Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Eur J Immunol ; 53(11): e2250236, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37673213

RESUMEN

Multiple sclerosis (MS) is a chronic and progressive autoimmune disease of the central nervous system (CNS), with both genetic and environmental factors contributing to the pathobiology of the disease. Although HLA genes have emerged as the strongest genetic factor linked to MS, consensus on the environmental risk factors is lacking. Recently, the gut microbiota has garnered increasing attention as a potential environmental factor in MS, as mounting evidence suggests that individuals with MS exhibit microbial dysbiosis (changes in the gut microbiome). Thus, there has been a strong emphasis on understanding the role of the gut microbiome in the pathobiology of MS, specifically, factors regulating the gut microbiota and the mechanism(s) through which gut microbes may contribute to MS. Among all factors, diet has emerged to have the strongest influence on the composition and function of gut microbiota. As MS patients lack gut bacteria capable of metabolizing dietary phytoestrogen, we will specifically discuss the role of a phytoestrogen diet and phytoestrogen metabolizing gut bacteria in the pathobiology of MS. A better understanding of these mechanisms will help to harness the enormous potential of the gut microbiota as potential therapeutics to treat MS and other autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Microbiota , Esclerosis Múltiple , Humanos , Fitoestrógenos , Bacterias , Dieta , Disbiosis
2.
Cells ; 12(18)2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37759490

RESUMEN

Preclinical studies have shown that chronic alcohol abuse leads to alterations in the gastrointestinal microbiota that are associated with behavior changes, physiological alterations, and immunological effects. However, such studies have been limited in their ability to evaluate the direct effects of alcohol-associated dysbiosis. To address this, we developed a humanized alcohol-microbiota mouse model to systematically evaluate the immunological effects of chronic alcohol abuse mediated by intestinal dysbiosis. Germ-free mice were colonized with human fecal microbiota from individuals with high and low Alcohol Use Disorders Identification Test (AUDIT) scores and bred to produce human alcohol-associated microbiota or human control-microbiota F1 progenies. F1 offspring colonized with fecal microbiota from individuals with high AUDIT scores had increased susceptibility to Klebsiella pneumoniae and Streptococcus pneumoniae pneumonia, as determined by increased mortality rates, pulmonary bacterial burden, and post-infection lung damage. These findings highlight the importance of considering both the direct effects of alcohol and alcohol-induced dysbiosis when investigating the mechanisms behind alcohol-related disorders and treatment strategies.


Asunto(s)
Alcoholismo , Microbiota , Neumonía Bacteriana , Humanos , Animales , Ratones , Alcoholismo/complicaciones , Disbiosis/complicaciones , Etanol
3.
Sci Adv ; 9(19): eadf5499, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37184968

RESUMEN

Mammalian species harbor compositionally distinct gut microbial communities, but the mechanisms that maintain specificity of symbionts to host species remain unclear. Here, we show that natural selection within house mice (Mus musculus domesticus) drives deterministic assembly of the house-mouse gut microbiota from mixtures of native and non-native microbiotas. Competing microbiotas from wild-derived lines of house mice and other mouse species (Mus and Peromyscus spp.) within germ-free wild-type (WT) and Rag1-knockout (Rag1-/-) house mice revealed widespread fitness advantages for native gut bacteria. Native bacterial lineages significantly outcompeted non-native lineages in both WT and Rag1-/- mice, indicating home-site advantage for native microbiota independent of host adaptive immunity. However, a minority of native Bacteriodetes and Firmicutes favored by selection in WT hosts were not favored or disfavored in Rag1-/- hosts, indicating that Rag1 mediates fitness advantages of these strains. This study demonstrates home-site advantage for native gut bacteria, consistent with local adaptation of gut microbiota to their mammalian species.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Ratones , Bacterias , Proteínas de Homeodominio/genética , Mamíferos
4.
Pathogens ; 12(5)2023 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37242309

RESUMEN

Intestinal dysbiosis increases susceptibility to infection through the alteration of metabolic profiles, which increases morbidity. Zinc (Zn) homeostasis in mammals is tightly regulated by 24 Zn transporters. ZIP8 is unique in that it is required by myeloid cells to maintain proper host defense against bacterial pneumonia. In addition, a frequently occurring ZIP8 defective variant (SLC39A8 rs13107325) is strongly associated with inflammation-based disorders and bacterial infection. In this study, we developed a novel model to study the effects of ZIP8-mediated intestinal dysbiosis on pulmonary host defense independent of the genetic effects. Cecal microbial communities from a myeloid-specific Zip8 knockout mouse model were transplanted into germ-free mice. Conventionalized ZIP8KO-microbiota mice were then bred to produce F1 and F2 generations of ZIP8KO-microbiota mice. F1 ZIP8KO-microbiota mice were also infected with S. pneumoniae, and pulmonary host defense was assessed. Strikingly, the instillation of pneumococcus into the lung of F1 ZIP8KO-microbiota mice resulted in a significant increase in weight loss, inflammation, and mortality when compared to F1 wild-type (WT)-microbiota recipients. Similar defects in pulmonary host defense were observed in both genders, although consistently greater in females. From these results, we conclude that myeloid Zn homeostasis is not only critical for myeloid function but also plays a significant role in the maintenance and control of gut microbiota composition. Further, these data demonstrate that the intestinal microbiota, independent of host genetics, play a critical role in governing host defense in the lung against infection. Finally, these data strongly support future microbiome-based interventional studies, given the high incidence of zinc deficiency and the rs13107325 allele in humans.

5.
Sci Rep ; 9(1): 10656, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31337812

RESUMEN

The Muc-1 oncoprotein is a tumor-associated mucin often overexpressed in pancreatic cancer. We report that knockout of Muc-1 reduced the degree of pancreatic inflammation that resulted from infection with Coxsackievirus B3 (CVB3) in a mouse model. CVB3-infected Muc-1-deficient (Muc-1KO) mice had significantly reduced infiltration of macrophages into the murine pancreas. We found that Muc-1 signaling through NF-κB increased expression of ICAM-1, a pro-inflammatory mediator that recruits macrophages. Further investigation revealed that bone marrow derived macrophages (BMDM) from the Muc-1KO mice exhibited defective migration properties, in part due to low expression of the C-C motif chemokine receptor (CCR2) and the integrin Very Late Antigen 4 (VLA-4). The results presented here provide novel insight into the role of Muc-1 in regulating the inflammatory response and the cellular microenvironment in pancreatitis.


Asunto(s)
Infecciones por Coxsackievirus/virología , Mucina-1/metabolismo , Pancreatitis/virología , Animales , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/metabolismo , Modelos Animales de Enfermedad , Enterovirus Humano B , Inflamación/genética , Inflamación/metabolismo , Inflamación/virología , Ratones , Ratones Noqueados , Mucina-1/genética , Pancreatitis/genética , Pancreatitis/metabolismo , Receptores CCR2/genética , Receptores CCR2/metabolismo
6.
EMBO Mol Med ; 8(6): 679-83, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27137491

RESUMEN

Biochemical experiments, animal models, and observational studies in humans all support a role of dipeptidyl peptidase 4 (DPP4) in the N-terminal truncation of CXCL10, which results in the generation of an antagonist form of the chemokine that limits T-cell and NK cell migration. Motivated by the ability to regulate lymphocyte trafficking in vivo, we conducted two prospective clinical trials to test the effects of DPP4 inhibition on CXCL10 processing in healthy donors and in chronic hepatitis C patients, a disease in which DPP4 levels are found to be elevated. Participants were treated daily with 100 mg sitagliptin, a clinically approved DPP4 inhibitor. Plasma samples were analyzed using an ultrasensitive single-molecule assay (Simoa) to distinguish the full-length CXCL101-77 from the NH2-truncated CXCL103-77, as compared to the total CXCL10 levels. Sitagliptin treatment resulted in a significant decrease in CXCL103-77 concentration, a reciprocal increase in CXCL101-77, with only minimal effects on total levels of the chemokine. These data provide the first direct evidence that in vivo DPP4 inhibition in humans can preserve the bioactive form of CXCL10, offering new therapeutic opportunities for DPP4 inhibitors.


Asunto(s)
Quimiocina CXCL10/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/administración & dosificación , Procesamiento Proteico-Postraduccional , Fosfato de Sitagliptina/administración & dosificación , Quimiocina CXCL10/sangre , Voluntarios Sanos , Hepatitis C Crónica/patología , Humanos , Placebos/administración & dosificación , Estudios Prospectivos , Proteolisis
7.
Front Immunol ; 6: 288, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26124756

RESUMEN

Dendritic cells (DCs) are key antigen-presenting cells that have an important role in autoimmune pathogenesis. DCs control both steady-state T cell tolerance and activation of pathogenic responses. The balance between these two outcomes depends on several factors, including genetic susceptibility, environmental signals that stimulate varied innate responses, and which DC subset is presenting antigen. Although the specific DC phenotype can diverge depending on the tissue location and context, there are four main subsets identified in both mouse and human: conventional cDC1 and cDC2, plasmacytoid DCs, and monocyte-derived DCs. In this review, we will discuss the role of these subsets in autoimmune pathogenesis and regulation, as well as the genetic and environmental signals that influence their function. Specific topics to be addressed include impact of susceptibility loci on DC subsets, alterations in DC subset development, the role of infection- and host-derived innate inflammatory signals, and the role of the intestinal microbiota on DC phenotype. The effects of these various signals on disease progression and the relative effects of DC subset composition and maturation level of DCs will be examined. These areas will be explored using examples from several autoimmune diseases but will focus mainly on type 1 diabetes.

8.
Diabetes ; 64(10): 3521-31, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26070317

RESUMEN

During autoimmunity, the normal ability of dendritic cells (DCs) to induce T-cell tolerance is disrupted; therefore, autoimmune disease therapies based on cell types and molecular pathways that elicit tolerance in the steady state may not be effective. To determine which DC subsets induce tolerance in the context of chronic autoimmunity, we used chimeric antibodies specific for DC inhibitory receptor 2 (DCIR2) or DEC-205 to target self-antigen to CD11b(+) (cDC2) DCs and CD8(+) (cDC1) DCs, respectively, in autoimmune-prone nonobese diabetic (NOD) mice. Antigen presentation by DCIR2(+) DCs but not DEC-205(+) DCs elicited tolerogenic CD4(+) T-cell responses in NOD mice. ß-Cell antigen delivered to DCIR2(+) DCs delayed diabetes induction and induced increased T-cell apoptosis without interferon-γ (IFN-γ) or sustained expansion of autoreactive CD4(+) T cells. These divergent responses were preceded by differential gene expression in T cells early after in vivo stimulation. Zbtb32 was higher in T cells stimulated with DCIR2(+) DCs, and overexpression of Zbtb32 in T cells inhibited diabetes development, T-cell expansion, and IFN-γ production. Therefore, we have identified DCIR2(+) DCs as capable of inducing antigen-specific tolerance in the face of ongoing autoimmunity and have also identified Zbtb32 as a suppressive transcription factor that controls T cell-mediated autoimmunity.


Asunto(s)
Anticuerpos , Antígenos CD/metabolismo , Autoinmunidad/fisiología , Linfocitos T CD4-Positivos/fisiología , Células Dendríticas/fisiología , Diabetes Mellitus/inmunología , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Represoras/metabolismo , Animales , Antígenos CD/genética , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/genética , Antígenos CD40/metabolismo , Diabetes Mellitus/prevención & control , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Lectinas Tipo C/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Antígenos de Histocompatibilidad Menor , Receptores de Superficie Celular/genética , Proteínas Represoras/genética , Organismos Libres de Patógenos Específicos
9.
J Leukoc Biol ; 95(2): 325-36, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24082013

RESUMEN

DCs are important mediators of peripheral tolerance for the prevention of autoimmunity. Chimeric αDEC-205 antibodies with attached antigens allow in vivo antigen-specific stimulation of T cells by CD8(+) DCs, resulting in tolerance in nonautoimmune mice. However, it is not clear whether DC-mediated tolerance induction occurs in the context of ongoing autoimmunity. We assessed the role of CD8(+) DCs in stimulation of autoreactive CD4(+) T cells in the NOD mouse model of type 1 diabetes. Targeting of antigen to CD8(+) DCs via αDEC-205 led to proliferation and expansion of ß-cell specific BDC2.5 T cells. These T cells also produced IL-2 and IFN-γ and did not up-regulate FoxP3, consistent with an activated rather than tolerant phenotype. Similarly, endogenous BDC peptide-reactive T cells, identified with I-A(g7) tetramers, did not become tolerant after antigen delivery via αDEC-205: no deletion or Treg induction was observed. We observed that CD8(+) DCs from NOD mice expressed higher surface levels of CD40 than CD8(+) DCs from C57BL/6 mice. Blockade of CD40-CD40L interactions reduced the number of BDC2.5 T cells remaining in mice, 10 days after antigen targeting to CD8 DCs, and blocked IFN-γ production by BDC2.5 T cells. These data indicate that the ability of autoreactive CD4(+) T cells to undergo tolerance mediated by CD8(+) DCs is defective in NOD mice and that blocking CD40-CD40L interactions can restore tolerance induction.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/antagonistas & inhibidores , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Animales , Anticuerpos/farmacología , Antígenos/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Factores de Transcripción Forkhead/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Interferón gamma/metabolismo , Ligandos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Péptidos/inmunología , Fenotipo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células TH1/efectos de los fármacos , Células TH1/metabolismo , Receptores Toll-Like/metabolismo
10.
Immunity ; 39(3): 560-72, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23993652

RESUMEN

The autoimmune regulator (Aire) is essential for prevention of autoimmunity; its role is best understood in the thymus, where it promotes self-tolerance through tissue-specific antigen (TSA) expression. Recently, extrathymic Aire-expressing cells (eTACs) have been described in murine secondary lymphoid organs, but the identity of such cells and their role in immune tolerance remains unclear. Here we have shown that eTACs are a discrete major histocompatibility complex class II (MHC II)(hi), CD80(lo), CD86(lo), epithelial cell adhesion molecule (EpCAM)(hi), CD45(lo) bone marrow-derived peripheral antigen-presenting cell (APC) population. We also have demonstrated that eTACs can functionally inactivate CD4⁺ T cells through a mechanism that does not require regulatory T cells (Treg) and is resistant to innate inflammatory stimuli. Together, these findings further define eTACs as a distinct tolerogenic cell population in secondary lymphoid organs.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Autotolerancia , Factores de Transcripción/metabolismo , Traslado Adoptivo , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/metabolismo , Autoinmunidad , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Células de la Médula Ósea , Moléculas de Adhesión Celular/metabolismo , Molécula de Adhesión Celular Epitelial , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos NOD , Factores de Transcripción/biosíntesis , Proteína AIRE
11.
Proc Natl Acad Sci U S A ; 108(6): 2408-13, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262836

RESUMEN

Steady-state development of plasmacytoid dendritic cells (pDCs) and conventional dendritic cells (cDCs) requires the ligand for FMS-like tyrosine kinase 3 receptor (flt3L), but little is known about how other cytokines may also control this process. In this study, we show that IL-2 inhibits the development of both pDCs and cDCs from bone marrow cells under flt3L stimulation, by acting on lineage(-) flt3(+) precursors. This inhibition of DC development by IL-2 requires IL-2Rα and IL2Rß. IL-2Rα is specifically expressed in one stage of the DC precursor: the monocyte and DC progenitors (MDPs). Furthermore, more MDPs are found in flt3L-stimulated bone marrow cultures when IL-2 is present, suggesting that IL-2 may be inhibiting DC development at the MDP stage. Consistent with our in vitro findings, we observe that nonobese diabetic (NOD) mice, which express less IL-2 compared with diabetes-resistant NOD.Idd3/5 mice, have more splenic pDCs. Additionally, DCs developed in vitro in the presence of flt3L and IL-2 display reduced ability to stimulate T-cell proliferation compared with DCs developed in the presence of flt3L alone. Although the addition of IL-2 does not increase the apoptosis of DCs during their development, DCs developed in the presence of IL-2 are more prone to apoptosis upon interaction with T cells. Together our data show that IL-2 can inhibit both the development and the function of DCs. This pathway may have implications for the loss of immune tolerance: Reduced IL-2 signaling may lead to increased DC number and T-cell stimulatory capacity.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Interleucina-2/inmunología , Proteínas de la Membrana/inmunología , Células Plasmáticas/inmunología , Transducción de Señal/inmunología , Animales , Apoptosis/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proliferación Celular , Células Dendríticas/citología , Células Dendríticas/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/inmunología , Subunidad beta del Receptor de Interleucina-2/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
12.
Infect Immun ; 78(12): 5295-306, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20921150

RESUMEN

Understanding the regulation of human immune responses is critical for vaccine development and treating infectious diseases. We have previously shown that simultaneous engagement of the T cell receptor (TCR) and complement regulator CD46 on human CD4(+) T cells in the presence of interleukin-2 (IL-2) induces potent secretion of the immunomodulatory cytokine IL-10. These T cells mediate IL-10-dependent suppression of bystander CD4(+) T cells activated in vitro with anti-CD3 and anti-CD28 costimulation, reflecting a T regulatory type 1 (Tr1)-like phenotype. However, CD46-mediated negative regulation of pathogen-specific T cells has not been described. Therefore, we studied the ability of CD46-activated human CD4(+) T cells to suppress T cell responses to Mycobacterium bovis BCG, the live vaccine that provides infants protection against the major human pathogen Mycobacterium tuberculosis. Our results demonstrate that soluble factors secreted by CD46-activated human CD4(+) T cells suppress mycobacterium-specific CD4(+), CD8(+), and γ(9)δ(2) TCR(+) T cells. Dendritic cell functions were not downregulated in our experiments, indicating that CD46-triggered factors directly suppress pathogen-specific T cells. Interestingly, IL-10 appeared to play a less pronounced role in our system, especially in the suppression of γ(9)δ(2) TCR(+) T cells, suggesting the presence of additional undiscovered soluble immunoregulatory factors. Blocking endogenous CD46 signaling 3 days after mycobacterial infection enhanced BCG-specific T cell responses in a subset of volunteers. Taken together, these results indicate that CD46-dependent negative regulatory mechanisms can impair T cell responses vital for immune defense against mycobacteria. Therefore, modulating CD46-induced immune regulation could be integral to the development of improved tuberculosis therapeutics or vaccines.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos/inmunología , Proteína Cofactora de Membrana/inmunología , Linfocitos T Reguladores/inmunología , Anticuerpos Monoclonales/inmunología , Vacuna BCG/inmunología , Células Cultivadas , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Hibridomas/inmunología , Interleucina-10/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología
13.
Blood ; 107(4): 1497-504, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16239430

RESUMEN

Concurrent activation of the T-cell receptor (TCR) and complement regulator CD46 on human CD4+ T lymphocytes induces Tr1-like regulatory T cells that suppress through IL-10 secretion bystander T-cell proliferation. Here we show that, despite their IL-10 production, CD46-induced T-regulatory T cells (Tregs) do not suppress the activation/maturation of dendritic cells (DCs). DC maturation by complement/CD46-induced Tregs is mediated through simultaneous secretion of GM-CSF and soluble CD40L, factors favoring DC differentiation and reversing inhibitory effects of IL-10. Thus, CD46-induced Tregs produce a distinct cytokine profile that inhibits T-cell responses but leaves DC activation unimpaired. Such "DC-sparing" Tregs could be desirable at host/environment interfaces such as the gastrointestinal tract where their specific cytokine profile provides a mechanism that ensures unresponsiveness to commensal bacteria while maintaining reactivity to invading pathogens.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Células Dendríticas/inmunología , Linfocitos T/inmunología , Antígenos CD/inmunología , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/inmunología , Humanos , Interleucina-10/inmunología , Activación de Linfocitos , Monocitos/inmunología , Valores de Referencia
14.
Springer Semin Immunopathol ; 27(3): 345-58, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16200405

RESUMEN

In the past 20 years, our understanding of the workings of complement regulatory protein, CD46 (membrane cofactor protein), has grown as has the impressive list of pathogens interacting with this membrane-bound complement inhibitor. Referred to as a "pathogen magnet," CD46 serves as a receptor for seven human pathogens. Initially discovered as a widely expressed C3b- and C4b-binding protein, it was subsequently shown to be a cofactor for the serine protease factor I to inactivate by limited proteolysis these two opsonins and components of the convertases. The involvement of CD46 in reproductive processes continues to be an emerging story. It is a protector of placental tissue, but it may also play a more direct role in reproduction through its expression on the inner acrosomal membrane of spermatozoa. Cross-linking CD46 with antibodies or natural or pathogenic ligands induces rapid turnover and signaling events. In this regard, much attention is currently focused on generating human T lymphocyte regulatory cells by cross-linking CD46. Finally, highlighting its importance in protecting cells against excessive complement activation is the discovery that even a heterozygous deficiency of CD46 predisposes to hemolytic uremic syndrome.


Asunto(s)
Proteína Cofactora de Membrana/inmunología , Secuencia de Aminoácidos , Animales , Activación de Complemento , Femenino , Expresión Génica , Humanos , Masculino , Proteína Cofactora de Membrana/química , Proteína Cofactora de Membrana/deficiencia , Proteína Cofactora de Membrana/genética , Modelos Moleculares , Datos de Secuencia Molecular , Embarazo , Receptores Virales/inmunología , Reproducción/inmunología , Linfocitos T Reguladores/inmunología
15.
Immunol Res ; 32(1-3): 31-43, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16106057

RESUMEN

Crosslinking of CD46 and CD3 on naïve human CD4+ T-lymphocytes induces interleukin-10 secretion and granzyme B expression. These highly proliferative T-regulatory type 1-like T-regulatory T-cells (Tregs) can suppress an immune response. We propose that this process is important in the prevention of chronic inflammation such as at epithelial borders and in deactivation of a successful immune response. Relative to the latter, once a complement-fixing polyclonal antibody response has been mounted, in most cases, the pathogen will be rapidly destroyed. At this time, the C3b/C4b-bearing immune complexes could initiate the deactivation arm of an immune response by shutting down immunocompetent cells through CD46-generated T-cells. Herein, we review this pathway for the induction of Tregs, focusing on a role for the complement system and especially signaling through CD46 on human T-cells.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Proteína Cofactora de Membrana/metabolismo , Linfocitos T Reguladores/inmunología , Secuencia de Aminoácidos , Animales , Granzimas , Humanos , Técnicas In Vitro , Interleucina-10/biosíntesis , Activación de Linfocitos , Masculino , Proteína Cofactora de Membrana/química , Proteína Cofactora de Membrana/genética , Ratones , Ratones Transgénicos , Modelos Inmunológicos , Datos de Secuencia Molecular , Serina Endopeptidasas/biosíntesis , Transducción de Señal/inmunología
16.
J Immunol ; 175(2): 677-84, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16002662

RESUMEN

Regulatory T cells (Tregs) participate in the control of the immune response. In the human system, an IL-10-secreting, T regulatory type 1 cell (Tr1)-like subset of Tregs can be induced by concurrent cross-linking of the TCR and CD46 on naive CD4(+) T cells. Because many viral and bacterial pathogens, including the major human pathogen Streptococcus pyogenes, bind to CD46, we asked whether this bacterium can directly induce Tr1-like cells through the streptococcal ligand for CD46, the M protein. The M5 and M22 proteins were found to induce T cells to develop into the IL-10-producing Tr1-like phenotype. Moreover, whole M5-expressing bacteria, but not isogenic M-negative bacteria, led to proliferation and IL-10 secretion by T cells. The interaction between the M5 protein and T cells was dependent on CD46 and the conserved C repeat region of M5. Supernatants derived from T cells stimulated with M proteins or M protein-expressing bacteria suppressed bystander T cell proliferation through IL-10 secretion. In addition, activation of CD46 through streptococcal M protein induced the expression of granzyme B, providing a second means for these cells to regulate an immune response. These findings suggest that binding to CD46 and exploiting its signaling pathway may represent a strategy employed by a number of important human pathogens to induce directly an immunosuppressive/regulatory phenotype in T cells.


Asunto(s)
Antígenos Bacterianos/fisiología , Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas Portadoras/fisiología , Inmunofenotipificación , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Antígenos CD/fisiología , Efecto Espectador/inmunología , Proliferación Celular , Sistema Libre de Células/inmunología , Citocinas/biosíntesis , Pruebas Inmunológicas de Citotoxicidad , Granzimas , Humanos , Activación de Linfocitos/inmunología , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/fisiología , Serina Endopeptidasas/biosíntesis , Streptococcus pyogenes/inmunología , Linfocitos T Colaboradores-Inductores/microbiología , Linfocitos T Reguladores/microbiología , Células TH1/citología , Células TH1/enzimología , Células TH1/inmunología , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA