Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Inflamm Bowel Dis ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38334263

ABSTRACT

Despite a known role for α4ß7 and MAdCAM-1 for the recruitment of antibody secreting cells to the lactating mammary gland, vedolizumab which targets integrin α4ß7 did not lower breastmilk IgA in lactating mothers with IBD receiving the drug. It is likely that antibody secreting cells alternatively employ α4ß1 to arrest on VCAM-1 also expressed by the lactating mammary gland.

2.
Inflamm Bowel Dis ; 29(8): 1327-1341, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36943800

ABSTRACT

Eighty percent of antibody secreting cells (ASCs) are found in the intestine, where they produce grams of immunoglobulin (Ig) A daily. immunoglobulin A is actively transcytosed into the lumen, where it plays a critical role in modulating the gut microbiota. Although loss of immune tolerance to bacterial antigens is the likely trigger of the dysregulated immune response that characterizes inflammatory bowel disease (IBD), little effort has been placed on understanding the interface between B cells, IgA, and the microbiota during initiation or progression of disease. This may be in part due to the misleading fact that IgA-deficient humans are mostly asymptomatic, likely due to redundant role of secretory (S) IgM. Intestinal B cell recruitment is critically dependent on integrin α4ß7-MAdCAM-1 interactions, yet antibodies that target α4ß7 (ie, vedolizumab), MAdCAM-1 (ie, ontamalimab), or both ß7 integrins (α4ß7 and αE [CD103] ß7; etrolizumab) are in clinical use or development as IBD therapeutics. The effect of such interventions on the biology of IgA is largely unknown, yet a single dose of vedolizumab lowers SIgA levels in stool and weakens the oral immunization response to cholera vaccine in healthy volunteers. Thus, it is critical to further understand the role of these integrins for the migration of ASC and other cellular subsets during homeostasis and IBD-associated inflammation and the mode of action of drugs that interfere with this traffic. We have recently identified a subset of mature ASC that employs integrin αEß7 to dock with intestinal epithelial cells, predominantly in the pericryptal region of the terminal ileum. This role for the integrin had not been appreciated previously, nor the αEß7-dependent mechanism of IgA transcytosis that it supports. Furthermore, we find that B cells more than T cells are critically dependent on α4ß7-MAdCAM-1 interactions; thus MAdCAM-1 blockade and integrin-ß7 deficiency counterintuitively hasten colitis in interleukin-10-deficient mice. In both cases, de novo recruitment of IgA ASC to the intestinal lamina propria is compromised, leading to bacterial overgrowth, dysbiosis, and lethal colitis. Thus, despite the safe and effective use of anti-integrin antibodies in patients with IBD, much remains to be learned about their various cell targets.


Loss of immune tolerance to bacterial antigens represents the likely trigger of the dysregulated immune response that characterizes IBD, yet the interface between B cells, IgA, and the microbiota during initiation and progression of disease has been understudied. Here we review important aspects of the biology of IgA, its role on the control of the microbiota in mouse models, and its potential role on the pathogenesis of IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Mice , Animals , Immunoglobulin A, Secretory , Integrins , Intestines , Immunoglobulin A
3.
Medicine (Baltimore) ; 101(36): e30216, 2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36086782

ABSTRACT

Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn disease (CD), has emerged as a global disease with an increasing incidence in developing and newly industrialized regions such as South America. This global rise offers the opportunity to explore the differences and similarities in disease presentation and outcomes across different genetic backgrounds and geographic locations. Our study includes 265 IBD patients. We performed an exploratory analysis of the databases of Chilean and North American IBD patients to compare the clinical phenotypes between the cohorts. We employed an unsupervised machine-learning approach using principal component analysis, uniform manifold approximation, and projection, among others, for each disease. Finally, we predicted the cohort (North American vs Chilean) using a random forest. Several unsupervised machine learning methods have separated the 2 main groups, supporting the differences between North American and Chilean patients with each disease. The variables that explained the loadings of the clinical metadata on the principal components were related to the therapies and disease extension/location at diagnosis. Our random forest models were trained for cohort classification based on clinical characteristics, obtaining high accuracy (0.86 = UC; 0.79 = CD). Similarly, variables related to therapy and disease extension/location had a high Gini index. Similarly, univariate analysis showed a later CD age at diagnosis in Chilean IBD patients (37 vs 24; P = .005). Our study suggests a clinical difference between North American and Chilean IBD patients: later CD age at diagnosis with a predominantly less aggressive phenotype (39% vs 54% B1) and more limited disease, despite fewer biological therapies being used in Chile for both diseases.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Chile/epidemiology , Colitis, Ulcerative/genetics , Ethnicity , Humans , Inflammatory Bowel Diseases/diagnosis , North America/epidemiology , Phenotype
4.
Mucosal Immunol ; 15(1): 109-119, 2022 01.
Article in English | MEDLINE | ID: mdl-34433904

ABSTRACT

T and B cells employ integrin α4ß7 to migrate to intestine under homeostatic conditions. Whether those cells differentially rely on α4ß7 for homing during inflammatory conditions has not been fully examined. This may have implications for our understanding of the mode of action of anti-integrin therapies in inflammatory bowel disease (IBD). Here, we examined the role of α4ß7 integrin during chronic colitis using IL-10-/- mice, ß7-deficient IL-10-/-, IgA-deficient IL-10-/- mice, and antibody blockade of MAdCAM-1. We found that α4ß7 was predominantly expressed by B cells. ß7 deficiency and MAdCAM-1 blockade specifically depleted antibody secreting cells (ASC) (not T cells) from the colonic LP, leading to a fecal pan-immunoglobulin deficit, severe colitis, and alterations of microbiota composition. Colitis was not due to defective regulation, as dendritic cells (DC), regulatory T cells, retinaldehyde dehydrogenase (RALDH) expression, activity, and regulatory T/B-cell cytokines were all comparable between the strains/treatment. Finally, an IgA deficit closely recapitulated the clinical phenotype and altered microbiota composition of ß7-deficient IL-10-/- mice. Thus, a luminal IgA deficit contributes to accelerated colitis in the ß7-deficient state. Given the critical/nonredundant dependence of IgA ASC on α4ß7:MAdCAM-1 for intestinal homing, B cells may represent unappreciated targets of anti-integrin therapies.


Subject(s)
Antibody-Producing Cells/immunology , Cell Adhesion Molecules/metabolism , Colitis/immunology , Gastrointestinal Microbiome/immunology , Inflammatory Bowel Diseases/immunology , Integrin alpha4/metabolism , Integrin beta Chains/metabolism , Intestines/physiology , Mucoproteins/metabolism , Animals , Chronic Disease , Disease Models, Animal , Humans , Immunoglobulin A/metabolism , Immunomodulation , Integrin beta Chains/genetics , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Front Cell Infect Microbiol ; 11: 752304, 2021.
Article in English | MEDLINE | ID: mdl-34869061

ABSTRACT

Over the last several years, many advances have been made in understanding the role of bacteria in the pathogenesis of gastrointestinal cancers. Beginning with Helicobacter pylori being recognized as the first bacterial carcinogen and the causative agent of most gastric cancers, more recent studies have examined the role of enteric microbes in colorectal cancer. In the digestive tract, these communities are numerous and have a complex interrelationship with local immune/inflammatory responses that impact the health of the host. As modifying the microbiome in the stomach has decreased the risk of gastric cancer, modifying the distal microbiome may decrease the risk of colorectal cancers. To date, very few studies have considered the notion that mucosal lymphocyte-dependent immune memory may confound attempts to change the microbial components in these communities. The goal of this review is to consider some of the factors impacting host-microbial interactions that affect colorectal cancer and raise questions about how immune memory responses to the local microbial consortium affect any attempt to modify the composition of the intestinal microbiome.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Helicobacter pylori , Colorectal Neoplasms/prevention & control , Humans , Immunologic Memory , Stomach
6.
Immunohorizons ; 5(12): 953-971, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911745

ABSTRACT

Lymphocytes within the intestinal epithelial layer (IEL) in mammals have unique composition compared with their counterparts in the lamina propria. Little is known about the role of some of the key colonic IEL subsets, such as TCRαß+CD8+ T cells, in inflammation. We have recently described liver-enriched innate-like TCRαß+CD8αα regulatory T cells, partly controlled by the non-classical MHC molecule, Qa-1b, that upon adoptive transfer protect from T cell-induced colitis. In this study, we found that TCRαß+CD8αα T cells are reduced among the colonic IEL during inflammation, and that their activation with an agonistic peptide leads to significant Qa-1b-dependent protection in an acute model of colitis. Cellular expression of Qa-1b during inflammation and corresponding dependency in peptide-mediated protection suggest that Batf3-dependent CD103+CD11b- type 1 conventional dendritic cells control the protective function of TCRαß+CD8αα T cells in the colonic epithelium. In the colitis model, expression of the potential barrier-protective gene, Muc2, is enhanced upon administration of a Qa-1b agonistic peptide. Notably, in steady state, the mucin metabolizing Akkermansia muciniphila was found in significantly lower abundance amid a dramatic change in overall microbiome and metabolome, increased IL-6 in explant culture, and enhanced sensitivity to dextran sulfate sodium in Qa-1b deficiency. Finally, in patients with inflammatory bowel disease, we found upregulation of HLA-E, a Qa-1b analog with inflammation and biologic non-response, in silico, suggesting the importance of this regulatory mechanism across species.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Homeostasis/immunology , Intestine, Large/immunology , Adoptive Transfer , Animals , Antigens, CD , CD8 Antigens , Female , Integrin alpha Chains , Intestine, Large/metabolism , Mammals/immunology , Mammals/metabolism , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocytes, Regulatory/immunology
7.
Mucosal Immunol ; 14(6): 1347-1357, 2021 11.
Article in English | MEDLINE | ID: mdl-34417548

ABSTRACT

Efficient IgA transcytosis is critical for the maintenance of a homeostatic microbiota. In the canonical model, locally-secreted dimeric (d)IgA reaches the polymeric immunoglobulin receptor (pIgR) on intestinal epithelium via simple diffusion. A role for integrin αE(CD103)ß7 during transcytosis has not been described, nor its expression by intestinal B cell lineage cells. We found that αE-deficient (αE-/-) mice have a luminal IgA deficit, despite normal antibody-secreting cells (ASC) recruitment, local IgA production and increased pIgR expression. This deficit was not due to dendritic cell (DC)-derived retinoic acid (RA) nor class-switching defects, as stool from RAG-/- mice reconstituted with αE-/- B cells was also IgA deficient. Flow cytometric, ultrastructural and transcriptional profiling showed that αEß7-expressing ASC represent an undescribed subset of terminally-differentiated intestinal plasma cells (PC) that establishes direct cell to cell contact with intestinal epithelium. We propose that IgA not only reaches pIgR through diffusion, but that αEß7+ PC dock with E-cadherin-expressing intestinal epithelium to directly relay IgA for transcytosis into the intestinal lumen.


Subject(s)
Immunoglobulin A/immunology , Integrins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Transcytosis/immunology , Animals , Cell Differentiation/immunology , Gene Expression , Gene Expression Regulation , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/immunology , Integrins/deficiency , Integrins/metabolism , Intestinal Mucosa/ultrastructure , Lymphocyte Activation , Mice , Mice, Knockout , Models, Biological , Plasma Cells/cytology , Plasma Cells/ultrastructure
10.
Drugs ; 81(9): 985-1002, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33983615

ABSTRACT

Sphingosine-1-phosphate (S1P) is a bioactive lipid metabolite that exerts its actions by engaging 5 G-protein-coupled receptors (S1PR1-S1PR5). S1P receptors are involved in several cellular and physiological events, including lymphocyte/hematopoietic cell trafficking. An S1P gradient (low in tissues, high in blood), maintained by synthetic and degradative enzymes, regulates lymphocyte trafficking. Because lymphocytes live long (which is critical for adaptive immunity) and recirculate thousands of times, the S1P-S1PR pathway is involved in the pathogenesis of immune-mediated diseases. The S1PR1 modulators lead to receptor internalization, subsequent ubiquitination, and proteasome degradation, which renders lymphocytes incapable of following the S1P gradient and prevents their access to inflammation sites. These drugs might also block lymphocyte egress from lymph nodes by inhibiting transendothelial migration. Targeting S1PRs as a therapeutic strategy was first employed for multiple sclerosis (MS), and four S1P modulators (fingolimod, siponimod, ozanimod, and ponesimod) are currently approved for its treatment. New S1PR modulators are under clinical development for MS, and their uses are being evaluated to treat other immune-mediated diseases, including inflammatory bowel disease (IBD), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and psoriasis. A clinical trial in patients with COVID-19 treated with ozanimod is ongoing. Ozanimod and etrasimod have shown promising results in IBD; while in phase 2 clinical trials, ponesimod has shown improvement in 77% of the patients with psoriasis. Cenerimod and amiselimod have been tested in SLE patients. Fingolimod, etrasimod, and IMMH001 have shown efficacy in RA preclinical studies. Concerns relating to S1PR modulators are leukopenia, anemia, transaminase elevation, macular edema, teratogenicity, pulmonary disorders, infections, and cardiovascular events. Furthermore, S1PR modulators exhibit different pharmacokinetics; a well-established first-dose event associated with S1PR modulators can be mitigated by gradual up-titration. In conclusion, S1P modulators represent a novel and promising therapeutic strategy for immune-mediated diseases.


Subject(s)
Immune System Diseases/drug therapy , Immune System Diseases/metabolism , Lysophospholipids/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Pharmaceutical Preparations/administration & dosage , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Animals , Humans , Sphingosine/metabolism
11.
Therap Adv Gastroenterol ; 13: 1756284820971214, 2020.
Article in English | MEDLINE | ID: mdl-33240396

ABSTRACT

BACKGROUND: Vedolizumab, an α4ß7 integrin antagonist, is an effective therapy for Crohn's disease (CD). Biomarkers are needed to guide therapy and predict outcomes. This study evaluated biomarker concentrations and outcomes in patients with CD undergoing vedolizumab treatment. METHODS: Sera at weeks 0, 2, 6, 14, and ⩾26 were collected from vedolizumab-treated, refractory CD patients. Concentrations of soluble (s)-Vascular Cell Adhesion Molecule (VCAM)-1, s-Intercellular Cell Adhesion Molecule (ICAM)-1, s-Mucosal Addressin Cell Adhesion Molecule (MAdCAM)-1, and s-α4ß7 integrin were evaluated for associations with achieving endoscopic remission. RESULTS: A total of 22 patients with CD were included. In all patients, s-MAdCAM-1 decreased significantly and s-α4ß7 increased compared with baseline. s-VCAM-1 and s-ICAM-1 changed differentially in patients who achieved remission. At week 6, median s-VCAM-1 (859.6 ng/ml versus 460.3 ng/ml, p = 0.03) and s-ICAM-1 (545.7 ng/ml versus 286.2 ng/ml, p = 0.03) concentrations were higher in patients who achieved endoscopic remission compared with those who did not, and similar differences were observed for s-ICAM-1 concentrations in patients who achieved clinical remission, compared with those who did not (669.1 ng/ml versus 291.0 ng/ml, p = 0.04). Week 14 s-α4ß7 concentrations were lower in patients who achieved endoscopic remission, compared with those who did not (7.5 ng/ml versus 17.6 ng/ml, p = 0.020). CONCLUSION: In all vedolizumab-treated CD patients, s-MAdCAM-1 decreased significantly and s-α4ß7 increased. However, higher concentrations of s-ICAM-1 and s-VCAM-1 at week 6 and lower concentrations of s-α4ß7 at week 14 differentiated patients who achieved endoscopic remission. These findings may help identify early predictors of response to vedolizumab treatment in patients with CD. Further validation in less refractory CD patients is needed.

12.
J Crohns Colitis ; 14(10): 1364-1377, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-32239151

ABSTRACT

BACKGROUND AND AIMS: Intestinal biopsy sampling during IBD trials represents a valuable adjunct strategy for understanding drug responses at the tissue level. Given the length and distinctive embryonic origins of the proximal and distal colon, we investigated whether inherent regional differences of immune cell composition could introduce confounders when sampling different disease stages, or pre/post drug administration. Here, we capitalise on novel mass cytometry technology to perform deep immunophenotyping of distinct healthy colonic segments, using the limited numbers of biopsies that can be harvested from patients. METHODS: Biopsies [2.8 mm] were collected from the caecum, transverse colon, descending colon, and rectum of normal volunteers. Intestinal leukocytes were isolated, stained with a panel of 37 antibodies, and mass cytometry data acquired. RESULTS: Site-specific patterns of leukocyte localisation were observed. The proximal colon featured increased CD8+ T cells [particularly resident memory], monocytes, and CD19+ B cells. Conversely, the distal colon and rectum tissues exhibited enrichment for CD4+ T cells and antibody-secreting cells. The transverse colon displayed increased abundance of both γδ T cells and NK cells. Subsets of leukocyte lineages also displayed gradients of expression along the colon length. CONCLUSIONS: Our results show an inherent regional immune cell variation within colonic segments, indicating that regional mucosal signatures must be considered when assessing disease stages or the prospective effects of trial drugs on leukocyte subsets. Precise protocols for intestinal sampling must be implemented to allow for the proper interpretation of potential differences observed within leukocyte lineages present in the colonic lamina propria.


Subject(s)
Antigens, CD19 , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Inflammatory Bowel Diseases , Intestinal Mucosa , Monocytes , Adult , Antigens, CD19/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Biopsy/methods , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , Female , Humans , Immunity, Cellular/drug effects , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Monocytes/immunology , Monocytes/pathology , Patient Acuity , Patient Selection
16.
Inflamm Bowel Dis ; 26(2): 216-228, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31807751

ABSTRACT

BACKGROUND: Lymphocytes recirculate from tissues to blood following the sphingosine-1-phosphate (S1P) gradient (low in tissues, high in blood), maintained by synthetic and degradative enzymes, among which the S1P lyase (SPL) irreversibly degrades S1P. The role of SPL in the intestine, both during homeostasis and IBD, is poorly understood. We hypothesized that modulation of tissue S1P levels might be advantageous over S1P receptor (S1PR) agonists (eg, fingolimod, ozanimod, etrasimod), as without S1PR engagement there might be less likelihood of potential off-target effects. METHODS: First we examined SPL mRNA transcripts and SPL localization in tissues by quantitative reverse transcription polymerase chain reaction and immunohistochemistry. The in vivo effects of the SPL inhibitors 4-deoxypyridoxine hydrochloride (30 mg/L) and 2-acetyl-4 (tetrahydroxybutyl)imidazole (50 mg/L) were assessed through their oral administration to adult TNF∆ARE mice, which spontaneously develop Crohn's-like chronic ileitis. The effect of SPL inhibition on circulating and tissue lymphocytes, transcriptional regulation of proinflammatory cytokines, and on the histological severity of ileitis was additionally examined. Tissue S1P levels were determined by liquid chromatography-mass spectrometry. Mechanistically, the potential effects of high S1P tissue levels on intestinal leukocyte apoptosis were assessed via terminal deoxynucleotidyl transferase dUTP nick end-labeling assay and annexin 5 staining. Finally, we examined the ability of T cells to home to the intestine, along with the effects of SPL inhibition on cellular subsets within immune compartments via flow and mass cytometry. RESULTS: S1P lyase was ubiquitously expressed. In the gut, immunohistochemistry predominantly localized it to small intestinal epithelia, although the lamina propria leukocyte fraction had higher mRNA transcripts. Inhibition of SPL markedly increased local intestinal S1P levels, induced peripheral lymphopenia, downregulated proinflammatory cytokines, and attenuated chronic ileitis in mice. SPL inhibition reduced T and myeloid cells in secondary lymphoid tissues and the intestine and decreased naïve T-cell recruitment. The anti-inflammatory activity of SPL inhibition was not mediated by leukocyte apoptosis, nor by interference with the homing of lymphocytes to the intestine, and was independent of its peripheral lymphopenic effect. However, SPL inhibition promoted thymic atrophy and depleted late immature T cells (CD4+CD8+ double positive), with accumulation of mature CD4+CD8- and CD4-CD8+ single-positive cells. CONCLUSIONS: Inhibition of the S1P lyase alters the S1P gradient and attenuates chronic ileitis via central immunosuppression. SPL inhibition could represent a potential way to tame an overactive immune response during IBD and other T-cell-mediated chronic inflammatory diseases.


Subject(s)
Aldehyde-Lyases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Crohn Disease/drug therapy , Ileitis/drug therapy , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Thymocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Crohn Disease/metabolism , Crohn Disease/pathology , Ileitis/metabolism , Ileitis/pathology , Mice , Sphingosine/metabolism , Thymocytes/pathology , Tumor Necrosis Factor-alpha/genetics
17.
Cell Mol Gastroenterol Hepatol ; 9(3): 369-385, 2020.
Article in English | MEDLINE | ID: mdl-31707128

ABSTRACT

BACKGROUND & AIMS: Integrin α4ß7 mediates lymphocyte trafficking to the gut and gut-associated lymphoid tissues, a process critical for recruitment of effector lymphocytes from the circulation to the gut mucosa in inflammatory bowel disease (IBD) and murine models of intestinal inflammation. Antibody blockade of ß7 integrins generally is efficacious in IBD; however, some patients fail to respond, and a few patients can experience exacerbations. This study examined the effects of loss of ß7 integrin function in murine models of IBD. METHODS: In a mouse IBD model caused by lack of interleukin 10, a cytokine important in CD25hiFoxP3+ regulatory T cell (Treg) function, genetic deletion of ß7 integrin or antibody blockade of α4ß7-mucosal addressin cell adhesion molecule-1 interaction paradoxically exacerbated colitis. RESULTS: Loss of ß7 impaired the capacity of Tregs homing to the gut and therefore suppress intestinal inflammation in an adoptive T-cell transfer model; however, the intrinsic suppressive function of ß7-deficient Tregs remained intact, indicating that the ß7 deficiency selectively impacts gut homing. Deletion of ß7 integrin did not worsen colitis in an acute dextran sodium sulfate model in which Treg number and function were normal. CONCLUSIONS: In Integrin subunit beta (Itgb)7-/-Il10-/- mice, loss of ß7-dependent Treg homing to gut-associated lymphoid tissues combined with loss of intrinsic Treg function exacerbated intestinal inflammation. These results suggest that IBD patients with reduced CD25hiFoxP3+ Treg numbers or function or lack of interleukin 10 could be at risk for failure of α4ß7 blocking therapy.


Subject(s)
Colitis, Ulcerative/immunology , Integrin beta Chains/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Adhesion/immunology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Integrin beta Chains/genetics , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism
18.
Front Pharmacol ; 10: 212, 2019.
Article in English | MEDLINE | ID: mdl-30930775

ABSTRACT

The inflammatory Bowel diseases (IBDs) are a chronic, relapsing inflammatory diseases of the gastrointestinal tract with heterogeneous behavior and prognosis. The introduction of biological therapies including anti-TNF, anti-IL-12/23, and anti-integrins, has revolutionized the treatment of IBD, but these drugs are not universally effective. Due to the complex molecular structures of biologics, they are uniformly immunogenic. New discoveries concerning the underlying mechanisms involved in the pathogenesis of IBD have allowed for progress in the development of new treatment options. The advantage of small molecules (SMs) over biological therapies includes their lack of immunogenicity, short half-life, oral administration, and low manufacturing cost. Among these, the Janus Kinases (JAKs) inhibition has emerged as a novel strategy to modulate downstream cytokine signaling during immune-mediated diseases. These drugs target various cytokine signaling pathways that participate in the pathogenesis of IBD. Tofacitinib, a JAK inhibitor targeting predominantly JAK1 and JAK3, has been approved for the treatment of ulcerative colitis (UC), and there are other specific JAK inhibitors under development that may be effective in Crohn's. Similarly, the traffic of lymphocytes can now be targeted by another SM. Sphingosine-1-phosphate receptor (S1PR) agonism is a novel strategy that acts, in part, by interfering with lymphocyte recirculation, through blockade of lymphocyte egress from lymph nodes. S1PR agonists are being studied in IBD and other immune-mediated disorders. This review will focus on SM drugs approved and under development, including JAK inhibitors (tofacitinib, filgotinib, upadacitinib, peficitinib) and S1PR agonists (KRP-203, fingolimod, ozanimod, etrasimod, amiselimod), and their mechanism of action.

19.
Clin Gastroenterol Hepatol ; 17(13): 2634-2643, 2019 12.
Article in English | MEDLINE | ID: mdl-31009791

ABSTRACT

Crohn's disease (CD) is an inflammatory bowel disease that can involve any region of the gastrointestinal tract. First described in 1932 as terminal ileitis or regional enteritis, it predominately involves the ileum with or without colonic involvement. Isolated colonic CD was first described in 1960 and since then the phenotypic classification of CD has evolved to stratify patients into isolated ileal, ileocolonic, or isolated colonic involvement. In the current review we evaluate the published literature regarding differences in epidemiology, natural history, pathogenesis, response to therapy, and disease monitoring, when stratified by disease location. Based on the available evidence consideration could be given to a new classification for CD, which splits it into ileum dominant (isolated ileal and ileocolonic) and isolated colonic disease. This may allow for a more optimized approach to clinical care and scientific research for CD.


Subject(s)
Colitis/physiopathology , Crohn Disease/classification , Crohn Disease/physiopathology , Ileitis/physiopathology , Autophagy/physiology , Colitis/epidemiology , Colitis/immunology , Colitis/therapy , Crohn Disease/epidemiology , Crohn Disease/therapy , Cytokines/immunology , Disease Progression , Gastrointestinal Microbiome/physiology , Humans , Ileitis/epidemiology , Ileitis/immunology , Ileitis/therapy , Risk Factors , T-Lymphocytes/immunology
20.
Inflamm Bowel Dis ; 25(2): 410-420, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30295781

ABSTRACT

Background: Vedolizumab inhibits α4ß7-mediated lymphocyte trafficking and is effective in ulcerative colitis (UC). This study evaluated drug and biomarker concentrations and patient outcomes during vedolizumab treatment in UC. Methods: Prospectively scored maintenance clinical (26.5 weeks; interquartile range [IQR], 16.3-37.0 weeks) and endoscopic (23.5 weeks; IQR, 16.8-35.6 weeks) outcomes were compared with serum vedolizumab concentrations, antivedolizumab antibodies, and serum biomarkers at baseline and weeks 2, 6, 14, and 26. A linear mixed-effects model compared biomarker trajectories over time between clinical and endoscopic remitters and nonremitters. Results: Thirty-two patients were included. Soluble (s)-tumor necrosis factor (TNF)-α, s-α4ß7, s-mucosal addressin cell adhesion molecule (s-MAdCAM-1), and s-amyloid A (s-AA) significantly changed with treatment. A linear mixed-effects model demonstrated that s-α4ß7 (P = 0.044) increased and s-MAdCAM-1 (P = 0.006) and s-vascular cell adhesion molecule-1 (s-VCAM-1, P = 0.001) decreased more rapidly in patients achieving clinical remission in maintenance. S-MAdCAM-1 (P = 0.005), s-intracellular adhesion molecule-1 (ICAM-1; P = 0.014), s-VCAM-1 (P < 0.001), and s-TNF (P = 0.052) decreased more rapidly in endoscopic remitters. In clinical remitters, higher week 14 (20.3 ng/mL vs 6.0 ng/mL; P = 0.013) and week 26 (14.1 ng/mL vs 8.6 ng/mL; P = 0.05) s-α4ß7 were observed. In endoscopic remitters, week 2 (6.7 pg/mL vs 17.8 pg/mL; P = 0.038) and week 6 (3.9 pg/mL vs 15.6 pg/mL; P = 0.005) s-TNF and week 14 s-VCAM (589.1 ng/mL vs 746.0 ng/mL; P = 0.05) were lower. Conclusion: Serum biomarkers were associated with outcomes in vedolizumab-treated UC patients. s-α4ß7 increased, whereas s-MAdCAM-1, s-VCAM-1, s-ICAM-1, and s-TNF decreased more rapidly in remitters. At individual time points, induction s-TNF and maintenance s-VCAM-1 concentrations were lower, whereas maintenance s-α4ß7 concentrations were higher in remitters.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/analysis , Colitis, Ulcerative/blood , Endoscopy, Gastrointestinal/methods , Gastrointestinal Agents/therapeutic use , Adolescent , Adult , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/drug therapy , Female , Follow-Up Studies , Humans , Male , Prognosis , Prospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...