Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(26): e2221744120, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37339214

RESUMO

Functional molecular characterization of the cochlea has mainly been driven by the deciphering of the genetic architecture of sensorineural deafness. As a result, the search for curative treatments, which are sorely lacking in the hearing field, has become a potentially achievable objective, particularly via cochlear gene and cell therapies. To this end, a complete inventory of cochlear cell types, with an in-depth characterization of their gene expression profiles right up to their final differentiation, is indispensable. We therefore generated a single-cell transcriptomic atlas of the mouse cochlea based on an analysis of more than 120,000 cells on postnatal day 8 (P8), during the prehearing period, P12, corresponding to hearing onset, and P20, when cochlear maturation is almost complete. By combining whole-cell and nuclear transcript analyses with extensive in situ RNA hybridization assays, we characterized the transcriptomic signatures covering nearly all cochlear cell types and developed cell type-specific markers. Three cell types were discovered; two of them contribute to the modiolus which houses the primary auditory neurons and blood vessels, and the third one consists in cells lining the scala vestibuli. The results also shed light on the molecular basis of the tonotopic gradient of the biophysical characteristics of the basilar membrane that critically underlies cochlear passive sound frequency analysis. Finally, overlooked expression of deafness genes in several cochlear cell types was also unveiled. This atlas paves the way for the deciphering of the gene regulatory networks controlling cochlear cell differentiation and maturation, essential for the development of effective targeted treatments.


Assuntos
Surdez , Transcriptoma , Animais , Camundongos , Cóclea/fisiologia , Membrana Basilar , Audição/fisiologia , Surdez/metabolismo
2.
Ann N Y Acad Sci ; 1413(1): 59-68, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29377165

RESUMO

Autoimmune diseases (AIDs) are chronic disorders characterized by inflammatory reactions against self-antigens that can be either systemic or organ specific. AIDs can differ in their epidemiologic features and clinical presentations, yet all share a remarkable complexity. AIDs result from an interplay of genetic and epigenetic factors with environmental components that are associated with imbalances in the immune system. Many of the pathogenic mechanisms of AIDs are also implicated in myasthenia gravis (MG), an AID in which inflammation of the thymus leads to a neuromuscular disorder. Our goal here is to highlight the similarities and differences between MG and other AIDs by reviewing the common transcriptome signatures and the development of germinal centers and by discussing some unresolved questions about autoimmune mechanisms. This review will propose hypotheses to explain the origin of regulatory T (Treg ) cell defects and the causes of chronicity and specificity of AIDs.


Assuntos
Autoimunidade/genética , Linfócitos B/imunologia , Predisposição Genética para Doença/genética , Lúpus Eritematoso Sistêmico/genética , Miastenia Gravis/genética , Linfócitos T Reguladores/imunologia , Autoimunidade/imunologia , Centro Germinativo/imunologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Miastenia Gravis/imunologia , Miastenia Gravis/patologia , Sarcoidose/genética , Sarcoidose/imunologia
3.
Ann N Y Acad Sci ; 1412(1): 137-145, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29125185

RESUMO

It has long been established that the thymus plays a central role in autoimmune myasthenia gravis (MG) because of either thymoma or thymic hyperplasia of lymphoproliferative origin. In this review, we discuss thymic changes associated with thymic hyperplasia and their implications in the development of an autoimmune response against the acetylcholine receptor (AChR).The hyperplastic MG thymus displays all the characteristics of tertiary lymphoid organs (TLOs): neoangiogenic processes with high endothelial venule and lymphatic vessel development, chemokine overexpression favoring peripheral cell recruitment, and ectopic germinal center development. As thymic epithelial cells or myoid cells express AChR, a specific antigen presentation can easily occur within the thymus in the presence of recruited peripheral cells, such as B cells and T follicular helper cells. How the thymus turns into a TLO is not known, but local inflammation seems mandatory. Interferon (IFN)-ß is overexpressed in MG thymus and could orchestrate thymic changes associated with MG. Knowledge about how IFN-ß is induced in MG thymus and why its expression is sustained even long after disease onset would be of interest in the future to better understand the etiological and physiopathological mechanisms involved in autoimmune MG.


Assuntos
Miastenia Gravis/etiologia , Timo/imunologia , Adulto , Idade de Início , Quimiocinas/genética , Feminino , Centro Germinativo/imunologia , Centro Germinativo/patologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Interferon beta/imunologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Miastenia Gravis/imunologia , Miastenia Gravis/patologia , Neovascularização Patológica , Receptores Colinérgicos/imunologia , Linfócitos T/imunologia , Timo/irrigação sanguínea , Timo/patologia , Hiperplasia do Timo/complicações , Hiperplasia do Timo/imunologia , Hiperplasia do Timo/patologia , Receptores Toll-Like/genética , Regulação para Cima
4.
JCI Insight ; 2(7): e89665, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28405609

RESUMO

Myasthenia gravis (MG) with anti-acetylcholine receptor (AChR) Abs is an autoimmune disease characterized by severe defects in immune regulation and thymic inflammation. Because mesenchymal stem cells (MSCs) display immunomodulatory features, we investigated whether and how in vitro-preconditioned human MSCs (cMSCs) could treat MG disease. We developed a new humanized preclinical model by subcutaneously grafting thymic MG fragments into immunodeficient NSG mice (NSG-MG model). Ninety percent of the animals displayed human anti-AChR Abs in the serum, and 50% of the animals displayed MG-like symptoms that correlated with the loss of AChR at the muscle endplates. Interestingly, each mouse experiment recapitulated the MG features of each patient. We next demonstrated that cMSCs markedly improved MG, reducing the level of anti-AChR Abs in the serum and restoring AChR expression at the muscle endplate. Resting MSCs had a smaller effect. Finally, we showed that the underlying mechanisms involved (a) the inhibition of cell proliferation, (b) the inhibition of B cell-related and costimulatory molecules, and (c) the activation of the complement regulator DAF/CD55. In conclusion, this study shows that a preconditioning step promotes the therapeutic effects of MSCs via combined mechanisms, making cMSCs a promising strategy for treating MG and potentially other autoimmune diseases.


Assuntos
Linfócitos B/imunologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Miastenia Gravis Autoimune Experimental/terapia , Receptores Colinérgicos/imunologia , Adolescente , Adulto , Animais , Anticorpos Monoclonais/sangue , Criança , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Adulto Jovem
5.
Mol Ther ; 20(8): 1571-81, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22643865

RESUMO

The administration of recombinant adeno-associated viral vectors (rAAV) for gene transfer induces strong humoral responses through mechanisms that remain incompletely characterized. To investigate the links between innate and adaptive immune responses to the vector, rAAVs were injected intravenously into mice deficient in cell-intrinsic components of innate responses (Toll-like receptors (TLRs), type-1 interferon (IFN) or inflammasome signaling molecules) and AAV-specific antibodies were measured. Of all molecules tested, only MyD88 was critically needed to mount immunoglobulin G (IgG) responses since MyD88(-/-) mice failed to develop high levels of AAV-specific IgG2 and IgG3, regardless of capsid serotype injected. None of the TLRs tested was essential here, but TLR9 ensured a Th1-biased antibody responses. Indeed, capsid-specific Th1 cells were induced upon injection of rAAV1, as directly confirmed with an epitope-tagged capsid, and the priming and development of these Th1 cells required T cell-extrinsic MyD88. Cell transfer experiments showed that autonomous MyD88 signaling in B cells, but not T cells, was sufficient to produce Th1-dependent IgGs. Therefore, rAAV triggers innate responses, at least via B cells, controlling the development of capsid-specific Th1-driven antibodies. MyD88 emerges as a critical and pivotal regulator of both T- and B-cell adaptive immunity against AAV.


Assuntos
Adenoviridae/imunologia , Anticorpos/imunologia , Anticorpos/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Células Th1/imunologia , Animais , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
6.
Transplantation ; 87(9 Suppl): S53-4, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19424007

RESUMO

Several reports have demonstrated the ability of mesenchymal stem cells (MSCs) to enhance hemopoietic engraftment and to exert a profound inhibitory effect on T-cell proliferation in vitro, making them a candidate for the prevention/treatment of graft-versus-host disease. Recent publications have highlighted the underlying mechanisms of the immunosuppressive function of MSCs, elicited by proinflammatory cytokines, such as interferon-gamma and tumor necrosis factor-alpha. These new findings have lead to a better understanding of the biology of MSCs, the outcome of graft-versus-host disease clinical trials and the conflicting results of preclinical T-cell-dependent pathologies. The potential use of MSCs as immunosuppressive elements in allogeneic stem-cell transplantation settings is thus discussed below.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Ensaios Clínicos como Assunto , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Interferon gama/imunologia , Ativação Linfocitária , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/imunologia
7.
Springer Semin Immunopathol ; 28(1): 25-9, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16838181

RESUMO

Alloreactive T cells present in a bone marrow transplant are responsible for graft-vs-host disease, but their depletion is associated with impaired engraftment, immunosuppression, and loss of the graft-vs-leukemia effect. The subpopulation of CD4(+)CD25(+) immunoregulatory T cells was first identified based on its crucial role in the control of autoimmune processes, but they also play a role in alloreactive responses. Moreover, these cells could be used to develop innovative strategies in the field of transplantation and particularly to prevent graft-vs-host disease. Indeed, high numbers of CD4(+)CD25(+) immunoregulatory T cells can modulate graft-vs-host disease if administered at the same time as allogeneic hematopoietic stem cell transplantation in mice. This review discusses various important issues regarding the possible use of CD4(+)CD25(+) immunoregulatory T cells to modulate alloreactivity in hematopoietic stem cell transplantation.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Linfócitos T Reguladores/imunologia , Animais , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos
8.
J Immunol ; 176(12): 7761-7, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16751424

RESUMO

Several reports have suggested that mesenchymal stem cells (MSCs) could exert a potent immunosuppressive effect in vitro, and thus may have a therapeutic potential for T cell-dependent pathologies. We aimed to establish whether MSCs could be used to control graft-vs-host disease (GVHD), a major cause of morbidity and mortality after allogeneic hemopoietic stem cell transplantation. From C57BL/6 and BALB/c mouse bone marrow cells, we purified and expanded MSCs characterized by the lack of expression of CD45 and CD11b molecules, their typical spindle-shaped morphology, together with their ability to differentiate into osteogenic, chondrogenic, and adipogenic cells. These MSCs suppressed alloantigen-induced T cell proliferation in vitro in a dose-dependent manner, independently of their MHC haplotype. However, when MSCs were added to a bone marrow transplant at a MSCs:T cells ratio that provided a strong inhibition of the allogeneic responses in vitro, they yielded no clinical benefit on the incidence or severity of GVHD. The absence of clinical effect was not due to MSC rejection because they still could be detected in grafted animals, but rather to an absence of suppressive effect on donor T cell division in vivo. Thus, in these murine models, experimental data do not support a significant immunosuppressive effect of MSCs in vivo for the treatment of GVHD.


Assuntos
Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/patologia , Proliferação de Células , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Terapia de Imunossupressão , Linfócitos/imunologia , Transplante de Células-Tronco Mesenquimais , Animais , Transplante de Medula Óssea/efeitos adversos , Separação Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Doença Enxerto-Hospedeiro/prevenção & controle , Imunofenotipagem , Teste de Cultura Mista de Linfócitos , Linfócitos/patologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Quimera por Radiação , Baço/citologia , Baço/imunologia , Baço/metabolismo
9.
J Immunol ; 176(2): 1266-73, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16394018

RESUMO

CD4+CD25+ immunoregulatory T cells (Tregs) can be administered to inhibit graft-vs-host disease (GVHD) while preserving graft-vs-leukemia activity after allogeneic bone marrow transplantation in mice. Preclinical studies suggest that it is necessary to infuse as many Tregs as conventional donor T cells to achieve a clinical effect on GVHD. Thus, it would be necessary to expand Tregs ex vivo before transplantation. Two strategies have been proposed: expansion of Tregs stimulated by anti-CD3/CD28-coated microbeads for polyclonal activation or by host-type allogeneic APCs for selecting Tregs specific for host Ags. In this study, we describe the mechanisms by which ex vivo-expanded Tregs act on donor T cells to prevent GVHD in mice. We demonstrate that expanded Tregs strongly inhibited the division, expansion, and differentiation of donor T cells, with a more pronounced effect with Tregs specific for host Ags. These latter cells permit the efficient and durable control of GVHD and favor immune reconstitution.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Linfócitos T Reguladores/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Animais , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/patologia , Diferenciação Celular , Divisão Celular , Feminino , Doença Enxerto-Hospedeiro/patologia , Técnicas In Vitro , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Doadores de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...