Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Crit Rev Immunol ; 41(4): 39-53, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35381142

RESUMO

The TCR-mediated recognition of self and microbial protein antigens by CD8+ T cells presented by the relatively nonvariable, class Ib MHC molecule, Qa-1 in mice and HLA-E in humans, is emerging as an important arm of the immune response. In this brief review, we will cover key examples of Qa1/HLA-E-restricted CD8+ T cells and their role in immunity against microbes and in cancer, but also as an important immunoregulatory pathway complementary to the FoxP3+CD4+ Treg. Although much remains to be learned, increased understanding of HLA-E-targeted immune responses can be potentially exploited in the development of broader and complementary immunotherapeutics against bacteria/viruses, tumors, and autoimmune diseases.


Assuntos
Antígenos de Histocompatibilidade Classe I , Neoplasias , Animais , Linfócitos T CD8-Positivos , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Camundongos , Neoplasias/metabolismo , Neoplasias/terapia , Linfócitos T Reguladores , Antígenos HLA-E
2.
J Immunol ; 201(10): 3017-3035, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30322964

RESUMO

Innate immune mechanisms play an important role in inflammatory chronic liver diseases. In this study, we investigated the role of type I or invariant NKT (iNKT) cell subsets in the progression of nonalcoholic steatohepatitis (NASH). We used α-galactosylceramide/CD1d tetramers and clonotypic mAb together with intracytoplasmic cytokine staining to analyze iNKT cells in choline-deficient l-amino acid-defined (CDAA)-induced murine NASH model and in human PBMCs, respectively. Cytokine secretion of hepatic iNKT cells in CDAA-fed C57BL/6 mice altered from predominantly IL-17+ to IFN-γ+ and IL-4+ during NASH progression along with the downmodulation of TCR and NK1.1 expression. Importantly, steatosis, steatohepatitis, and fibrosis were dependent upon the presence of iNKT cells. Hepatic stellate cell activation and infiltration of neutrophils, Kupffer cells, and CD8+ T cells as well as expression of key proinflammatory and fibrogenic genes were significantly blunted in Jα18-/- mice and in C57BL/6 mice treated with an iNKT-inhibitory RAR-γ agonist. Gut microbial diversity was significantly impacted in Jα18-/- and in CDAA diet-fed mice. An increased frequency of CXCR3+IFN-γ+T-bet+ and IL-17A+ iNKT cells was found in PBMC from NASH patients in comparison with nonalcoholic fatty liver patients or healthy controls. Consistent with their in vivo activation, iNKT cells from NASH patients remained hyporesponsive to ex-vivo stimulation with α-galactosylceramide. Accumulation of plasmacytoid dendritic cells in both mice and NASH patients suggest their role in activation of iNKT cells. In summary, our findings indicate that the differential activation of iNKT cells play a key role in mediating diet-induced hepatic steatosis and fibrosis in mice and its potential involvement in NASH progression in humans.


Assuntos
Ativação Linfocitária/imunologia , Células T Matadoras Naturais/imunologia , Hepatopatia Gordurosa não Alcoólica/imunologia , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Subpopulações de Linfócitos T/imunologia
3.
BMC Immunol ; 20(1): 42, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31718550

RESUMO

BACKGROUND: Myeloid cells, especially mononuclear phagocytes, which include monocytes, macrophages and dendritic cells (DC), play vital roles in innate immunity, and in the initiation and maintenance of adaptive immunity. While T cell-associated activation pathways and cytokines have been identified and evaluated in inflammatory bowel disease (IBD) patients (Neurath, Nat Rev Gastroenterol Hepatol 14:269-78, 1989), the role of mononuclear phagocytes are less understood. Recent reports support the crucial role of DC subsets in the development of acute colitis models (Arimura et al., Mucosal Immunol 10:957-70, 2017), and suggest they may contribute to the pathogenesis of ulcerative colitis (UC) by inducing Th1/Th2/Th17 responses (Matsuno et al., Inflamm Bowel Dis 23:1524-34, 2017). RESULTS: We performed in silico analysis and evaluated the enrichment of immune cells, with a focus on mononuclear phagocytes in IBD patient colonic biopsies. Samples were from different gut locations, with different levels of disease severity, and with treatment response to current therapies. We observe enrichment of monocytes, M1 macrophages, activated DCs (aDCs) and plasmacytoid dendritic cells (pDCs) in inflamed tissues from various gut locations. This enrichment correlates with disease severity. Additionally, the same mononuclear phagocytes subsets are among the top enriched cell types in both infliximab and vedolizumab treatment non-responder samples. We further investigated the enrichment of selected DC and monocyte subsets based on gene signatures derived from a DC- and monocyte-focused single cell RNA-seq (scRNA-seq) study (Villani et al., Science 356:eaah4573, 2017), and verified enrichment in both inflamed tissues and those with treatment resistance. Moreover, we validated an increased mononuclear phagocyte subset abundance in a Dextran Sulphate Sodium (DSS) induced colitis model in C57Bl/6 mice representative of chronic inflammation. CONCLUSIONS: We conducted an extensive analysis of immune cell populations in IBD patient colonic samples and identified enriched subsets of monocytes, macrophages and dendritic cells in inflamed tissues. Understanding how they interact with other immune cells and other cells in the colonic microenvironment such as epithelial and stromal cells will help us to delineate disease pathogenesis.


Assuntos
Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Sistema Fagocitário Mononuclear/imunologia , Sistema Fagocitário Mononuclear/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Biópsia , Microambiente Celular , Colo/imunologia , Colo/metabolismo , Colo/patologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Resistência a Medicamentos , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/patologia , Infliximab/farmacologia , Infliximab/uso terapêutico , Contagem de Leucócitos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Sistema Fagocitário Mononuclear/patologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
4.
Am J Physiol Gastrointest Liver Physiol ; 316(5): G585-G597, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30817180

RESUMO

We investigated the migration of intestinal immune cells to the liver and their contribution to alcoholic liver disease. In mice fed ethanol, we found that an increased number of invariant natural killer T (iNKT) cells, which respond to the antigen presented by CD1d, migrated from mesenteric lymph nodes to the liver. iNKT cells react to lipid antigens, so we studied their activities in mice with intestinal epithelial cell-specific deletion of Pparg (PpargΔIEC) as a model for altering intestinal lipidomic profiles. Levels of CD1d increased in intestines of ethanol-fed PpargΔIEC mice, and in cell-tracking experiments, more iNKT cells migrated to the liver, compared with mice without disruption of Pparg. Livers of PpargΔIEC mice had increased markers of apoptosis and liver injury after ethanol feeding. iNKT cells isolated from livers of ethanol-fed PpargΔIEC mice induced apoptosis of cultured hepatocytes. An inhibitor of iNKT cells reduced ethanol-induced liver injury in PpargΔIEC mice. Duodenal tissues from patients with alcohol-use disorder have been found to have increased levels of CD1d compared with tissues from patients without alcohol overuse. Ethanol use, therefore, activates iNKT cells in the intestine to migrate to liver, where they-along with the resident hepatic iNKT cells-contribute to hepatocyte death and injury. NEW & NOTEWORTHY In this article, we studied migration of intestinal immune cells into the liver in response to ethanol-induced liver disease. We found that chronic ethanol feeding induces expression of CD1d by enterocytes, which activate invariant natural killer T (iNKT) cells in mesenteric lymph nodes; activation is further increased with loss of peroxisome proliferator-activated receptor gamma gene and altered lipid profiles. The activated iNKT cells migrate into the liver, where they promote hepatocyte apoptosis. Patients with alcohol use disorder have increased expression of CD1d in the small intestine. Strategies to block these processes might be developed to treat alcoholic liver disease.


Assuntos
Enterócitos , Etanol/farmacologia , Hepatócitos , Hepatopatias Alcoólicas , Células T Matadoras Naturais , Animais , Antígenos CD1d/metabolismo , Apoptose , Ensaios de Migração de Leucócitos/métodos , Movimento Celular , Depressores do Sistema Nervoso Central/farmacologia , Enterócitos/efeitos dos fármacos , Enterócitos/imunologia , Enterócitos/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatopatias Alcoólicas/metabolismo , Hepatopatias Alcoólicas/patologia , Ativação Linfocitária , Camundongos , Células T Matadoras Naturais/efeitos dos fármacos , Células T Matadoras Naturais/metabolismo
5.
Immunogenetics ; 68(8): 665-76, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27405300

RESUMO

Type II natural killer T cells (NKT) are a subset of the innate-like CD1d-restricted lymphocytes that are reactive to lipid antigens. Unlike the type I NKT cells, which express a semi-invariant TCR, type II NKT cells express a broader TCR repertoire. Additionally, other features, such as their predominance over type I cells in humans versus mice, the nature of their ligands, CD1d/lipid/TCR binding, and modulation of immune responses, distinguish type II NKT cells from type I NKT cells. Interestingly, it is the self-lipid-reactivity of type II NKT cells that has helped define their physiological role in health and in disease. The discovery of sulfatide as one of the major antigens for CD1d-restricted type II NKT cells in mice has been instrumental in the characterization of these cells, including the TCR repertoire, the crystal structure of the CD1d/lipid/TCR complex, and their function. Subsequently, several other glycolipids and phospholipids from both endogenous and microbial sources have been shown to activate type II NKT cells. The activation of a specific subset of type II NKT cells following administration with sulfatide or lysophosphatidylcholine (LPC) leads to engagement of a dominant immunoregulatory pathway associated with the inactivation of type I NKT cells, conventional dendritic cells, and inhibition of the proinflammatory Th1/Th17 cells. Thus, type II NKT cells have been shown to be immunosuppressive in autoimmune diseases, inflammatory liver diseases, and in cancer. Knowing their relatively higher prevalence in human than type I NKT cells, understanding their biology is imperative for health and disease.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD1d/imunologia , Células T Matadoras Naturais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Humanos , Camundongos
6.
Haematologica ; 98(10): 1650-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23716558

RESUMO

Induction of heme oxygenase-1, a stress-inducible enzyme with anti-inflammatory activity, reduces the immunogenicity of therapeutic factor VIII in experimental hemophilia A. In humans, heme oxygenase-1 expression is modulated by polymorphisms in the promoter of the heme oxygenase-1-encoding gene (HMOX1). We investigated the relationship between polymorphisms in the HMOX1 promoter and factor VIII inhibitor development in severe hemophilia A. We performed a case-control study on 99 inhibitor-positive patients and 263 patients who did not develop inhibitors within the first 150 cumulative days of exposure to therapeutic factor VIII. Direct sequencing and DNA fragment analysis were used to study (GT)n polymorphism and single nucleotide polymorphisms located at -1135 and -413 in the promoter of HMOX1. We assessed associations between the individual allele frequencies or genotypes, and inhibitor development. Our results demonstrate that inhibitor-positive patients had a higher frequency of alleles with large (GT)n repeats (L: n≥30), which are associated with lesser heme oxygenase-1 expression (odds ratio 2.31; 95% confidence interval 1.46-3.66; P<0.001]. Six genotypes (L/L, L/M, L/S, M/M, M/S and S/S) of (GT)n repeats were identified (S: n<21; M: 21≤n<30). The genotype group including L alleles (L/L, L/M and L/S) was statistically more frequent among inhibitor-positive than inhibitor-negative patients, as compared to the other genotypes (33.3% versus 17.1%) (odds ratio 2.21, 95% confidence interval 1.30-3.76; P<0.01). To our knowledge, this is the first association identified between HMOX1 promoter polymorphism and development of anti-drug antibodies. Our study paves the way towards modulation of the endogenous anti-inflammatory machinery of hemophilia patients to reduce the risk of inhibitor development.


Assuntos
Fator VIII/uso terapêutico , Heme Oxigenase-1/genética , Hemofilia A/genética , Repetições de Microssatélites/genética , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Anticorpos/sangue , Estudos de Casos e Controles , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Humanos , Índice de Gravidade de Doença
7.
Blood ; 115(13): 2682-5, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19890094

RESUMO

Replacement therapy with exogenous factor VIII (FVIII) to treat hemorrhages induces anti-FVIII inhibitory immunoglobulin G in up to 30% of patients with hemophilia A. Chronic inflammation associated with recurrent bleedings is a proposed risk factor for FVIII inhibitor development. Heme oxygenase-1 (HO-1) is a stress-inducible enzyme with potent anti-inflammatory activity. Here, we demonstrate that induction of HO-1 before FVIII administration drastically reduces the onset of the anti-FVIII humoral immune response. The protective effect was specific for HO-1 because it was reproduced on administration of the end products of HO-1 activity, carbon monoxide, and bilirubin, and prevented by the pharmacologic inhibition of HO-1 using tin mesoporphyrin IX. HO-1 induction was associated with decreased major histocompatibility complex class II expression by splenic antigen-presenting cells and reduced T-cell proliferation. Triggering the endogenous anti-inflammatory machinery before FVIII administration may represent a novel therapeutic option for preventing the development of FVIII inhibitors in hemophilia A patients.


Assuntos
Fator VIII/uso terapêutico , Heme Oxigenase-1/fisiologia , Hemina/administração & dosagem , Hemofilia A/imunologia , Imunoglobulina G/biossíntese , Isoanticorpos/biossíntese , Proteínas de Membrana/fisiologia , Animais , Células Apresentadoras de Antígenos/imunologia , Esquema de Medicação , Fator VIII/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Hemina/farmacologia , Hemina/uso terapêutico , Hemofilia A/tratamento farmacológico , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Imunoglobulina G/imunologia , Inflamação , Isoanticorpos/imunologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Metaloporfirinas/farmacologia , Camundongos , Camundongos Knockout , Baço/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Fatores de Tempo
8.
J Immunol ; 185(7): 4101-8, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20817872

RESUMO

The importance of gut commensal bacteria in maintaining immune homeostasis is increasingly understood. We recently described that alteration of the gut microflora can affect a population of Foxp3(+)T(reg) cells that regulate demyelination in experimental autoimmune encephalomyelitis (EAE), the experimental model of human multiple sclerosis. We now extend our previous observations on the role of commensal bacteria in CNS demyelination, and we demonstrate that Bacteroides fragilis producing a bacterial capsular polysaccharide Ag can protect against EAE. Recolonization with wild type B. fragilis maintained resistance to EAE, whereas reconstitution with polysaccharide A-deficient B. fragilis restored EAE susceptibility. Enhanced numbers of Foxp3(+)T(reg) cells in the cervical lymph nodes were observed after intestinal recolonization with either strain of B. fragilis. Ex vivo, CD4(+)T cells obtained from mice reconstituted with wild type B. fragilis had significantly enhanced rates of conversion into IL-10-producing Foxp3(+)T(reg) cells and offered greater protection against disease. Our results suggest an important role for commensal bacterial Ags, in particular B. fragilis expressing polysaccharide A, in protecting against CNS demyelination in EAE and perhaps human multiple sclerosis.


Assuntos
Antígenos de Bactérias/imunologia , Cápsulas Bacterianas/imunologia , Bacteroides fragilis/imunologia , Encefalomielite Autoimune Experimental/imunologia , Mucosa Intestinal/microbiologia , Animais , Separação Celular , Encefalomielite Autoimune Experimental/microbiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Immunohorizons ; 5(12): 953-971, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34911745

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Homeostase/imunologia , Intestino Grosso/imunologia , Transferência Adotiva , Animais , Antígenos CD , Antígenos CD8 , Feminino , Cadeias alfa de Integrinas , Intestino Grosso/metabolismo , Mamíferos/imunologia , Mamíferos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T alfa-beta , Linfócitos T Reguladores/imunologia
10.
Blood ; 112(2): 240-9, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18469198

RESUMO

Procoagulant factor VIII (FVIII) is either produced endogenously under physiologic conditions, or administered exogenously as a therapeutic hemostatic drug in patients with hemophilia A. In the circulation, FVIII interacts with a multitude of glycoproteins, and may be used for coagulation at the sites of bleeding, eliminated by scavenger cells, or processed by the immune system, either as a self-constituent or as a foreign antigen. The fate of FVIII is dictated by the immune status of the individual, the location of FVIII in the body at a given time point, and the inflammatory microenvironment. It also depends on the local concentration of FVIII and of each interacting partner, and on the affinity of the respective interactions. FVIII, by virtue of its promiscuity, thus constitutes the core of a dynamic network that links the coagulation cascade, cells of the immune system, and, presumably, the inflammatory compartment. We describe the different interactions that FVIII is prone to establish during its life cycle, with a special focus on players of the innate and adaptive immune response. Lessons can be learned from understanding the dynamics of FVIII interactions--lessons that should pave the way to the conception of long-lasting hemostatic drugs devoid of iatrogenic immunogenicity.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Anticorpos , Fator VIII/metabolismo , Humanos , Sistema Imunitário/citologia , Tolerância Imunológica
11.
J Immunol ; 181(3): 1609-15, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18641295

RESUMO

Alloimmunization is a crippling concern in the management of patients undergoing administration of protein therapeutics as evidenced in replacement therapy and other treatment procedures. Several issues in the genesis and modulation of such deleterious immune responses have been studied. While authors have focused on the downstream events of the specific immune response and suggested modification of protein therapeutics to eliminate epitopes that interact with B cell receptors, T cell receptors, or MHCII molecules, the mechanisms underlying Ag interaction with APCs, a step upstream of immune effectors, have been grossly neglected. We wish to emphasize that the recent knowledge in understanding the capacities of an APC to handle an Ag and the importance of the surrounding microenvironment in this process are crucial for designing novel protein therapeutics with reduced immunogenicity.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Proteínas/imunologia , Animais , Endocitose/imunologia , Humanos , Imunogenética , Inflamação/imunologia , Inflamação/metabolismo
12.
Front Immunol ; 11: 393, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32273875

RESUMO

The development of an immune response against therapeutic factor VIII is the major complication in hemophilia A patients. Oligomannose carbohydrates at N239 and/or N2118 on factor VIII allow its binding to the macrophage mannose receptor expressed on human dendritic cells, thereby leading to factor VIII endocytosis and presentation to CD4+ T lymphocytes. Here, we investigated whether altering the interaction of factor VIII with mannose-sensitive receptors on antigen-presenting cells may be a strategy to reduce factor VIII immunogenicity. Gene transfer experiments in factor VIII-deficient mice indicated that N239Q and/or N2118Q factor VIII mutants have similar specific activities as compared to non-mutated factor VIII; N239Q/N2118Q mutant corrected blood loss upon tail clip. Production of the corresponding recombinant FVIII mutants or light chains indicated that removal of the N-linked glycosylation site at N2118 is sufficient to abrogate in vitro the activation of FVIII-specific CD4+ T cells by human monocyte-derived dendritic cells. However, removal of mannose-ending glycans at N2118 did not alter factor VIII endocytosis and presentation to CD4+ T cells by mouse antigen-presenting cells. In agreement with this, the N2118Q mutation did not reduce factor VIII immunogenicity in factor VIII-deficient mice. Our results highlight differences in the endocytic pathways between human and mouse dendritic cell subsets, and dissimilarities in tissue distribution and function of endocytic receptors such as CD206 in both species. Further investigations in preclinical models of hemophilia A closer to humans are needed to decipher the exact role of mannose-ending glycans in factor VIII immunogenicity.


Assuntos
Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Fator VIII/imunologia , Ativação Linfocitária/imunologia , Animais , Fator VIII/química , Fator VIII/genética , Humanos , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Manose/química , Manose/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/imunologia , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Mutação , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo
13.
Inflamm Bowel Dis ; 26(10): 1498-1508, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32840322

RESUMO

BACKGROUND: Currently, 2 coprimary end points are used by health authorities to determine the effectiveness of therapeutic interventions in patients with Crohn's disease (CD): symptomatic remission (patient-reported outcome assessment) and endoscopic remission (ileocolonoscopy). However, there is lack of accepted biomarkers to facilitate regulatory decision-making in the development of novel therapeutics for the treatment of CD. METHODS: With support from the Helmsley Charitable Trust, Critical Path Institute formed the Crohn's Disease Biomarkers preconsortium (CDBpC) with members from the pharmaceutical industry, academia, and nonprofit organizations to evaluate the CD biomarker landscape. Biomarkers were evaluated based on biological relevance, availability of biomarker assays, and clinical validation data. RESULTS: The CDBpC identified the most critical need as pharmacodynamic/response biomarkers to monitor disease activity in response to therapeutic intervention. Fecal calprotectin (FC) and serum C-reactive protein (CRP) were identified as biomarkers ready for the regulatory qualification process. A number of exploratory biomarkers and potential panels of these biomarkers was also identified for additional development. Given the different factors involved in CD and disease progression, a combination of biomarkers, including inflammatory, tissue injury, genetic, and microbiome-associated biomarkers, will likely have the most utility. CONCLUSIONS: The primary focus of the Inflammatory Bowel Disease Regulatory Science Consortium will be development of exploratory biomarkers and the qualification of FC and CRP for IBD. The Inflammatory Bowel Disease Regulatory Science Consortium, focused on tools to support IBD drug development, will operate in the precompetitive space to share data, biological samples for biomarker testing, and assay information for novel biomarkers.


Assuntos
Proteína C-Reativa/análise , Tomada de Decisão Clínica/métodos , Doença de Crohn/diagnóstico , Monitoramento de Medicamentos/métodos , Complexo Antígeno L1 Leucocitário/análise , Biomarcadores/análise , Consenso , Doença de Crohn/metabolismo , Doença de Crohn/terapia , Descoberta de Drogas , Fezes/química , Humanos , Reprodutibilidade dos Testes , Índice de Gravidade de Doença
14.
Haematologica ; 93(1): 83-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18166789

RESUMO

BACKGROUND: The development of factor VIII (FVIII) inhibitors remains the major hurdle in the clinical management of patients with hemophilia A. FVIII uptake by professional antigen-presenting cells (APC) is the first step involved in initiation of immune responses to FVIII. Studies on FVIII catabolism have highlighted the role played by CD91/LRP as a potential target for increasing FVIII half-life in patients and prolonging treatment efficiency. We investigated the involvement of CD91 in FVIII endocytosis by human dendritic cells (DC), a model of professional APC. DESIGN AND METHODS: Immature DC were generated from circulating monocytes from healthy donors. Surface expression of CD91 was assessed by flow cytometry. Uptake of fluorescein isothiocyanate-conjugated ligands by immature DC was studied in the presence of various blocking agents. RESULTS: CD91 was expressed on approximately 20% of DC and mediated the internalization of its model ligand, alpha2-macroglobulin. DC internalized FVIII and activated a human FVIII-specific T-cell clone in a dose-dependent manner. FVIII uptake by DC and subsequent T-cell activation were not inhibited by receptor-associated protein. CONCLUSIONS: Our results indicate that CD91 and other members of the LDL receptor family are not strongly implicated in FVIII internalization by monocyte-derived DC, and suggest the involvement of alternative divalent ion-dependent endocytic receptors.


Assuntos
Antígenos CD/biossíntese , Células Dendríticas/citologia , Fator VIII/biossíntese , Linfócitos T/citologia , Animais , Separação Celular , Endocitose , Fator VIII/metabolismo , Hemofilia A/genética , Humanos , Leucócitos Mononucleares/citologia , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Ativação Linfocitária , Camundongos , Monócitos/citologia , Monócitos/metabolismo
15.
Thromb Res ; 122 Suppl 2: S3-6, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18549909

RESUMO

Up to 33% of patients with severe haemophilia A develop inhibitory antibodies to factor VIM (FVIII) that can significantly impair treatment with FVIII. The plasma protein von Willebrand factor (VWF) binds to FVIII and is known to be important for the functioning of FVIII. Accumulating data suggest that VWF may also be important for reducing the immunogenicity of therapeutically administered FVIII in patients with haemophilia A. Although contradictory results have been reported for studies in patients, studies in mice have shown that the levels of FVIII-binding antibodies induced following treatment with recombinant FVIII (rFVIII) are higher than those following treatment with plasma-derived FVIII preparations containing VWF, and that the addition of VWF to rFVIII reduces the levels of FVIII-binding antibodies induced. In in vitro studies, VWF has been shown to inhibit both the uptake of FVIII by immature dendritic cells and the activation of FVIII-specificT-cells in a dose-dependent manner. However, recombinant VWF (rVWF) lacking the FVIII-binding domain did not inhibit T-cell activation. These data suggest that VWF may reduce the immunogenicity of FVIII by inhibiting the uptake of FVIII by antigen presenting cells, the first step in the development of an immune response against a foreign antigen. Further studies are required to confirm the applicability of these results to patients with haemophilia. If confirmed, these data would encourage the use of VWF in conjunction with FVIII in the management of patients with haemophilia A.


Assuntos
Fator VIII/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Fator de von Willebrand/imunologia , Apresentação de Antígeno/imunologia , Humanos
17.
Immunol Lett ; 110(1): 23-8, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17467813

RESUMO

A number of diseases are treated by passive administration of human proteins. Human coagulation factor VIII (FVIII) is one such protein which is administered to hemophilia A patients in order to manage and treat hemorrhagic incidences. This mode of therapy suffers from the side effect of generating anti-FVIII antibodies (inhibitors) which neutralizes the function of the infused FVIII. At a time when efficient viral screening procedures are at place, development of inhibitors poses the greatest threat to such a therapy. Various predisposing factors, both patient and product-related, are responsible for the development of inhibitory antibodies. A proper understanding of these "risk-factors" would eventually help to better design therapeutic regimen to tackle hemophilia A.


Assuntos
Anticorpos/sangue , Fator VIII/imunologia , Hemofilia A/tratamento farmacológico , Animais , Anticorpos/imunologia , Fator VIII/efeitos adversos , Fator VIII/uso terapêutico , Hemofilia A/imunologia , Humanos , Fatores de Risco , Resultado do Tratamento
18.
Haematologica ; 92(10): 1423-6, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18024377

RESUMO

Von Willebrand factor (VWF) has been proposed to reduce the immunogenicity of therapeutic factor VIII (FVIII) in patients with hemophilia A. Using FVIII-deficient mice, we compared the immunogenicity of different preparations of plasma-derived (pd) and recombinant (r) FVIII. Treatment of mice with pdFVIII induced significantly lower titers of FVIII inhibitors, as measured by ELISA and in vitro coagulation assays, compared with rFVIII. Furthermore, pre-incubation of rFVIII with excess VWF significantly reduced rFVIII immunogenicity. Our data confirm that pdFVIII induces lower levels of inhibitors than rFVIII, and that VWF is an immuno-chaperone molecule for FVIII.


Assuntos
Modelos Animais de Doenças , Fator VIII/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Animais , Fator VIII/genética , Feminino , Hemofilia A/genética , Humanos , Imunoglobulina G/imunologia , Masculino , Camundongos
19.
PLoS One ; 12(11): e0188600, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29190678

RESUMO

Type I interferons (IFNα, IFNß) are key regulators of innate and adaptive immunity, modulating the severity of both viral and bacterial infections. While type I IFN signaling leads to improved outcomes in viral infections, its role in bacterial infections is more contextual and depends on the specific pathogen and route of infection. Given the limited evidence on whether type I IFN signaling affects enteric bacterial pathogens, we investigated the role of this signaling pathway in Salmonella enterica serovar Typhimurium (S. typhimurium)-induced colitis. Comparing mice deficient in IFNAR1- the common receptor for IFNα and IFNß- with wild-type mice, we found that type I IFN signaling leads to more rapid death, more severe colonic inflammation, higher serum levels of pro-inflammatory cytokines, and greater bacterial dissemination. Specific ablation of plasmacytoid dendritic cells (pDCs), which are prominent producers of type I IFNs in antiviral responses, did not alter survival after infection. This result established that pDCs do not play a major role in the pathogenesis of S. typhimurium colitis. Flow cytometric analysis of macrophages and conventional dendritic cells (cDCs) during active colitis demonstrated an increase in CD11c- macrophages and CD103+ cDCs in the colon of Ifnar1-/- animals. Interestingly, cells expressing the anti-inflammatory cytokine receptor IL-10R are more abundant within these subsets in Ifnar1-/- than in wild-type mice. Moreover, blockade of IL-10R in Ifnar1-/- mice increased their susceptibility to S. typhimurium colitis, suggesting that altered numbers of these immunoregulatory cells may underlie the difference in disease severity. This cross-talk between type I IFN and IL-10R signaling pathways may represent a key host cellular mechanism to investigate further in order to unravel the balance between pathogenic inflammation and homeostasis of the colon. Taken together, our data clearly demonstrate that type I IFN signaling is pathogenic in S. typhimurium colitis.


Assuntos
Colite/patologia , Células Dendríticas/metabolismo , Interferon Tipo I/metabolismo , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Receptores de Interleucina-10/metabolismo , Infecções por Salmonella/metabolismo , Transdução de Sinais , Animais , Colite/metabolismo , Colite/microbiologia , Citocinas/metabolismo , Mucosa Intestinal/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Índice de Gravidade de Doença
20.
Immunol Lett ; 103(1): 3-7, 2006 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-16427704

RESUMO

Immunoglobulins have initially been illustrated as proteins produced by the immune system for binding and neutralizing foreign molecules potentially harmful to the organism. The number of V(H), D(H), J(H), V(L) and J(L) genes that encode the variable regions of immunoglobulins and the junctional diversity that occurs at the time of somatic rearrangement determine the extent of the repertoire of antibodies that may be potentially produced by an organism. This potential repertoire includes antibodies the antigen binding site of which may recognize external as well as autologous antigens, or may structurally resemble the active site of enzymes and be endowed with enzymatic activity. Under physiological conditions, B cell clones that produce antibodies naturally endowed with catalytic activity are negatively regulated and subjected to apoptosis. Catalytic antibodies are expressed only following active immunization, or if the physiological regulatory mechanisms that control the expression of catalytic antibody-producing B cell clones are perturbed, e.g. in the context of pregnancy or in the course of autoimmune diseases.


Assuntos
Anticorpos Catalíticos , Doenças Autoimunes/imunologia , Doenças Autoimunes/fisiopatologia , Doença de Hashimoto/imunologia , Doença de Hashimoto/fisiopatologia , Humanos , Imunização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA