Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur Respir J ; 62(4)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37696564

RESUMO

BACKGROUND: Around 20% of people with cystic fibrosis (pwCF) do not have access to the triple combination elexacaftor/tezacaftor/ivacaftor (ETI) in Europe because they do not carry the F508del allele on the CF transmembrane conductance regulator (CFTR) gene. Considering that pwCF carrying rare variants may benefit from ETI, including variants already validated by the US Food and Drug Administration (FDA), a compassionate use programme was launched in France. PwCF were invited to undergo a nasal brushing to investigate whether the pharmacological rescue of CFTR activity by ETI in human nasal epithelial cell (HNEC) cultures was predictive of the clinical response. METHODS: CFTR activity correction was studied by short-circuit current in HNEC cultures at basal state (dimethyl sulfoxide (DMSO)) and after ETI incubation and expressed as percentage of normal (wild-type (WT)) CFTR activity after sequential addition of forskolin and Inh-172 (ΔI ETI/DMSO%WT). RESULTS: 11 pwCF carried variants eligible for ETI according to the FDA label and 28 carried variants not listed by the FDA. ETI significantly increased CFTR activity of FDA-approved CFTR variants (I601F, G85E, S492F, M1101K, R347P, R74W;V201M;D1270N and H1085R). We point out ETI correction of non-FDA-approved variants, including N1303K, R334W, R1066C, Q552P and terminal splicing variants (4374+1G>A and 4096-3C>G). ΔI ETI/DMSO%WT was significantly correlated to change in percentage predicted forced expiratory volume in 1 s and sweat chloride concentration (p<0.0001 for both). G85E, R74W;V201M;D1270N, Q552P and M1101K were rescued more efficiently by other CFTR modulator combinations than ETI. CONCLUSIONS: Primary nasal epithelial cells hold promise for expanding the prescription of CFTR modulators in pwCF carrying rare mutants. Additional variants should be discussed for ETI indication.


Assuntos
Fibrose Cística , Humanos , Fibrose Cística/tratamento farmacológico , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Dimetil Sulfóxido , Mutação
2.
J Immunol ; 195(1): 156-65, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26026063

RESUMO

Extrathymic T cell precursors can be detected in many tissues and represent an immediately competent population for rapid T cell reconstitution in the event of immunodeficiencies. Blood T cell progenitors have been detected, but their source in the bone marrow (BM) remains unclear. Prospective purification of BM-resident and circulating progenitors, together with RT-PCR single-cell analysis, was used to evaluate and compare multipotent progenitors (MPPs) and common lymphoid progenitors (CLPs). Molecular analysis of circulating progenitors in comparison with BM-resident progenitors revealed that CCR9(+) progenitors are more abundant in the blood than CCR7(+) progenitors. Second, although Flt3(-) CLPs are less common in the BM, they are abundant in the blood and have reduced Cd25(+)-expressing cells and downregulated c-Kit and IL-7Rα intensities. Third, in contrast, stage 3 MPP (MPP3) cells, the unique circulating MPP subset, have upregulated Il7r, Gata3, and Notch1 in comparison with BM-resident counterparts. Evaluation of the populations' respective abilities to generate splenic T cell precursors (Lin(-)Thy1.2(+)CD25(+)IL7Rα(+)) after grafting recipient nude mice revealed that MPP3 cells were the most effective subset (relative to CLPs). Although several lymphoid genes are expressed by MPP3 cells and Flt3(-) CLPs, the latter only give rise to B cells in the spleen, and Notch1 expression level is not modulated in the blood, as for MPP3 cells. We conclude that CLPs have reached the point where they cannot be a Notch1 target, a limiting condition on the path to T cell engagement.


Assuntos
Linfócitos B/metabolismo , Células da Medula Óssea/metabolismo , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Células-Tronco Multipotentes/metabolismo , Linfócitos T/metabolismo , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular , Linhagem da Célula/imunologia , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Subunidade alfa de Receptor de Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/imunologia , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/imunologia , Receptor Notch1/genética , Receptor Notch1/imunologia , Receptores CCR/genética , Receptores CCR/imunologia , Receptores CCR7/genética , Receptores CCR7/imunologia , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/imunologia , Análise de Célula Única , Linfócitos T/citologia , Linfócitos T/imunologia , Tirosina Quinase 3 Semelhante a fms/deficiência , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/imunologia
3.
Proc Natl Acad Sci U S A ; 110(24): E2199-208, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23716674

RESUMO

Diverse hematopoietic progenitors, including myeloid populations arising in inflammatory and tumoral conditions and multipotent cells, mobilized by hematopoietic growth factors or emerging during parasitic infections, display tolerogenic properties. Innate immune stimuli confer regulatory functions to various mature B-cell subsets but immature B-cell progenitors endowed with suppressive properties per se or after differentiating into more mature regulatory B cells remain to be characterized. Herein we provide evidence for innate pro-B cells (CpG-proBs) that emerged within the bone marrow both in vitro and in vivo upon Toll-like receptor-9 activation and whose adoptive transfer protected nonobese diabetic mice against type 1 diabetes (T1D). These cells responded to IFN-γ released by activated effector T cells (Teffs), by up-regulating their Fas ligand (FasL) expression, which enabled them to kill Teffs through apoptosis. In turn, IFN-γ derived from CpG-proBs enhanced IFN-γ while dramatically reducing IL-21 production by Teffs. In keeping with the crucial pathogenic role played by IL-21 in T1D, adoptively transferred IFN-γ-deficient CpG-proBs did not prevent T1D development. Additionally, CpG-proBs matured in vivo into diverse pancreatic and splenic suppressive FasL(high) B-cell subsets. CpG-proBs may become instrumental in cell therapy of autoimmune diseases either on their own or as graft complement in autologous stem cell transplantation.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Células Precursoras de Linfócitos B/imunologia , Linfócitos T/imunologia , Receptor Toll-Like 9/imunologia , Transferência Adotiva , Animais , Apoptose/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proliferação de Células , Diabetes Mellitus Tipo 1/prevenção & controle , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/metabolismo , Citometria de Fluxo , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucinas/imunologia , Interleucinas/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos NOD , Camundongos Knockout , Oligodesoxirribonucleotídeos/imunologia , Oligodesoxirribonucleotídeos/farmacologia , Células Precursoras de Linfócitos B/metabolismo , Células Precursoras de Linfócitos B/transplante , Linfócitos T/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo
4.
J Immunol ; 191(5): 2266-72, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23878314

RESUMO

G-CSF prevents type 1 diabetes in the NOD mouse by promoting the local recruitment of T regulatory cells (Tregs). This is an indirect effect because adoptive transfer of G-CSF-induced tolerogenic dendritic cells (DCs) promotes Treg accumulation. However, the identity of the particular DC subset and the molecule(s) mediating this effect remain unknown. We demonstrate in this study that the adoptive transfer of CD11c(high)CD8α(-) DCs isolated from pegylated G-CSF (pegG-CSF) recipients, but not that of other DC subtypes, enhanced the pancreatic recruitment of CD4(+)CD25(+)Foxp3(+) Tregs, which generated increased amounts of TGF-ß. Likewise, only CD11c(high)CD8α(-) DCs from pegG-CSF recipients secreted the chemokine CCL22 at levels that effectively attracted Tregs. PegG-CSF was more efficient at enhancing the synthesis of CCL22 by CD11c(high)CD8α(-) DCs from the pancreatic lymph nodes compared with those from the spleen. Accordingly, CD11c(high)CD8α(-) DCs from the pancreatic lymph nodes of pegG-CSF recipients were more efficient than their splenic counterparts in the recruitment of Tregs upon adoptive transfer. Predictably, CD11c(high)CD8α(-) DCs failed to recruit these Tregs both in vivo and in vitro following intracellular neutralization of CCL22. These data assign a key role to CD8α(-) DCs and CCL22 in Treg recruitment in the protection of NOD mice against type 1 diabetes following the treatment with G-CSF.


Assuntos
Quimiocina CCL2/imunologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Antígenos CD8/imunologia , Antígenos CD8/metabolismo , Quimiocina CCL2/metabolismo , Quimiotaxia de Leucócito/imunologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos NOD , Células Mieloides/imunologia , Células Mieloides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/metabolismo
5.
Front Immunol ; 13: 790564, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35479094

RESUMO

Development of Graft Versus Host Disease (GVHD) represents a major impediment in allogeneic hematopoietic stem cell transplantation (HSCT). The observation that the presence of bone marrow and circulating hematogones correlated with reduced GVHD risks prompted us to evaluate whether B-cell progenitors, which provide protection in various autoimmune disease models following activation with the TLR-9 agonist CpG (CpG-proBs), could likewise reduce this allogeneic disorder. In a murine model of GVHD that recapitulates an initial phase of acute GVHD followed by a phase of chronic sclerodermatous GVHD, we found that CpG-proBs, adoptively transferred during the initial phase of disease, reduced the diarrhea score and mostly prevented cutaneous fibrosis. Progenitors migrated to the draining lymph nodes and to the skin where they mainly differentiated into follicular B cells. CpG activation and IFN-γ expression were required for the protective effect, which resulted in reduced CD4+ T-cell-derived production of critical cytokines such as TGF-ß, IL-13 and IL-21. Adoptive transfer of CpG-proBs increased the T follicular regulatory to T follicular helper (Tfr/Tfh) ratio. Moreover, CpG-proBs privileged the accumulation of IL-10-positive CD8+ T cells, B cells and dendritic cells in the skin. However, CpG-proBs did not improve survival. Altogether, our findings support the notion that adoptively transferred CpG-proBs exert immunomodulating effect that alleviates symptoms of GVHD but require additional anti-inflammatory strategy to improve survival.


Assuntos
Linfócitos B Reguladores , Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Animais , Linfócitos T CD8-Positivos , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas/métodos , Camundongos , Camundongos Endogâmicos C57BL
6.
Sci Rep ; 11(1): 4735, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637789

RESUMO

Early innate education of hematopoietic progenitors within the bone marrow (BM) stably primes them for either trained immunity or instead immunoregulatory functions. We herein demonstrate that in vivo or in vitro activation within the BM via Toll-like receptor-9 generates a population of plasmacytoid dendritic cell (pDC) precursors (CpG-pre-pDCs) that, unlike pDC precursors isolated from PBS-incubated BM (PBS-pre-pDCs), are endowed with the capacity to halt progression of ongoing experimental autoimmune encephalomyelitis. CpG activation enhances the selective migration of pDC precursors to the inflamed spinal cord, induces their immediate production of TGF-ß, and after migration, of enhanced levels of IL-27. CpG-pre-pDC derived TGF-ß and IL-27 ensure protection at early and late phases of the disease, respectively. Spinal cords of CpG-pre-pDC-protected recipient mice display enhanced percentages of host-derived pDCs expressing TGF-ß as well as an accumulation of IL-10 producing B cells and of CD11c+ CD11b+ dendritic cells. These results reveal that pDC precursors are conferred stable therapeutic properties by early innate activation within the BM. They further extend to the pDC lineage promising perspectives for cell therapy of autoimmune diseases with innate activated hematopoietic precursor cells.


Assuntos
Células da Medula Óssea/citologia , Células Dendríticas/citologia , Esclerose Múltipla/patologia , Medula Espinal/citologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Feminino , Interleucina-27/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/imunologia , Medula Espinal/imunologia , Medula Espinal/metabolismo , Receptor Toll-Like 9 , Fator de Crescimento Transformador beta/metabolismo
7.
Blood ; 112(6): 2575-8, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18617637

RESUMO

Mechanisms of protection against autoimmune diseases by transplantation of autologous hematopoietic progenitors remain poorly defined. We recently demonstrated that, unlike medullary hematopoietic stem cells (HSCs), mobilized hematopoietic progenitors (HPCs) stimulate peripheral Foxp3(+) regulatory T cell (Treg)-expansion through cell-contact activation of Notch signaling and through as yet undetermined soluble factor(s), distinct from TGF-beta1. Herein we identified one such soluble factor as granulocyte macrophage-colony stimulating factor (GM-CSF), which is produced at higher levels by HPCs than HSCs and whose neutralization significantly reduces the growth-promoting effect of HPCs on Treg. Treg express a functional GM-CSF receptor alpha-chain CD116 and proliferate in response to this cytokine independently from IL2. GM-CSF-expanded Treg-like HPC-expanded Treg-display enhanced suppressive capacity relative to control Treg. Hence, mobilized progenitors stimulate Treg expansion both by cell-contact dependent mechanisms and by their production of GM-CSF.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Células-Tronco Hematopoéticas/imunologia , Tolerância Imunológica , Animais , Comunicação Celular , Proliferação de Células , Fator Estimulador de Colônias de Granulócitos e Macrófagos/análise , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Camundongos , Camundongos Endogâmicos NOD , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos , Linfócitos T Reguladores/citologia
8.
Proc Natl Acad Sci U S A ; 104(48): 19085-90, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18006654

RESUMO

Infiltration of inflammatory cells into pancreatic islets of Langerhans and selective destruction of insulin-secreting beta-cells are characteristics of type 1 diabetes. Uncoupling protein 2 (UCP2) is a mitochondrial protein expressed in immune cells. UCP2 controls macrophage activation by modulating the production of mitochondrial reactive oxygen species (ROS) and MAPK signaling. We investigated the role of UCP2 on immune cell activity in type 1 diabetes in Ucp2-deficient mice. Using the model of multiple low-dose streptozotocin (STZ)-induced diabetes, we found that autoimmune diabetes was strongly accelerated in Ucp2-KO mice, compared with Ucp2-WT mice with increased intraislet lymphocytic infiltration. Macrophages from STZ-treated Ucp2-KO mice had increased IL-1beta and nitric oxide (NO) production, compared with WT macrophages. Moreover, more macrophages were recruited in islets of STZ-treated Ucp2-KO mice, compared with Ucp2-WT mice. This finding also was accompanied by increased NO/ROS-induced damage. Altogether, our data show that inflammation is stronger in Ucp2-KO mice and islets, leading to the exacerbated disease in these mice. Our results highlight the mitochondrial protein UCP2 as a new player in autoimmune diabetes.


Assuntos
Diabetes Mellitus Experimental/imunologia , Canais Iônicos/fisiologia , Ilhotas Pancreáticas/imunologia , Macrófagos Peritoneais/imunologia , Proteínas Mitocondriais/fisiologia , Animais , Glicemia/análise , Células Cultivadas/efeitos dos fármacos , Citocinas/metabolismo , Diabetes Mellitus Experimental/patologia , Progressão da Doença , Inflamação , Interferon gama/farmacologia , Canais Iônicos/deficiência , Canais Iônicos/genética , Ilhotas Pancreáticas/patologia , Lipopolissacarídeos/farmacologia , Linfócitos/imunologia , Linfócitos/patologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/fisiologia , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Estreptozocina/administração & dosagem , Estreptozocina/toxicidade , Proteína Desacopladora 2
9.
Front Immunol ; 11: 607180, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33643294

RESUMO

Allogeneic Hematopoietic Stem Cell Transplantation (Allo-HSCT) is routinely performed with peripheral blood stem cells (PBSCs) mobilized by injection of G-CSF, a growth factor which not only modulates normal hematopoiesis but also induces diverse immature regulatory cells. Based on our previous evidence that G-CSF-mobilized multipotent hematopoietic progenitors (MPP) can increase survival and proliferation of natural regulatory T cells (Tregs) in autoimmune disorders, we addressed the question how these cells come into play in mice and humans in an alloimmune setting. Using a C57BL/6 mouse model, we demonstrate that mobilized MPP enhance the immunosuppressant effect exerted by Tregs, against alloreactive T lymphocytes, both in vitro and in vivo. They do so by migrating to sites of allopriming, interacting with donor Tregs and increasing their numbers, thus reducing the lethality of graft-versus-host disease (GVHD). Protection correlates likewise with increased allospecific Treg counts. Furthermore, we provide evidence for a phenotypically similar MPP population in humans, where it shares the capacity to promote selective Treg expansion in vitro. We postulate that G-CSF-mobilized MPPs might become a valuable cellular therapy to expand donor Tregs in vivo and prevent GVHD, thereby making allo-HSCT safer for the treatment of leukemia patients.


Assuntos
Transferência Adotiva , Proliferação de Células , Doença Enxerto-Hospedeiro/prevenção & controle , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Ativação Linfocitária , Células-Tronco Multipotentes/imunologia , Linfócitos T Reguladores/imunologia , Animais , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Multipotentes/metabolismo , Linfócitos T Reguladores/metabolismo
10.
Front Immunol ; 11: 607175, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424854

RESUMO

Achieving immunoregulation via in vivo expansion of Foxp3+ regulatory CD4+ T cells (Treg) remains challenging. We have shown that mobilization confers to multipotent hematopoietic progenitors (MPPs) the capacity to enhance Treg proliferation. Transcriptomic analysis of Tregs co-cultured with MPPs revealed enhanced expression of genes stabilizing the suppressive function of Tregs as well as the activation of IL-1ß-driven pathways. Adoptive transfer of only 25,000 MPPs effectively reduced the development of experimental autoimmune encephalomyelitis (EAE), a pre-clinical model for multiple sclerosis (MS). Production of the pathogenic cytokines IL-17 and GM-CSF by spinal cord-derived CD4+ T-cells in MPP-protected recipients was reduced while Treg expansion was enhanced. Treg depletion once protection by MPPs was established, triggered disease relapse to the same level as in EAE mice without MPP injection. The key role of IL-1ß was further confirmed in vivo by the lack of protection against EAE in recipients of IL-1ß-deficient MPPs. Mobilized MPPs may thus be worth considering for cell therapy of MS either per se or for enrichment of HSC grafts in autologous bone marrow transplantation already implemented in patients with severe refractory multiple sclerosis.


Assuntos
Transferência Adotiva , Proliferação de Células , Encefalomielite Autoimune Experimental/prevenção & controle , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Ativação Linfocitária , Células-Tronco Multipotentes/imunologia , Medula Espinal/imunologia , Linfócitos T Reguladores/imunologia , Animais , Autoimunidade , Células Cultivadas , Técnicas de Cocultura , Citocinas/genética , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Multipotentes/metabolismo , Medula Espinal/metabolismo , Linfócitos T Reguladores/metabolismo , Transcriptoma
12.
Diabetes ; 54(1): 78-84, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15616013

RESUMO

Accumulating evidence that granulocyte colony-stimulating factor (G-CSF), the key hematopoietic growth factor of the myeloid lineage, not only represents a major component of the endogenous response to infections, but also affects adaptive immune responses, prompted us to investigate the therapeutic potential of G-CSF in autoimmune type 1 diabetes. Treatment with G-CSF protected NOD mice from developing spontaneous diabetes. G-CSF triggered marked recruitment of dendritic cells (DCs), particularly immature CD11c(lo)B220(+) plasmacytoid DCs, with reduced costimulatory signal expression and higher interferon-alpha but lower interleukin-12p70 release capacity than DCs in excipient-treated mice. G-CSF recipients further displayed accumulation of functional CD4(+)CD25(+) regulatory T-cells that produce transforming growth factor-beta1 (TGF-beta1) and actively suppressed diabetes transfer by diabetogenic effector cells in secondary NOD-SCID recipients. G-CSF's ability to promote key tolerogenic interactions between DCs and regulatory T-cells was demonstrated by enhanced recruitment of TGF-beta1-expressing CD4(+)CD25(+) cells after adoptive transfer of DCs isolated from G-CSF- relative to vehicle-treated mice into naive NOD recipients. The present results suggest that G-CSF, a promoter of tolerogenic DCs, may be evaluated for the treatment of human type 1 diabetes, possibly in association with direct inhibitors of T-cell activation. They also provide a rationale for a protective role of the endogenous G-CSF produced during infections in early diabetes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Receptores de Interleucina-2/imunologia , Envelhecimento , Animais , Antígenos CD4/efeitos dos fármacos , Antígenos CD4/imunologia , Citocinas/imunologia , Células Dendríticas/efeitos dos fármacos , Diabetes Mellitus Tipo 1/prevenção & controle , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos NOD , Receptores de Interleucina-2/efeitos dos fármacos , Proteínas Recombinantes , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta1
13.
J Leukoc Biol ; 78(5): 1106-17, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16266974

RESUMO

The Fas/Fas ligand (FasL) pathway has been largely implicated in the homeostasis of mature cells. However, it is still unclear whether it plays a role at the progenitor level. To address this issue, we created chimeric mice by transferring C57BL/6 bone marrow (BM) cells of the lpr (Fas-FasL+) or gld (Fas+FasL-) genotype into Rag-2-/- hosts of the same genetic background. In this model, the consequences of a deficient Fas/FasL pathway on lymphoid differentiation could be evaluated without endogenous competition. Analysis of the chimerism revealed a differential sensitivity of hematopoietic lineages to the lack of Fas receptor signaling. While donor-derived myelo-monocytic cells were similarly distributed in all chimeric mice, mature B cells were deleted in the BM and the spleen of lpr chimera, leading to the absence of the marginal zone (MZ) as detected by immunohistology. In contrast, B cell hematopoiesis was complete in gld chimera but MZ macrophages undetectable. These defects suggest a direct and determinant dual role of FasL regulation in negative selection of B cells and in maintenance of the MZ.


Assuntos
Linfócitos B/imunologia , Diferenciação Celular/imunologia , Glicoproteínas de Membrana/imunologia , Transdução de Sinais/imunologia , Fatores de Necrose Tumoral/imunologia , Receptor fas/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Proliferação de Células , Proteína Ligante Fas , Genótipo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Quimera por Radiação/genética , Quimera por Radiação/imunologia , Baço/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fatores de Necrose Tumoral/genética , Receptor fas/genética
14.
Methods Mol Biol ; 1371: 79-88, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26530795

RESUMO

Control of T-cell responses can be achieved by several subsets of B cells with immunoregulatory functions, mostly acting by provision of the anti-inflammatory cytokine IL-10 or exhibiting killing properties through Fas ligand (Fas-L) or granzyme B-induced cell death. We herein describe the characterization as well as the cellular and molecular mechanisms mediating the suppressive properties of bone marrow immature innate pro-B cell progenitors that emerge upon transient activation of Toll-like receptor 9. They are licensed by activated T-cell-derived IFN-γ to become suppressive by up-regulating their Fas-L expression and inducing effector CD4(+) T-cell apoptosis. They also up-regulate their own IFN-γ production which dramatically reduces T-cell production of a major pathogenic cytokine, IL-21. A single adoptive transfer of as little as 60,000 of them efficiently prevents the onset of spontaneous type 1 diabetes in recipient nonobese diabetes (NOD) mice, highlighting the remarkable regulatory potency of these so-called CpG-proB cell progenitors compared to regulatory cells of diverse lineages so far described. The CpG-proB cell activity is prolonged in vivo by their differentiation after migration in the pancreas and the spleen into B-cell progeny with high Fas-L expression that can keep up inducing apoptosis of effector T cells in the long term.


Assuntos
Imunidade Inata , Imunomodulação , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/fisiologia , Animais , Apoptose/imunologia , Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Proteína Ligante Fas/metabolismo , Humanos , Tolerância Imunológica , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptores Toll-Like/metabolismo
16.
Nat Commun ; 7: 12134, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27396388

RESUMO

The influence of signals perceived by immature B cells during their development in bone marrow on their subsequent functions as mature cells are poorly defined. Here, we show that bone marrow cells transiently stimulated in vivo or in vitro through the Toll-like receptor 9 generate proB cells (CpG-proBs) that interrupt experimental autoimmune encephalomyelitis (EAE) when transferred at the onset of clinical symptoms. Protection requires differentiation of CpG-proBs into mature B cells that home to reactive lymph nodes, where they trap T cells by releasing the CCR7 ligand, CCL19, and to inflamed central nervous system, where they locally limit immunopathogenesis through interleukin-10 production, thereby cooperatively inhibiting ongoing EAE. These data demonstrate that a transient inflammation at the environment, where proB cells develop, is sufficient to confer regulatory functions onto their mature B-cell progeny. In addition, these properties of CpG-proBs open interesting perspectives for cell therapy of autoimmune diseases.


Assuntos
Linfócitos B Reguladores/fisiologia , Transplante de Medula Óssea , Encefalomielite Autoimune Experimental/terapia , Células Precursoras de Linfócitos B/transplante , Animais , Linfócitos B Reguladores/citologia , Diferenciação Celular , Movimento Celular , Quimiocina CCL19/fisiologia , Feminino , Interferon gama/metabolismo , Interleucina-10/metabolismo , Linfonodos/fisiologia , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos , Células Precursoras de Linfócitos B/fisiologia
17.
Sci Transl Med ; 7(281): 281ra42, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25834108

RESUMO

Granulocyte colony-stimulating factor (G-CSF) is routinely used to collect peripheral blood stem cells (PBSCs) from healthy donors for allogeneic hematopoietic stem cell transplantation (allo-HSCT). We show that, in both humans and mice, G-CSF mobilizes a subset of CD34(+) cells with mature monocyte features. These cells, which are phenotypically and functionally conserved in mice and humans, are transcriptionally distinct from myeloid and monocytic precursors but similar to mature monocytes and endowed with immunosuppressive properties. In response to interferon-γ released by activated T cells, these cells produce nitric oxide, which induces allogeneic T cell death both in vitro and in vivo. These apoptotic T cells are engulfed by macrophages that release transforming growth factor-ß and promote regulatory T cell expansion. Indeed, the fraction of CD34(+) monocytes in peripheral blood CD34(+) cells inversely correlates with the incidence of acute graft-versus-host disease (GVHD) in humans. Therefore, G-CSF-mobilized cells are an attractive candidate population to be expanded ex vivo for cellular therapy against GVHD.


Assuntos
Antígenos CD34/metabolismo , Doença Enxerto-Hospedeiro/imunologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Monócitos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Tolerância Imunológica/efeitos dos fármacos , Terapia de Imunossupressão , Interferon gama/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Monócitos/efeitos dos fármacos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Transplante Homólogo
18.
Immunity ; 25(5): 823-34, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17081781

RESUMO

Cellular interactions promoting the in vivo expansion of CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells for maintenance of immune tolerance remain poorly defined. Here we report that mobilized Lin(-)Sca-1(+)c-kit(+) (LSK) hematopoietic progenitor cells (HPCs), unlike medullary hematopoietic stem cells (HSCs), selectively drove the direct, immediate expansion of functional host-derived Treg cells, thereby preventing the progression to overt spontaneous autoimmune diabetes in nonobese diabetic mice. Treg cell expansion required cell-to-cell contact and Notch3 signaling, which was mediated selectively through the Notch ligand Jagged2 expressed by the multipotent HPC subset, as assessed by small interfering RNA (siRNA) silencing. Conversely, notwithstanding their similar multilineage microchimerism, neither sorted Jagged2(-) HPCs nor Jagged2(lo) medullary HSCs were able to expand Treg cells. These data provide evidence for a productive Notch-mediated interaction between a unique subset of mobilized hematopoietic progenitors and Treg cells. They open therapeutic perspectives for autologous transplantation of Jagged2(+) LSK progenitors to promote Treg cell expansion in T cell-mediated diseases.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Tolerância Imunológica , Proteínas de Membrana/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/metabolismo , Transferência Adotiva , Animais , Comunicação Celular/imunologia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/prevenção & controle , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Imuno-Histoquímica , Proteína Jagged-2 , Ativação Linfocitária/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T Reguladores/imunologia
19.
J Immunol ; 175(11): 7085-91, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16301609

RESUMO

In recent years, several investigators have unraveled a previously unrecognized role for G-CSF in the regulation of T cell and dendritic cell functions. The experimental evidence in favor of G-CSF-mediated immune regulation includes the ability to switch T cell cytokine secretion profile to Th2 responses and the promotion of regulatory T cell and tolerogenic dendritic cell differentiation. Interestingly, G-CSF is beneficial in animals for the prevention and/or treatment of immune-mediated diseases, e.g., graft-vs-host disease, multiple sclerosis, systemic lupus erythematosus, inflammatory bowel disease, and diabetes, suggesting a potential role in human autoimmune diseases. This review summarizes the growing body of evidence that supports a critical role for G-CSF as a novel mediator of T cell tolerance.


Assuntos
Fator Estimulador de Colônias de Granulócitos/imunologia , Tolerância Imunológica/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Animais , Apresentação de Antígeno/imunologia , Doenças Autoimunes/tratamento farmacológico , Citocinas/imunologia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Humanos
20.
J Autoimmun ; 24(2): 125-34, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15829405

RESUMO

Cyclophosphamide (CY) accelerates autoimmune diabetes in the NOD mouse at different levels, including critical targeting of a regulatory T cell subset, exacerbation of pro-Th1 IFN-gamma production and promotion of inflammation in pancreatic islets. Here we evaluated the ability of G-CSF to antagonize the acceleration of the disease induced by CY. Human recombinant G-CSF, administered daily at 200 microg/kg by s.c. injection, protected NOD mice from CY-accelerated onset of glycosuria and insulitis. G-CSF accelerated the recovery of the T cell compartment after the depletion of the lymphoid compartment triggered by CY injection. It selectively prevented the loss of the immunoregulatory T cells expressing the CD4(+)CD25+ phenotype that also stained CD62L+ in peripancreatic lymph nodes and promoted their expansion in the spleen. In addition to this, it abrogated the robust cytokine--particularly IFN-gamma- and chemokine burst triggered in immune cells by CY. G-CSF promoted only slight changes in the inflammatory effects of CY at the target tissue site, assessed by chemokine induction within the pancreas. Thus the immunoregulatory properties of G-CSF were critical in the early control of the accelerating effects of CY on autoimmune diabetes in the NOD mouse.


Assuntos
Ciclofosfamida/antagonistas & inibidores , Ciclofosfamida/farmacologia , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/patologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Animais , Antígenos CD/imunologia , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Quimiocinas/biossíntese , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/biossíntese , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/imunologia , Progressão da Doença , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA