Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Article in English | MEDLINE | ID: mdl-34654708

ABSTRACT

BACKGROUND AND OBJECTIVES: Costimulation by CD40 and its ligand CD40L (CD154) is important for the functional differentiation of T cells. Preclinical studies have recognized the importance of this costimulatory interaction in the pathogenesis of experimental models of multiple sclerosis (MS). To determine safety, pharmacokinetics, and immune effect of a humanized monoclonal antibody (mAb) against CD40 ligand (toralizumab/IDEC-131) in patients with relapsing-remitting MS (RRMS). METHODS: This single-institution open-label dose-escalation study (phase I) enrolled 12 patients with RRMS to receive 4 doses of 1, 5, 10, or 15 mg/kg of humanized αCD40L (toralizumab) IV infusion every other week. Patients were followed up to 18 weeks, annually, and finally at 5 years. In addition to safety and pharmacokinetics, other secondary and exploratory measurements are immune effects, clinical, MRI, laboratory, and neuropsychological evaluations. RESULTS: Fifteen adverse events, all of mild to moderate severity, were considered to be of possible or of unknown relationship to treatment. No serious adverse events, including thromboembolic events, occurred during the 18-week defined study period. Annual and long-term follow-up at 5 years revealed no delayed toxicity. Pharmacokinetics were nonlinear between the 5 and 10 mg/kg dose groups. The serum half-life of toralizumab was consistent between the dose groups with a mean of 15.3 days (SD = 1.9). Flow cytometry revealed no depletion of lymphocyte subsets. An increase in the CD25+/CD3+ and CD25+/CD4+ ratio and a shift toward an anti-inflammatory cytokine response were seen after treatment. DISCUSSION: Our study suggests that blocking CD40L is safe and well tolerated in patients with RRMS while increasing CD25 + T cells and anti-inflammatory cytokine profile. These findings support further studies to assess the efficacy of blocking CD40L as a potential treatment of RRMS. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence on the safety, pharmacokinetics, and immune effects of an mAb to CD40L in patients with RRMS.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Immunologic Factors/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Adult , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , CD40 Ligand , Female , Follow-Up Studies , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Male , Middle Aged , Outcome Assessment, Health Care
2.
Front Immunol ; 12: 662807, 2021.
Article in English | MEDLINE | ID: mdl-34025663

ABSTRACT

The symbiotic relationship between animals and their resident microorganisms has profound effects on host immunity. The human microbiota comprises bacteria that reside in the gastrointestinal tract and are involved in a range of inflammatory and autoimmune diseases. The gut microbiota's immunomodulatory effects extend to extraintestinal tissues, including the central nervous system (CNS). Specific symbiotic antigens responsible for inducing immunoregulation have been isolated from different bacterial species. Polysaccharide A (PSA) of Bacteroides fragilis is an archetypical molecule for host-microbiota interactions. Studies have shown that PSA has beneficial effects in experimental disease models, including experimental autoimmune encephalomyelitis (EAE), the most widely used animal model for multiple sclerosis (MS). Furthermore, in vitro stimulation with PSA promotes an immunomodulatory phenotype in human T cells isolated from healthy and MS donors. In this review, we discuss the current understanding of the interactions between gut microbiota and the host in the context of CNS inflammatory demyelination, the immunomodulatory roles of gut symbionts. More specifically, we also discuss the immunomodulatory effects of B. fragilis PSA in the gut-brain axis and its therapeutic potential in MS. Elucidation of the molecular mechanisms responsible for the microbiota's impact on host physiology offers tremendous promise for discovering new therapies.


Subject(s)
Brain/metabolism , Demyelinating Diseases/etiology , Demyelinating Diseases/metabolism , Disease Susceptibility , Feedback, Physiological , Gastrointestinal Tract/metabolism , Animals , Bacteroides fragilis/immunology , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gastrointestinal Microbiome/immunology , Humans , Immunomodulation , Polysaccharides, Bacterial/immunology
3.
Front Immunol ; 11: 510113, 2020.
Article in English | MEDLINE | ID: mdl-33193297

ABSTRACT

The intestinal microbiota constitutes a complex ecosystem in constant reciprocal interactions with the immune, neuroendocrine, and neural systems of the host. Recent molecular technological advances allow for the exploration of this living organ and better facilitates our understanding of the biological importance of intestinal microbes in health and disease. Clinical and experimental studies demonstrate that intestinal microbes may be intimately involved in the progression of diseases of the central nervous system (CNS), including those of affective and psychiatric nature. Gut microbes regulate neuroinflammatory processes, play a role in balancing the concentrations of neurotransmitters and could provide beneficial effects against neurodegeneration. In this review, we explore some of these reciprocal interactions between gut microbes and the CNS during experimental disease and suggest that therapeutic approaches impacting the gut-brain axis may represent the next avenue for the treatment of psychiatric disorders.


Subject(s)
Central Nervous System/immunology , Gastrointestinal Microbiome/immunology , Mental Disorders , Neurotransmitter Agents/immunology , Animals , Humans , Mental Disorders/immunology , Mental Disorders/microbiology
5.
Article in English | MEDLINE | ID: mdl-29311123

ABSTRACT

The microbiome can be defined as the sum of the microbial and host's genome. Recent information regarding this complex organ suggests that in animal models of multiple sclerosis (MS), the composition of the gut microbiome can be altered, giving rise to both the effector and regulatory phases of central nervous system (CNS) demyelination. Experimental findings during the past decade in animal models of MS have provided clear evidence for the significant role of gut microbes in both the effector and regulatory phase of this condition. There is mounting evidence in preliminary human studies suggesting that a dysbiotic MS gut microbiome could affect disease progression. We propose considering the gut microbiome as a key organ for the regulation of tolerance mechanisms and speculate that the gut microbiome is the major environmental risk factor for CNS demyelinating disease. Accordingly, we hypothesize that intervention of the gut microbiome could result in safer novel therapeutic strategies to treat MS.


Subject(s)
Gastrointestinal Microbiome/physiology , Multiple Sclerosis/microbiology , Multiple Sclerosis/physiopathology , Animals , Disease Progression , Humans , Multiple Sclerosis/immunology , Neuroimmunomodulation
6.
Ann Clin Transl Neurol ; 4(11): 825-829, 2017 11.
Article in English | MEDLINE | ID: mdl-29159195

ABSTRACT

Multiple sclerosis, an immune-mediated disease of the central nervous system, is characterized by the impaired function of regulatory cells that fail to suppress self-reactive effector cells. We have previously found that polysaccharide A, a capsular antigen derived from the human gut commensal Bacteroides fragilis, can induce a population of regulatory T cells. Herein, we demonstrate that naïve T cells isolated from patients with multiple sclerosis have the capacity to acquire regulatory characteristics when stimulated in vitro with polysaccharide A. This study demonstrates the amplification of a regulatory T cell response by a gut-derived commensal antigen in those with multiple sclerosis.

7.
Gut Microbes ; 8(6): 561-573, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28708466

ABSTRACT

The gut microbiome plays an important role in the development of inflammatory disease as shown using experimental models of central nervous system (CNS) demyelination. Gut microbes influence the response of regulatory immune cell populations in the gut-associated lymphoid tissue (GALT), which drive protection in acute and chronic experimental autoimmune encephalomyelitis (EAE). Recent observations suggest that communication between the host and the gut microbiome is bidirectional. We hypothesized that the gut microbiota differs between the acute inflammatory and chronic progressive stages of a murine model of secondary-progressive multiple sclerosis (SP-MS). This non-obese diabetic (NOD) model of EAE develops a biphasic pattern of disease that more closely resembles the human condition when transitioning from relapsing-remitting (RR)-MS to SP-MS. We compared the gut microbiome of NOD mice with either mild or severe disease to that of non-immunized control mice. We found that the mice which developed a severe secondary form of EAE harbored a dysbiotic gut microbiome when compared with the healthy control mice. Furthermore, we evaluated whether treatment with a cocktail of broad-spectrum antibiotics would modify the outcome of the progressive stage of EAE in the NOD model. Our results indicated reduced mortality and clinical disease severity in mice treated with antibiotics compared with untreated mice. Our findings support the hypothesis that there are reciprocal effects between experimental CNS inflammatory demyelination and modification of the microbiome providing a foundation for the establishment of early therapeutic interventions targeting the gut microbiome that could potentially limit disease progression.


Subject(s)
Bacterial Physiological Phenomena , Disease Models, Animal , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Multiple Sclerosis/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Physiological Phenomena/drug effects , Disease Progression , Dysbiosis/drug therapy , Dysbiosis/prevention & control , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/microbiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Gastrointestinal Microbiome/drug effects , Humans , Immunomodulation , Mice , Mice, Inbred NOD , Peptide Fragments/pharmacology , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
8.
Ann Transl Med ; 5(6): 145, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28462225

ABSTRACT

Recent findings suggest that the intestinal microbiota of patients suffering from relapsing remitting multiple sclerosis (MS) shows changes on the relative abundances of archaeal and bacterial genera. Although the richness and overall structure of the microbiota may be similar compared to the intestinal microbiota of healthy controls, elevated and reduced frequencies suggest a dysbiotic microbiota in MS. Over the past decade experimental evidence obtained in murine models of the disease highlighted the important relevance of the microbiota in the regulation of the immune system and in the severity of the disease. More recent findings on peripheral immune cells derived from human MS patients support the initial observations that changes in the microbiota may affect immunological pathways that could exacerbate disease. However, important questions remain to be answered. For instance, it is unclear whether dysbiosis precedes disease or, if in the contrary, an autoimmune disease such as MS can lead to gut dysbiosis. In this brief discussion, we speculate about this later possibility based on findings observed in murine models of disease. Further human studies are needed to answer the dilemma and determine specific immunomodulatory pathways that could have an impact on the therapeutic approaches to treat MS.

9.
J Neuroimmunol ; 303: 22-30, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087077

ABSTRACT

While examining the therapeutic value of anti-CD52 antibody against EAE/MS, we identified a unique subset of CD39+ Tregs in repopulating GALT tissues, a major lymphoid reservoir, which was accompanied by amelioration of disease. Furthermore, anti-CD52 treatment leads to increased expression of BDNF, IL-10, and SMAD3 in the brains of EAE mice. This condition is associated with suppression of IL-17, a critical inflammatory factor in EAE/MS progression. Additionally, we found elevated levels of CD4+CD39+ Tregs in PBMCs of RRMS patients treated with humanized anti-CD52 mAb. Thus, anti-CD52 can affect multiple immune mediated pathways involved in the pathogenesis of EAE/MS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Apyrase/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Forkhead Transcription Factors/metabolism , Glycoproteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Apyrase/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD52 Antigen , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Forkhead Transcription Factors/immunology , Glycoproteins/antagonists & inhibitors , Glycoproteins/immunology , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Treatment Outcome
10.
J Immunol ; 198(2): 596-604, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28069755

ABSTRACT

Autoimmune disorders of the CNS have complex pathogeneses that are not well understood. In multiple sclerosis and neuromyelitis optica spectrum disorders, T cells destroy CNS tissue, resulting in severe disabilities. Mounting evidence suggests that reducing inflammation in the CNS may start with modulation of the gut microbiome. The lymphoid tissues of the gut are specialized for the induction of regulatory cells, which are directly responsible for the suppression of CNS-damaging autoreactive T cells. Whether cause or effect, the onset of dysbiosis in the gut of patients with multiple sclerosis and neuromyelitis optica provides evidence of communication along the gut-brain axis. Thus, current and future therapeutic interventions directed at microbiome modulation are of considerable appeal.


Subject(s)
Autoimmunity/immunology , Demyelinating Autoimmune Diseases, CNS/immunology , Demyelinating Autoimmune Diseases, CNS/microbiology , Gastrointestinal Microbiome/immunology , Animals , Humans
11.
Transl Res ; 179: 126-138, 2017 01.
Article in English | MEDLINE | ID: mdl-27519147

ABSTRACT

There is considerable interest in trying to understand the importance of the gut microbiome in human diseases. The association between dysbiosis, an altered microbial composition, as related to human disease is being explored in the context of different autoimmune conditions, including multiple sclerosis (MS). Recent studies suggest that MS affects the composition of the gut microbiota by altering the relative abundances of specific bacteria and archaea species. Remarkably, some of the bacterial species shown reduced in the gut of MS patients are known to promote immunosuppressive regulatory T cells (Tregs). In MS, the function of a phenotype of Tregs that express CD39, an ectoenzyme involved in the catabolism of adenosine triphosphate as immunomodulatory cells, appears to be reduced. In this review, we discuss the involvement of the gut microbiota in the regulation of experimental models of central nervous system inflammatory demyelination and review the evidence that link the gut microbiome with MS. Further, we hypothesize that the gut microbiome is an essential organ for the control of tolerance in MS patients and a potential source for safer novel therapeutics.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Central Nervous System/pathology , Demyelinating Diseases/metabolism , Gastrointestinal Tract/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Gastrointestinal Microbiome , Humans
12.
Neurol Neuroimmunol Neuroinflamm ; 3(6): e291, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27766282

ABSTRACT

OBJECTIVE: To determine whether as an orally delivered treatment, teriflunomide, an inhibitor of the mitochondrial enzyme dihydroorotate dehydrogenase approved to treat relapsing forms of multiple sclerosis, could affect gut-associated lymphoid tissue (GALT) immune responses functionally. METHODS: C57BL/6 mice were treated orally with teriflunomide and flow cytometric analysis of immune GALT cells performed ex vivo, and adoptive transfer experiments were used to test the protective effects of GALT regulatory T (Treg) cells. RESULTS: Teriflunomide reduced the percentages of antigen-presenting cells of Peyer patches when compared to controls. Conversely, a significant increase of the relative frequency of CD39+ Treg cells was observed. In vivo, the protective effect of GALT-derived teriflunomide-induced CD39+ Treg cells was established by adoptive transfer into recipient experimental autoimmune encephalomyelitis mice. CONCLUSIONS: Our results identify specific GALT-derived CD39+ Treg cells as a mechanism of action that may contribute to the efficacy of teriflunomide during CNS inflammatory demyelination and as an oral therapeutic in relapsing multiple sclerosis.

13.
Curr Obes Rep ; 5(1): 51-64, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26865085

ABSTRACT

The gut-brain axis is a bi-directional integrated system composed by immune, endocrine, and neuronal components by which the gap between the gut microbiota and the brain is significantly impacted. An increasing number of different gut microbial species are now postulated to regulate brain function in health and disease. The westernized diet is hypothesized to be the cause of the current obesity levels in many countries, a major socio-economical health problem. Experimental and epidemiological evidence suggest that the gut microbiota is responsible for significant immunologic, neuronal, and endocrine changes that lead to obesity. We hypothesize that the gut microbiota, and changes associated with diet, affect the gut-brain axis and may possibly contribute to the development of mental illness. In this review, we discuss the links between diet, gut dysbiosis, obesity, and immunologic and neurologic diseases that impact brain function and behavior.


Subject(s)
Gastrointestinal Microbiome , Obesity/microbiology , Animals , Brain/physiology , Central Nervous System Diseases/complications , Central Nervous System Diseases/microbiology , Diet , Dysbiosis , Humans , Obesity/complications
14.
Lancet Neurol ; 15(1): 35-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26621682

ABSTRACT

BACKGROUND: Relapses of multiple sclerosis decrease during pregnancy, when the hormone estriol is increased. Estriol treatment is anti-inflammatory and neuroprotective in preclinical studies. In a small single-arm study of people with multiple sclerosis estriol reduced gadolinium-enhancing lesions and was favourably immunomodulatory. We assessed whether estriol treatment reduces multiple sclerosis relapses in women. METHODS: We did a randomised, double-blind, placebo-controlled phase 2 trial at 16 academic neurology centres in the USA, between June 28, 2007, and Jan 9, 2014. Women aged 18-50 years with relapsing-remitting multiple sclerosis were randomly assigned (1:1) with a random permuted block design to either daily oral estriol (8 mg) or placebo, each in combination with injectable glatiramer acetate 20 mg daily. Patients and all study personnel, except for pharmacists and statisticians, were masked to treatment assignment. The primary endpoint was annualised relapse rate after 24 months, with a significance level of p=0.10. Relapses were confirmed by an increase in Expanded Disability Status Scale score assessed by an independent physician. Analysis was by intention to treat. The trial is registered with ClinicalTrials.gov, number NCT00451204. FINDINGS: We enrolled 164 patients: 83 were allocated to the estriol group and 81 were allocated to the placebo group. The annualised confirmed relapse rate was 0.25 relapses per year (95% CI 0.17-0.37) in the estriol group versus 0.37 relapses per year (0.25-0.53) in the placebo group (adjusted rate ratio 0.63, 95% CI 0.37-1.05; p=0.077). The proportion of patients with serious adverse events did not differ substantially between the estriol group and the placebo group (eight [10%] of 82 patients vs ten [13%] of 76 patients). Irregular menses were more common in the estriol group than in the placebo group (19 [23%] vs three [4%], p=0.0005), but vaginal infections were less common (one [1%] vs eight [11%], p=0.0117). There were no differences in breast fibrocystic disease, uterine fibroids, or endometrial lining thickness as assessed by clinical examination, mammogram, uterine ultrasound, or endometrial lining biopsy. INTERPRETATION: Estriol plus glatiramer acetate met our criteria for reducing relapse rates, and treatment was well tolerated over 24 months. These results warrant further investigation in a phase 3 trial. FUNDING: National Institutes of Health, National Multiple Sclerosis Society, Conrad N Hilton Foundation, Jack H Skirball Foundation, Sherak Family Foundation, and the California Community Foundation.


Subject(s)
Estriol/administration & dosage , Glatiramer Acetate/administration & dosage , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Adjuvants, Immunologic/administration & dosage , Adult , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Middle Aged
15.
Gut Microbes ; 6(4): 234-42, 2015 Jul 04.
Article in English | MEDLINE | ID: mdl-26230152

ABSTRACT

Polysaccharide A (PSA) derived from the human commensal Bacteroides fragilis is a symbiosis factor that stimulates immunologic development within mammalian hosts. PSA rebalances skewed systemic T helper responses and promotes T regulatory cells (Tregs). However, PSA-mediated induction of Foxp3 in humans has not been reported. In mice, PSA-generated Foxp3(+) Tregs dampen Th17 activity thereby facilitating bacterial intestinal colonization while the increased presence and function of these regulatory cells may guard against pathological organ-specific inflammation in hosts. We herein demonstrate that PSA induces expression of Foxp3 along with CD39 among naïve CD4 T cells in vitro while promoting IL-10 secretion. PSA-activated dendritic cells are essential for the mediation of this regulatory response. When cultured with isolated Foxp3(+) Tregs, PSA enriched Foxp3 expression, enhanced the frequency of CD39(+)HLA-DR(+) cells, and increased suppressive function as measured by decreased TNFα expression by LPS-stimulated monocytes. Our findings are the first to demonstrate in vitro induction of human CD4(+)Foxp3(+) T cells and enhanced suppressive function of circulating Foxp3(+) Tregs by a human commensal bacterial symbiotic factor. Use of PSA for the treatment of human autoimmune diseases, in particular multiple sclerosis and inflammatory bowel disease, may represent a new paradigm in the approach to treating autoimmune disease.


Subject(s)
Bacteroides fragilis/immunology , Bacteroides fragilis/physiology , CD4-Positive T-Lymphocytes/immunology , Symbiosis , T-Lymphocyte Subsets/immunology , Antigens, CD/analysis , Apyrase/analysis , Cells, Cultured , Dendritic Cells/immunology , Forkhead Transcription Factors/analysis , Humans , Immunophenotyping , Lipopolysaccharides/immunology , T-Lymphocyte Subsets/chemistry
16.
Curr Treat Options Neurol ; 17(4): 344, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25843302

ABSTRACT

OPINION STATEMENT: The gut microbiome is made up of a wide range of (chiefly) bacterial species that colonize the small and large intestine. The human gut microbiome contains a subset of thousands of bacterial species, with up to 10(14) total bacteria. Studies examining this bacterial content have shown wide variations in which species are present between individuals. The gut microbiome has been shown to have profound effects on the development and maintenance of immune system in both animal models and in humans. A growing body of evidence has implicated the human gut microbiome in a range of disorders, including obesity, inflammatory bowel diseases, and cardiovascular disease. Animal studies present compelling evidence that the gut microbiome plays a significant role in the progression of demyelinating disease, and that modulation of the microbiome can lead to either exacerbation or amelioration of symptoms. Differences in diet, vitamin D insufficiency, smoking, and alcohol use have all been implicated as risk factors in MS, and all have the ability to affect the composition of the gut microbiota. Preliminary clinical trials aimed at modulating the gut microbiota in MS patients are underway and may prove to be a promising and lower-risk treatment option in the future.

17.
Ann Clin Transl Neurol ; 1(8): 622-31, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25356432

ABSTRACT

Multiple sclerosis (MS) is a complex disorder of the central nervous system that appears to be driven by a shift in immune functioning toward excess inflammation that results in demyelination and axonal loss. Beta interferons were the first class of disease-modifying therapies to be approved for patients with MS after treatment with this type I interferon improved the course of MS on both clinical and radiological measures in clinical trials. The mechanism of action of interferon-beta appears to be driven by influencing the immune system at many levels, including antigen-presenting cells, T cells, and B cells. One effect of these interactions is to shift cytokine networks in favor of an anti-inflammatory effect. The pleiotropic mechanism of action may be a critical factor in determining the efficacy of interferon-beta in MS. This review will focus on select immunological mechanisms that are influenced by this type I cytokine.

18.
FEBS Lett ; 588(22): 4214-22, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25286403

ABSTRACT

Humans are colonized after birth by microbial organisms that form a heterogeneous community, collectively termed microbiota. The genomic pool of this macro-community is named microbiome. The gut microbiota is essential for the complete development of the immune system, representing a binary network in which the microbiota interact with the host providing important immune and physiologic function and conversely the bacteria protect themselves from host immune defense. Alterations in the balance of the gut microbiome due to a combination of environmental and genetic factors can now be associated with detrimental or protective effects in experimental autoimmune diseases. These gut microbiome alterations can unbalance the gastrointestinal immune responses and influence distal effector sites leading to CNS disease including both demyelination and affective disorders. The current range of risk factors for MS includes genetic makeup and environmental elements. Of interest to this review is the consistency between this range of MS risk factors and the gut microbiome. We postulate that the gut microbiome serves as the niche where different MS risk factors merge, thereby influencing the disease process.


Subject(s)
Autoimmunity , Central Nervous System/immunology , Gastrointestinal Tract/microbiology , Microbiota , Animals , Demyelinating Autoimmune Diseases, CNS/immunology , Demyelinating Autoimmune Diseases, CNS/microbiology , Humans , Risk Factors
19.
FEBS Lett ; 588(22): 4101, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25239394
20.
J Interferon Cytokine Res ; 34(8): 605-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25084177

ABSTRACT

There is increasing support for the importance of risk factors such as genetic makeup, obesity, smoking, vitamin D insufficiency, and antibiotic exposure contributing to the development of autoimmune diseases, including human multiple sclerosis (MS). Perhaps the greatest environmental risk factor associated with the development of immune-mediated conditions is the gut microbiome. Microbial and helminthic agents are active participants in shaping the immune systems of their hosts. This concept is continually reinforced by studies in the burgeoning area of commensal-mediated immunomodulation. The clinical importance of these findings for MS is suggested by both their participation in disease and, perhaps of greater clinical importance, attenuation of disease severity. Observations made in murine models of central nervous system demyelinating disease and a limited number of small studies in human MS suggest that immune homeostasis within the gut microbiome may be of paramount importance in maintaining a disease-free state. This review describes three immunological factors associated with the gut microbiome that are central to cytokine network activities in MS pathogenesis: T helper cell polarization, T regulatory cell function, and B cell activity. Comparisons are drawn between the regulatory mechanisms attributed to first-line therapies and those described in commensal-mediated amelioration of central nervous system demyelination.


Subject(s)
B-Lymphocytes/immunology , Central Nervous System/immunology , Demyelinating Diseases/immunology , Intestines/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/immunology , Demyelinating Diseases/microbiology , Humans , Immune Tolerance , Immunity, Mucosal , Intestines/microbiology , Lymphocyte Activation , Microbiota , Symbiosis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...