the patients’ immune systems, which leads to the patients’ own immune cells overreacting or incorrectly attacking ‘self’ antigens in vital organs. 

Tregs expressing the transcription factor FoxP3 (FOXP3Tregs) are essential to maintain immunologic homeostasis, self-tolerance, and to prevent runaway immune responses.[23] 

Upregulation and/or maintenance of regulatory T cells (Tregs) during autoimmune insults may have therapeutic efficacy in autoimmune diseases.
The main trend in current therapies is to rely heavily on nonspecific immunosuppressive drugs such as steroids. 
However, these drugs relieve only some of the symptoms, and often have serious side effects particularly if they are used for the long term, due to their indiscriminate immunosuppressive function.


Type I Hypersensitivity


Usually, Treg cells, TR1, and Th3 cells at mucosal surfaces suppress Th2, mast cells, and eosinophils, which mediate allergic response. Deficits in Treg cells or their localization to mucosa have been implicated in asthma and atopic dermatitis.[32]

Attempts have been made to reduce hypersensitivity reactions by oral tolerance and other means of repeated exposure. Repeated administration of the allergen in slowly increasing doses, subcutaneously or sublingually appears to be effective for allergic rhinitis.[33] Repeated administration of antibiotics, which can form haptens to cause allergic reactions, can also reduce antibiotic allergies in children.[34]

Type II Hypersensitivity


Type II hypersensitivity reaction refers to an antibody-mediated immune reaction in which antibodies (IgG or IgM) are directed against cellular or extracellular matrix antigens with the resultant cellular destruction, functional loss, or damage to tissues.

For the treatment/management of Type II hypersensitivity, read [40] for more details.

Type III Hypersensitivity


In type III hypersensitivity reaction, an abnormal immune response is mediated by the formation of antigen-antibody aggregates called "immune complexes." They can precipitate in various tissues such as skin, joints, vessels, or glomeruli, and trigger the classical complement pathway. Complement activation leads to the recruitment of inflammatory cells (monocytes and neutrophils) that release lysosomal enzymes and free radicals at the site of immune complexes, causing tissue damage.
The principle feature that separates type III reactions from other hypersensitivity reactions is that in type III reaction, the antigen-antibody complexes are pre-formed in the circulation before their deposition in tissues.
For the treatment/management of Type III hypersensitivity, read [42] for more details.

Type IV Hypersensitivity


Type four hypersensitivity reaction is a cell-mediated reaction that can occur in response to contact with certain allergens resulting in what is called contact dermatitis or in response to some diagnostic procedures as in the tuberculin skin test. Certain allergens must be avoided to treat this condition.

For the treatment/management of Type IV hypersensitivity, read [43] for more details.

References

  1. Profound Treg perturbations correlate with COVID-19 severity
  2. Regulatory T-cell therapy shows promise for COVID-19-related respiratory distress
  3. All About Regulatory T cells (Tregs) & How to Increase Them
  4. Itch expression by Treg cells controls Th2 inflammatory responses
  5. Are You Th1 or Th2 Dominant? Effects + Immune Response
  6. Immunity against Helminths: Interactions with the Host and the Intercurrent Infections
  7. Endurance Exercise Diverts the Balance between Th17 Cells and Regulatory T Cells
  8. Influence of Dietary Components on Regulatory T Cells
  9. Chen W (August 2011). "Tregs in immunotherapy: opportunities and challenges". Immunotherapy. 3 (8): 911–4.
  10. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK (May 2006). "Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells". Nature. 441 (7090): 235–8.
  11. Vignali, D.A., Collison, L.W., and Workman, C.J. (2008). How regulatory T cells work. Nat. Rev. Immunol. 8, 523–532.
  12. Panduro, M., Benoist, C., and Mathis, D. (2016). Tissue Tregs. Annu. Rev Immunol. 34, 609–633.
  13. De Simone, M., Arrigoni, A., Rossetti, G., Gruarin, P., Ranzani, V., Politano, C., Bonnal, R.J.P., Provasi, E., Sarnicola, M.L., Panzeri, I. et al. (2016). Transcriptional Landscape of Human Tissue Lymphocytes Unveils Uniqueness of Tumor-Infiltrating T Regulatory Cells. Immunity. 45, 1135–1147.
  14. Plitas, G., Konopacki, C., Wu, K., Bos, P.D., Morrow, M., Putintseva, E.V., Chudakov, D.M., and Rudensky, A.Y. (2016). Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity. 45, 1122–1134.
  15. Magnuson, A.M., Kiner, E., Ergun, A., Park, J.S., Asinovski, N., Ortiz-Lopez, A., Kilcoyne, A., Paoluzzi-Tomada, E., Weissleder, R., Mathis, D. et al. (2018). Identification and validation of a tumor-infiltrating Treg transcriptional signature conserved across species and tumor types. Proc Natl Acad Sci U S A. 115, E10672–E10681.
  16. Lund, J.M., Hsing, L., Pham, T.T., and Rudensky, A.Y. (2008). Coordination of early protective immunity to viral infection by regulatory T cells. Science. 320, 1220–1224.
  17. Almanan, M., Raynor, J., Sholl, A., Wang, M., Chougnet, C., Cardin, R.D., and Hildeman, D.A. (2017). Tissue-specific control of latent CMV reactivation by regulatory T cells. PLoS Pathog. 13, e1006507.
  18. Belkaid, C. A. Piccirillo, S. Mendez, E. M. Shevach, D. L. Sacks, Nature 420, 502 (2002).
  19. Y. Belkaid, B. T. Rouse, Nat. Immunol. 6, 353 (2005).
  20. B. T. Rouse, P. P. Sarangi, S. Suvas, Immunol. Rev. 212, 272 (2006).
  21. G. Peng et al., Science 309, 1380 (2005).
  22. C. Pasare, R. Medzhitov, Science 299, 1033 (2003).
  23. Josefowicz, S.Z., Lu, L.F., and Rudensky, A.Y. (2012). Regulatory T cells: mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531–564.
  24. Extracellular NAD+ shapes the Foxp3+ regulatory T cell compartment through the ART2–P2X7 pathway
  25. Role of CD4+ CD25+ regulatory T cells in melatonin-mediated inhibition of murine gastric cancer cell growth in vivo and in vitro
  26. A comparison of low-dose cyclophosphamide treatment with artemisinin treatment in reducing the number of regulatory T cells in murine breast cancer model
  27. Metabolic regulation of regulatory T cell development and function
  28. Regulatory T cells Facilitate Cutaneous Wound Healing
  29. Bodnar RJ. Epidermal Growth Factor and Epidermal Growth Factor Receptor: The Yin and Yang in the Treatment of Cutaneous Wounds and Cancer. Adv Wound Care. 2013;2:24–29.
  30. Immune- and non-immune-mediated roles of regulatory T-cells during wound healing
  31. Th17 cytokines in mucosal immunity and inflammation
  32. Soyer, OU; Akdis M; Ring J; Behrendt H; Crameri R; Lauener R; Akdis CA (2012). "Mechanisms of peripheral tolerance to allergens". Allergy. 68 (2): 161–170.
  33. Petalas, K; Durham SR (2013). "Allergen immunotherapy for allergic rhinitis". Rhinology. 51 (2): 99–110.
  34. Cernadas, JR (Feb 2013). "Desensitization to antibiotics in children". Pediatr Allergy Immunol. 24 (1): 3–9.
  35. Oestreich KJ, Weinmann AS. Master regulators or lineage-specifying? Changing views on CD4+ T cell transcription factors. Nat Rev Immunol. 2012;12:799–804.
  36. Impact of suppressing retinoic acid-related orphan receptor gamma t (ROR)γt in ameliorating central nervous system autoimmunity
  37. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK (May 2006). "Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells". Nature. 441 (7090): 235–8.
  38. Curiel TJ (May 2007). "Tregs and rethinking cancer immunotherapy". The Journal of Clinical Investigation. 117 (5): 1167–74.
  39. Type II hypersensitivity (Wikipedia)
  40. Type II Hypersensitivity Reaction
  41. Committee IoMUVS. Adverse Events Associated with Childhood Vaccines: Evidence Bearing on Causality. Washington,(DC: National Academies Press (US), 1994.
  42. Type III Hypersensitivity Reaction
  43. Type IV Hypersensitivity Reaction
  44. Responsiveness of Naive CD4 T Cells to Polarizing Cytokine Determines the Ratio of Th1 and Th2 Cell Differentiation
  45. Expanding Role of T Cells in Human Autoimmune Diseases of the Central Nervous System (good)
    • Upregulation of serum IL-23 strengthens the case for Th17 activity
  46. Ulusoy C, Tuzun E, Kurtuncu M, Turkoglu R, Akman-Demir G, Eraksoy M. Comparison of the cytokine profiles of patients with neuronal-antibody-associated central nervous system disorders. Int J Neurosci (2012) 122(6):284–9.
  47. Dendritic Cells—Messengers between Innate and Adaptive Immune Systems
  48. Viruses and Autoimmunity: A Review on the Potential Interaction and Molecular Mechanisms

Osteoarthritis—Knowing the Basics

An estimated 10 to 15% of all adults aged over 60 have some degree of osteoarthritis (OA), with prevalence higher among women than men. According to the United Nations, by 2050, people aged over 60 will account for more than 20% of the world’s population.

Figure 1.  Synovial fluid (By Madhero88)

Osteoarthritis (OA)


Osteoarthritis (OA) is a degenerative disease of the articular joints with progressive nature involving the synovium, articular cartilage, and subchondral bone(see Figure 1).  It is characterized by the breakdown of cartilage, joint lining, ligaments, and underlying bone. It typically involves an entire joint, with the most commonly affected joints being the hips, knees, hands, and spine

Common manifestations of OA are stiffness and pain. There are a variety of risk factors for OA, including:[3] 
  • High-impact sports
  • Obesity
    • The risk of suffering osteoarthritis can be decreased with weight loss. 
    • This reduction of risk is related in part with the decrease of the load on the joint, but also in the decrease of fatty mass, the central adipose tissue and the low-level inflammation associated with obesity and systemic factors.
  • Bone deformities
The prevalence of OA increases with obesity and age.[4,5]  Knee OA is the most leading cause of disability and pain in the adult and old age population.

Figure 2.  Bone Grown (cancellous bone vs cortical bone)

Research Highlights


  • Levels of several inflammatory mediators, such as IL-6 and CCL2 (belongs to the CC chemokine family), were higher in OA sera compared to healthy sera[16]
  • Read here for a summary of dietary interventions that may be of benefit in OA
  • Obesity leads to low-grade systemic inflammation and weight reduction can reduce adipose tissue and restore normal secretion patterns.[13]
  • Leptin is associated with inflammation and cartilage degradation and may be involved in OA pathophysiology at a local and systemic level.[14-15]
    • Leptin can affect bone metabolism via direct signaling from the brain. 
    • Leptin decreases cancellous bone, but increases cortical bone (see Figure 2). This "cortical-cancellous dichotomy" may represent a mechanism for enlarging bone size, and thus bone resistance, to cope with increased body weight.
  • In a cell study with human chondrocytes (cells that make up cartilage), the anti-inflammatory action of IL-10 protects against damage from tumor necrosis factor-alpha (TNF-α), a pro-inflammatory mediator elevated in OA.[1] 
  • In another study, it shows that bioavailable turmeric extract is as effective as paracetamol in reducing pain and other symptoms of knee OA and found to be safe and more effective in reducing CRP and TNF-α.[3]

TNF Promotes Inflammatory Response


Tumor Necrosis Factor (TNF) promotes the inflammatory response, which, in turn, causes many of the clinical problems associated with autoimmune disorders such as
These disorders are sometimes treated by using a TNF inhibitor. On the other hand some patients treated with TNF inhibitors develop an aggravation of their disease or new onset of autoimmunity.  In one study, it provides some evidence that acute exercise may inhibit TNF production.[6]

TNF seems to have an immunosuppressive facet as well. One explanation for a possible mechanism is this observation that TNF has a positive effect on regulatory T cells (Tregs), due to its binding to the tumor necrosis factor receptor 2 (TNFR2).[7]

TNF-α and IL-6 concentrations are elevated in obesity.[8-10]  Monoclonal antibody against TNF-α is associated with increases rather than decreases in obesity, indicating that inflammation is the result, rather than the cause, of obesity.[10]

Figure 3.  Schematic diagram of the proposed mode of action for type II collagen (UC-II) 

UC-II®


UC-II® contains active epitopes that are able to interact with Peyer’s patches and induce oral tolerance.  A possible mechanism of action for UC-II activity is briefly summarized below (see Figure 3):
  1. Transforms naïve T-cells into Treg 
    • When consumed, UC-II® is believed to be taken up by the Peyer’s patches, where it activates immune cells. It transforms naive T-cells into T regulatory (Treg) cells that specifically target type II collagen
  2. Treg cells then migrate through the circulation
  3. Encounters type II collagen in joint cartilage
    • When they recognize type II collagen in joint cartilage, Treg cells secrete anti-inflammatory mediators (cytokines), including TGF-βIL-4 and IL-10
      • Note that the effect of TGF-β has been shown to be highly context-dependent.[11]
    • This action helps reduce joint inflammation and promotes cartilage repair. 
This process initiates anti-inflammatory and cartilage protective pathways that prevent the immune system from injuring its joint cartilage while promoting cartilage repair and regeneration. 


Summary of cytokines and their functions

CytokineFamilyMain sourcesFunction
IL-1βIL-1Macrophages, monocytesPro-inflammation, proliferation, apoptosis, differentiation
IL-4IL-4Th-cellsAnti-inflammation, T-cell and B-cell proliferation, B-cell differentiation
IL-6IL-6Macrophages, T-cells, adipocytePro-inflammation, differentiation, cytokine production
IL-8CXCMacrophages, epithelial cells, endothelial cellsPro-inflammation, chemotaxis, angiogenesis
IL-10IL-10Monocytes, T-cells, B-cellsAnti-inflammation, inhibition of the pro-inflammatory cytokines
IL-12IL-12Dendritic cells, macrophages, neutrophilsPro-inflammation, cell differentiation, activates NK cell
IL-11IL-6Fibroblasts, neurons, epithelial cellsAnti-inflammation, differentiation, induces acute phase protein
TNF-αTNFMacrophages, NK cells, CD4+lymphocytes, adipocytePro-inflammation, cytokine production, cell proliferation, apoptosis, anti-infection
IFN-γINFT-cells, NK cells, NKT cellsPro-inflammation, innate, adaptive immunity anti-viral
GM-CSFIL-4T-cells, macrophages, fibroblastsPro-inflammation, macrophage activation, increase neutrophil and monocyte function
TGF-βTGFMacrophages, T cellsAnti-inflammation, inhibition of pro-inflammatory cytokine production


References

  1. Müller R.D., John T., Kohl B., Oberholzer A., Gust T., Hostmann A., Hellmuth M., Laface D., Hutchins B., Laube G., et al. IL-10 overexpression differentially affects cartilage matrix gene expression in response to TNF-alpha in human articular chondrocytes in vitro. Cytokine. 2008;44:377–385.
  2. Transmission, pathogenesis, replication of SARS-CoV-2 (COVID-19)
  3. Bioavailable turmeric extract for knee osteoarthritis: a randomized, non-inferiority trial versus paracetamol
  4. Altman R, Asch E, Bloch G, et al. Development of criteria for the classification and reporting of osteoarthritis: classification of osteoarthritis of the knee. Arthritis Rheum. 1986;29:1039–49.
  5. Hochberg MC, Altman RD, April KT, et al. American College of Rheumatology 2012 recommendations for the use of nonpharmacologic and pharmacologic therapies in osteoarthritis of the hand, hip, and knee. Arthritis Care Res. 2012;64:465–74.
  6. Pedersen BK (December 2009). "The diseasome of physical inactivity – and the role of myokines in muscle–fat cross talk". J Physiol. 587 (23): 5559–5568.
  7. Salomon BL, Leclerc M, Tosello J, Ronin E, Piaggio E, Cohen JL (2018). "Tumor Necrosis Factor α and Regulatory T Cells in Oncoimmunology". Front. Immunol. 9: 444.
  8. Coppack SW (August 2001). "Pro-inflammatory cytokines and adipose tissue". The Proceedings of the Nutrition Society. 60 (3): 349–56.
  9. Kern L, Mittenbühler MJ, Vesting AJ, Wunderlich FT (2018). "Obesity-Induced TNFα and IL-6 Signaling: The Missing Link between Obesity and Inflammation-Driven Liver and Colorectal Cancers". cancers. 11(1): 24.
  10. Virdis A, Colucci R, Bernardini N, Masi S (2019). "Microvascular Endothelial Dysfunction in Human Obesity: Role of TNF-α". The Journal of Clinical Endocrinology and Metabolism. 104 (2): 341–348.
  11. Wahl SM (February 2007). "Transforming growth factor-beta: innately bipolar". Current Opinion in Immunology. 19 (1): 55–62.
  12. What is the evidence for a role for diet and nutrition in osteoarthritis
  13. Hauner H. Secretory factors from human adipose tissue and their functional role. Proc Nutr Soc. 2005 May; 64(2):163-9.
  14. Leptin produced by joint white adipose tissue induces cartilage degradation via upregulation and activation of matrix metalloproteinases.
  15. Leptin in osteoarthritis: Focus on articular cartilage and chondrocytes.
  16. Sohn DH, Sokolove J, Sharpe O, Erhart JC, Chandra PE, Lahey LJ, Lindstrom TM, Hwang I, Boyer KA, Andriacchi TP, et al. Plasma proteins present in osteoarthritic synovial fluid can stimulate cytokine production via Toll-like receptor 4. Arthritis Res Ther. 2012;14:R7.
  17. Slide show: Hand exercises for people with arthritis (Mayo Clinic)
  18. Sulforaphane represses matrix-degrading proteases and protects cartilage from destruction in vitro and in vivo
    • Sulforaphane can be found in cruciferous vegetables like broccoli, kale, cabbage, and watercress.