Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Immunol ; 195(12): 5805-15, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26553075

RESUMO

In transplantation tolerance, numerous regulatory populations have the capacity to inhibit allograft rejection; however, their compensatory capacities have never been clearly evidenced. We have previously demonstrated that the tolerogenic effect mediated by CD8(+)CD45RC(low) regulatory T cells (Tregs) in a model of organ transplantation with CD40Ig could be abrogated by permanent depletion of CD8(+) cells that resulted in allograft rejection in half of the recipients. This result demonstrated that CD8(+) Tregs were essential, but also that half of the recipients still survived indefinitely. We also demonstrated that no other regulatory populations, besides CD8(+) Tregs, could induce and maintain allograft tolerance in CD40Ig-treated tolerant animals. In the current study, we analyzed the mechanisms that arose following CD8(+) Treg depletion and allowed establishment of networks of new regulatory cells to maintain allograft survival. We identified regulatory B cells (Bregs) and regulatory myeloid cells (RegMCs) as being responsible of the maintenance of the long-term allograft survival. We demonstrated that both regulatory cell subsets efficiently inhibited antidonor immune responses in adoptively transferred recipients. Although Bregs were induced, they were not essential for the maintenance of the graft as demonstrated in IgM-deficient recipients. In addition, we showed that RegMCs were the most suppressive and acted alone, whereas Bregs activity was associated with increased suppressive activity of other subsets in adoptively transferred recipients. Altogether, to our knowledge, we demonstrated in this study for the first time the emergence of both Bregs and RegMCs following Tregs depletion and highlighted the importance of regulatory cell networks and their synergistic potential in transplantation.


Assuntos
Linfócitos B Reguladores/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração , Células Mieloides/imunologia , Transferência Adotiva , Animais , Linfócitos B Reguladores/transplante , Comunicação Celular , Células Cultivadas , Imunomodulação , Células Mieloides/transplante , Ratos , Ratos Endogâmicos Lew , Tolerância ao Transplante
2.
Cell Rep ; 43(7): 114390, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38900636

RESUMO

Timed feeding drives adipose browning, although the integrative mechanisms for the same remain unclear. Here, we show that twice-a-night (TAN) feeding generates biphasic oscillations of circulating insulin and leptin, representing their entrainment by timed feeding. Insulin and leptin surges lead to marked cellular, functional, and metabolic remodeling of subcutaneous white adipose tissue (sWAT), resulting in increased energy expenditure. Single-cell RNA-sequencing (scRNA-seq) analyses and flow cytometry demonstrate a role for insulin and leptin surges in innate lymphoid type 2 (ILC2) cell recruitment and sWAT browning, since sWAT depot denervation or loss of leptin or insulin receptor signaling or ILC2 recruitment each dampens TAN feeding-induced sWAT remodeling and energy expenditure. Consistently, recreating insulin and leptin oscillations via once-a-day timed co-injections is sufficient to favorably remodel innervated sWAT. Innervation is necessary for sWAT remodeling, since denervation of sWAT, but not brown adipose tissue (BAT), blocks TAN-induced sWAT remodeling and resolution of inflammation. In sum, reorganization of nutrient-sensitive pathways remodels sWAT and drives the metabolic benefits of timed feeding.

3.
Sci Adv ; 8(17): eabm7012, 2022 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-35476450

RESUMO

The immune checkpoint B7-H3 (CD276) is a member of the B7 family that has been studied in the tumor microenvironment and immunotherapy, but its potential role in metabolism remains largely unknown. Here, we show that B7-H3 is highly expressed in mouse and human adipose tissue at steady state, with the highest levels in adipocyte progenitor cells. B7-H3 is rapidly down-regulated upon the initiation of adipocyte differentiation. Combined RNA sequencing and metabolic studies reveal that B7-H3 stimulates glycolytic and mitochondrial activity of adipocyte progenitors. Loss of B7-H3 in progenitors results in impaired oxidative metabolism program and increased lipid accumulation in derived adipocytes. Consistent with these observations, mice knocked out for B7-H3 develop spontaneous obesity, metabolic dysfunction, and adipose tissue inflammation. Our results reveal an unexpected metabolic role for B7-H3 in adipose tissue and open potential new avenues for the treatment of metabolic diseases by targeting the B7-H3 pathway.

5.
Curr Opin Organ Transplant ; 15(6): 751-6, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20881498

RESUMO

PURPOSE OF REVIEW: Recent advances in phenotypic identification of CD8 T cells with regulatory properties has allowed better definition of this subset of CD8 Tregs (also called CD8 T suppressor cells in the past). Analysis of their potential contribution in transplantation has attracted most of the interest in the past few years. A better understanding of the mechanisms of action and role of antigenic specificity of the different subtypes of CD8 Tregs and their potential in solid organ transplantation will bring considerable advances in the field. This review will focus and summarize the potential of this subset in cell-based treatments of human transplantation and highlight major discovery in the last months. RECENT FINDINGS: Identification of markers such as PD-1 has allowed better definition of the different CD8 Tregs subsets. Recent animal and human studies have identified mechanisms used by antigen-specific CD8 Treg cells in suppressing anti-allograft immune responses such as perforin-dependent cytotoxicity. New strategies of ex-vivo stimulation (through the T-cell receptor or not) of CD8 Tregs have shown future direction for solid organ transplantation. SUMMARY: CD8 Treg cells represent a subset of regulatory T cells with great potential for future therapeutic application and researchers are moving ahead for a better understanding of their action in antigraft immune response suppression.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Transplante de Órgãos , Animais , Linfócitos T CD8-Positivos/transplante , Humanos , Receptores de Antígenos de Linfócitos T/imunologia
6.
Cell Metab ; 30(1): 12-13, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269422

RESUMO

Highly suppressive tumor microenvironments often result in T cell dysfunction and inability to control tumors, but factors regulating this process remain elusive. A new study by Ma et al. (2019) reports that tumor cholesterol devitalizes T cells by modulating endoplasmic reticulum stress pathways, revealing a new mechanism underlying T cell exhaustion.


Assuntos
Neoplasias , Microambiente Tumoral , Linfócitos T CD8-Positivos , Colesterol , Estresse do Retículo Endoplasmático , Humanos
7.
Cell Rep ; 29(13): 4245-4255.e6, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31875536

RESUMO

To reduce the use of non-specific immunosuppressive drugs detrimental to transplant patient health, therapies in development aim to achieve antigen-specific tolerance by promoting antigen-specific regulatory T cells (Tregs). However, identification of the natural antigens recognized by Tregs and the contribution of their dominance in transplantation has been challenging. We identify epitopes derived from distinct major histocompatibility complex (MHC) class II molecules, sharing a 7-amino acid consensus sequence positioned in a central mobile section in complex with MHC class I, recognized by cross-reactive CD8+ Tregs, enriched in the graft. Antigen-specific CD8+ Tregs can be induced in vivo with a 16-amino acid-long peptide to trigger transplant tolerance. Peptides derived from human HLA class II molecules, harboring the rat consensus sequence, also activate and expand human CD8+ Tregs, suggesting its potential in human transplantation. Altogether, this work should facilitate the development of therapies with peptide epitopes for transplantation and improve our understanding of CD8+ Treg recognition.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Reações Cruzadas/imunologia , Rejeição de Enxerto/imunologia , Linfócitos T Reguladores/imunologia , Doadores de Tecidos , Aloenxertos/imunologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência Consenso , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/química , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Ativação Linfocitária/imunologia , Peptídeos/química , Peptídeos/imunologia , Ratos , Vacinação
8.
JCI Insight ; 2(3): e90088, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28194440

RESUMO

Rat and human CD4+ and CD8+ Tregs expressing low levels of CD45RC have strong immunoregulatory properties. We describe here that human CD45 isoforms are nonredundant and identify distinct subsets of cells. We show that CD45RC is not expressed by CD4+ and CD8+ Foxp3+ Tregs, while CD45RA/RB/RO are. Transient administration of a monoclonal antibody (mAb) targeting CD45RC in a rat cardiac allotransplantation model induced transplant tolerance associated with inhibition of allogeneic humoral responses but maintained primary and memory responses against cognate antigens. Anti-CD45RC mAb induced rapid death of CD45RChigh T cells through intrinsic cell signaling but preserved and potentiated CD4+ and CD8+ CD45RClow/- Tregs, which are able to adoptively transfer donor-specific tolerance to grafted recipients. Anti-CD45RC treatment results in distinct transcriptional signature of CD4+ and CD8+ CD45RClow/- Tregs. Finally, we demonstrate that anti-human CD45RC treatment inhibited graft-versus-host disease (GVHD) in immune-humanized NSG mice. Thus, short-term anti-CD45RC is a potent therapeutic candidate to induce transplantation tolerance in human.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Doença Enxerto-Hospedeiro/tratamento farmacológico , Antígenos Comuns de Leucócito/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Doença Enxerto-Hospedeiro/imunologia , Transplante de Coração , Humanos , Imunidade Humoral/efeitos dos fármacos , Camundongos , Ratos , Tolerância ao Transplante
9.
Clin Cancer Res ; 22(14): 3425-3431, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27208063

RESUMO

B7-H3 (CD276) is an important immune checkpoint member of the B7 and CD28 families. Induced on antigen-presenting cells, B7-H3 plays an important role in the inhibition of T-cell function. Importantly, B7-H3 is highly overexpressed on a wide range of human solid cancers and often correlates with both negative prognosis and poor clinical outcome in patients. Challenges remain to identify the receptor(s) of B7-H3 and thus better elucidate the role of the B7-H3 pathway in immune responses and tumor evasion. With a preferential expression on tumor cells, B7-H3 is an attractive target for cancer immunotherapy. Based on the clinical success of inhibitory immune checkpoint blockade (CTLA-4, PD-1, and PD-L1), mAbs against B7-H3 appear to be a promising therapeutic strategy worthy of development. An unconventional mAb against B7-H3 with antibody-dependent cell-mediated cytotoxicity is currently being evaluated in a phase I clinical trial and has shown encouraging preliminary results. Additional therapeutic approaches in targeting B7-H3, such as blocking mAbs, bispecific mAbs, chimeric antigen receptor T cells, small-molecule inhibitors, and combination therapies, should be evaluated, as these technologies have already shown positive results in various cancer settings. A better understanding of the B7-H3 pathway in humans will surely help to further optimize associated cancer immunotherapies. Clin Cancer Res; 22(14); 3425-31. ©2016 AACR.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antígenos B7/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Ensaios Clínicos Fase I como Assunto , Humanos , Imunoterapia/métodos , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
10.
J Clin Invest ; 125(10): 3952-64, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26389674

RESUMO

Cytokines and metabolic pathway-controlling enzymes regulate immune responses and have potential as powerful tools to mediate immune tolerance. Blockade of the interaction between CD40 and CD40L induces long-term cardiac allograft survival in rats through a CD8+CD45RClo Treg potentiation. Here, we have shown that the cytokine IL-34, the immunoregulatory properties of which have not been previously studied in transplantation or T cell biology, is expressed by rodent CD8+CD45RClo Tregs and human FOXP3+CD45RCloCD8+ and CD4+ Tregs. IL-34 was involved in the suppressive function of both CD8+ and CD4+ Tregs and markedly inhibited alloreactive immune responses. Additionally, in a rat cardiac allograft model, IL-34 potently induced transplant tolerance that was associated with a total inhibition of alloantibody production. Treatment of rats with IL-34 promoted allograft tolerance that was mediated by induction of CD8+ and CD4+ Tregs. Moreover, these Tregs were capable of serial tolerance induction through modulation of macrophages that migrate early to the graft. Finally, we demonstrated that human macrophages cultured in the presence of IL-34 greatly expanded CD8+ and CD4+ FOXP3+ Tregs, with a superior suppressive potential of antidonor immune responses compared with non-IL-34-expanded Tregs. In conclusion, we reveal that IL-34 serves as a suppressive Treg-specific cytokine and as a tolerogenic cytokine that efficiently inhibits alloreactive immune responses and mediates transplant tolerance.


Assuntos
Interleucinas/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Tolerância ao Transplante , Transferência Adotiva , Aloenxertos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Dependovirus/genética , Fatores de Transcrição Forkhead/análise , Vetores Genéticos , Células HEK293 , Transplante de Coração , Humanos , Interleucinas/biossíntese , Interleucinas/genética , Interleucinas/farmacologia , Antígenos Comuns de Leucócito/análise , Ativação Linfocitária , Fator Estimulador de Colônias de Macrófagos/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos Lew , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Proteínas Recombinantes de Fusão/genética , Análise Serial de Tecidos , Transdução Genética
11.
PLoS One ; 10(3): e0119686, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25763980

RESUMO

We previously described that in a rat model of heart transplantation tolerance was dependent on CD8+CD45RClow Tregs that over-expressed fibrinogen-like protein 2 (FGL2)/fibroleukin. Little is known on the immunoregulatory properties of FGL2. Here we analyzed the transplantation tolerance mechanisms that are present in Lewis 1A rats treated with FGL2. Over-expression of FGL2 in vivo through adenovirus associated virus -mediated gene transfer without any further treatment resulted in inhibition of cardiac allograft rejection. Adoptive cell transfer of splenocytes from FGL2-treated rats with long-term graft survival (> 80 days) in animals that were transplanted with cardiac allografts inhibited acute and chronic organ rejection in a donor-specific and transferable tolerance manner, since iterative adoptive transfer up to a sixth consecutive recipient resulted in transplantation tolerance. Adoptive cell transfer also efficiently inhibited anti-donor antibody production. Analysis of all possible cell populations among splenocytes revealed that B lymphocytes were sufficient for this adoptive cell tolerance. These B cells were also capable of inhibiting the proliferation of CD4+ T cells in response to allogeneic stimuli. Moreover, gene transfer of FGL2 in B cell deficient rats did not prolong graft survival. Thus, this is the first description of FGL2 resulting in long-term allograft survival. Furthermore, allograft tolerance was transferable and B cells were the main cells responsible for this effect.


Assuntos
Aloenxertos/transplante , Linfócitos B Reguladores/metabolismo , Fibrinogênio/administração & dosagem , Rejeição de Enxerto/prevenção & controle , Rejeição de Enxerto/terapia , Sobrevivência de Enxerto , Animais , Fibrinogênio/genética , Técnicas de Transferência de Genes , Rejeição de Enxerto/genética , Rejeição de Enxerto/metabolismo , Masculino , Ratos
12.
J Clin Invest ; 124(6): 2497-512, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24789907

RESUMO

In a rat heart allograft model, preventing T cell costimulation with CD40Ig leads to indefinite allograft survival, which is mediated by the induction of CD8+CD45RClo regulatory T cells (CD8+CD40Ig Tregs) interacting with plasmacytoid dendritic cells (pDCs). The role of TCR-MHC-peptide interaction in regulating Treg activity remains a topic of debate. Here, we identified a donor MHC class II-derived peptide (Du51) that is recognized by TCR-biased CD8+CD40Ig Tregs and activating CD8+CD40Ig Tregs in both its phenotype and suppression of antidonor alloreactive T cell responses. We generated a labeled tetramer (MHC-I RT1.Aa/Du51) to localize and quantify Du51-specific T cells within rat cardiac allografts and spleen. RT1.Aa/Du51-specific CD8+CD40Ig Tregs were the most suppressive subset of the total Treg population, were essential for in vivo tolerance induction, and expressed a biased, restricted Vß11-TCR repertoire in the spleen and the graft. Finally, we demonstrated that treatment of transplant recipients with the Du51 peptide resulted in indefinite prolongation of allograft survival. These results show that CD8+CD40Ig Tregs recognize a dominant donor antigen, resulting in TCR repertoire alterations in the graft and periphery. Furthermore, this allopeptide has strong therapeutic activity and highlights the importance of TCR-peptide-MHC interaction for Treg generation and function.


Assuntos
Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Linfócitos T Reguladores/imunologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno , Antígenos CD8/metabolismo , Transplante de Coração , Tolerância Imunológica , Isoantígenos/genética , Isoantígenos/imunologia , Complexo Principal de Histocompatibilidade , Masculino , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/imunologia , Ratos , Ratos Endogâmicos Lew , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Reguladores/classificação
13.
Immunotherapy ; 3(4 Suppl): 35-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21524168

RESUMO

Recent studies in the field of CD8(+) Tregs have allowed a better identification and characterization of this subset of regulatory cells. Their key role in the regulation of allogeneic responses is now well established. To take full advantage of CD8(+) Treg cells in future therapeutic applications, a better knowledge is required, particularly concerning the contribution of the T-cell receptor (TCR) in cell function as well as the role and importance of its antigenic specificity. Here, we focused on the CD8(+)CD45RC(low) Tregs, which in rats induce an indefinite long-term allograft acceptance. We summarized recent findings on their interaction properties with antigen-presenting cells. Identification of the antigenic targets and TCR repertoire of CD8(+) Tregs will allow a better understanding of their recognition properties and will highlight the potential of such of specific population in cell-based treatment.


Assuntos
Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Reguladores/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/patologia , Antígenos CD8/biossíntese , Comunicação Celular , Humanos , Modelos Animais , Ratos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Imunologia de Transplantes , Tolerância ao Transplante , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA