Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
2.
Stem Cells ; 33(6): 1892-901, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25753288

RESUMO

The immunomodulatory capacity of mesenchymal stem cells (MSCs) is critical for their use in therapeutic applications. MSC response to specific inflammatory cues allows them to switch between a proinflammatory (MSC1) or anti-inflammatory (MSC2) phenotype. Regulatory mechanisms controlling this switch remain to be defined. One characteristic feature of MSC2 is their ability to respond to IFNγ with induction of indoleamine 2,3-dioxygenase (IDO), representing the key immunoregulatory molecule released by human MSC. Here, we show that STAT1 and PI3Kα pathways interplay regulates IFNγ-induced IDO production in MSC. Chemical phosphoinositide 3-kinase (PI3K) pan-inhibition, PI3Kα-specific inhibition or shRNA knockdown diminished IFNγ-induced IDO production. This effect involved PI3Kα-mediated upregulation of STAT1 protein levels and phosphorylation at Ser727. Overexpression of STAT1 or of a constitutively active PI3Kα mutant failed to induce basal IDO production, but shifted MSC into an MSC2-like phenotype by strongly enhancing IDO production in response to IFNγ as compared to controls. STAT1 overexpression strongly enhanced MSC-mediated T-cell suppression. The same effect could be induced using short-term pretreatment of MSC with a chemical inhibitor of the counter player of PI3K, phosphatase and tensin homolog. Finally, downregulation of STAT1 abrogated the immunosuppressive capacity of MSC. Our results for the first time identify critical upstream signals for the induced production of IDO in MSCs that could be manipulated therapeutically to enhance their immunosuppressive phenotype.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Transcrição STAT1/metabolismo , Classe I de Fosfatidilinositol 3-Quinases , Regulação para Baixo , Humanos , Interferon gama/metabolismo , Fator de Transcrição STAT3/metabolismo , Regulação para Cima
3.
Stem Cells ; 33(5): 1523-31, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25641589

RESUMO

Type 1 diabetes (T1D) is an immune-mediated disease resulting in destruction of insulin-producing pancreatic beta cells. Mesenchymal stem cells (MSCs) possess potent immunomodulatory properties, garnering increasing attention as cellular therapy for T1D and other immunologic diseases. However, MSCs generally lack homing molecules, hindering their colonization at inflammatory sites following intravenous (IV) administration. Here, we analyzed whether enforced E-selectin ligand expression on murine MSCs could impact their effect in reversing hyperglycemia in nonobese diabetic (NOD) mice. Although murine MSCs natively do not express the E-selectin-binding determinant sialyl Lewis(x) (sLe(x) ), we found that fucosyltransferase-mediated α(1,3)-exofucosylation of murine MSCs resulted in sLe(x) display uniquely on cell surface CD44 thereby creating hematopoietic cell E-/L-selectin ligand (HCELL), the E-selectin-binding glycoform of CD44. Following IV infusion into diabetic NOD mice, allogeneic HCELL(+) MSCs showed threefold greater peri-islet infiltrates compared to buffer-treated (i.e., HCELL(-) ) MSCs, with distribution in proximity to E-selectin-expressing microvessels. Exofucosylation had no effect on MSC immunosuppressive capacity in in vitro assays; however, although engraftment was temporary for both HCELL(+) and HCELL(-) MSCs, administration of HCELL(+) MSCs resulted in durable reversal of hyperglycemia, whereas only transient reversal was observed following administration of HCELL(-) MSCs. Notably, exofucosylation of MSCs generated from CD44(-/-) mice induced prominent membrane expression of sLe(x) , but IV administration of these MSCs into hyperglycemic NOD mice showed no enhanced pancreatotropism or reversal of hyperglycemia. These findings provide evidence that glycan engineering to enforce HCELL expression boosts trafficking of infused MSCs to pancreatic islets of NOD mice and substantially improves their efficacy in reversing autoimmune diabetes. Stem Cells 2013;33:1523-1531.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Receptores de Hialuronatos/metabolismo , Células Secretoras de Insulina/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Sobrevivência Celular , Diabetes Mellitus Tipo 1/complicações , Selectina E/metabolismo , Imunofluorescência , Fucose/metabolismo , Fucosiltransferases/metabolismo , Humanos , Hiperglicemia/complicações , Hiperglicemia/patologia , Terapia de Imunossupressão , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Ligação Proteica , Estresse Mecânico , Galactosídeo 2-alfa-L-Fucosiltransferase
4.
Am J Pathol ; 184(12): 3344-58, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25455688

RESUMO

Clinical trials investigating mesenchymal stromal cell (MSC) therapy for bronchopulmonary dysplasia have been initiated; however, the optimal delivery route and functional effects of MSC therapy in newborns remain incompletely established. We studied the morphologic and functional effects of intranasal versus i.p. MSC administration in a rodent model of neonatal lung injury. Cultured human cord tissue MSCs (0.1, 0.5, or 1 × 10(6) cell per pup) were given intranasally or i.p. to newborn severe combined immunodeficiency-beige mice exposed to 90% O2 from birth; sham controls received an equal volume of phosphate-buffered saline. Lung mechanics, engraftment, lung growth, and alveolarization were evaluated 8 weeks after transplantation. High-dose i.p. MSC administration to newborn mice exposed to 90% O2 resulted in the restoration of normal lung compliance, elastance, and pressure-volume loops (tissue recoil). Histologically, high-dose i.p. MSC administration was associated with alveolar septal widening, suggestive of interstitial matrix modification. Intranasal MSC or lower-dose i.p. administration had no significant effects on lung function or alveolar remodeling. Pulmonary engraftment was rare in all the groups. These findings suggest that high-dose systemic administration of human cultured MSCs can restore normal compliance in neonatally injured lungs, possibly by paracrine modulation of the interstitial matrix. Intranasal delivery had no obvious pulmonary effects.


Assuntos
Lesão Pulmonar/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Cordão Umbilical/metabolismo , Administração Intranasal , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/metabolismo , Linhagem da Célula , Células Cultivadas , Humanos , Hiperóxia , Injeções Intraperitoneais , Pulmão/patologia , Camundongos , Camundongos SCID , Oxigênio/química
5.
J Leukoc Biol ; 96(2): 283-93, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24676276

RESUMO

Whereas DC have increasingly been recognized for their role in activating the inflammatory cascades during IRIs, the mechanisms by which oxidative stress enhances DC activation remain to be explored. We examined the role of oxidative stress on two important features of DC: T cell activation and trafficking. Bone marrow-derived OS-DC were compared with untreated DC. DC exposed to oxidative stress augmented allogeneic T cell proliferation and showed increased migration in a chemotaxis chamber. These results were confirmed by using hypoxanthine and xanthine oxidase as another inducer of oxidative stress. We used OT-II and OT-I mice to assess the effect of oxidative stress on DC activation of OVA-specific CD4(+) and CD8(+) T cells, respectively. Oxidative stress increased DC capacity to promote OVA-specific CD4(+) T cell activity, demonstrated by an increase in their proliferation and production of IFN-γ, IL-6, and IL-2 proinflammatory cytokines. Whereas oxidative stress increased the DC ability to stimulate IFN-γ production by OVA-specific CD8(+) T cells, cellular proliferation and cytotoxicity were not affected. Compared with untreated DC, oxidative stress significantly reduced the capacity of DC to generate T(regs), which were restored by using anti-IL-6. With regard to DC trafficking, whereas oxidative stress increased DC expression of p-Akt and p-NF-κB, targeting PI3Kγ and NF-κB pathways abrogated the observed increase in DC migration. Our data propose novel insights on the activation of DC by oxidative stress and provide rationales for targeted therapies, which can potentially attenuate IRI.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Movimento Celular/imunologia , Células Dendríticas/imunologia , Estresse Oxidativo/imunologia , Linfócitos T Reguladores/imunologia , Regulação para Cima/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Movimento Celular/genética , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , Estresse Oxidativo/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Linfócitos T Reguladores/citologia , Regulação para Cima/genética
6.
Methods Mol Biol ; 1098: 211-27, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24166380

RESUMO

Regulatory T cells (Tregs) are amongst the most widely studied cells in a variety of immune-mediated conditions, including transplantation and Graft Versus Host Disease (GVHD), cancer and autoimmunity; indeed, there is great interest in the tolerogenic potential of Treg-based therapy. Consequently, the need to establish the mechanisms that determine Treg survival and longevity, in addition to developing new tools to monitor these parameters, is paramount. Using both a mouse model of GVHD and a mouse model of Type 1 Diabetes (T1D), we describe herein a dual reporter system based on Gluc and multiplexed with SEAP and non-secreted Firefly luciferase (Fluc), which permits simultaneous imaging and noninvasive tracking of two different T-cell populations (CD4(+)CD25(+) Tregs and CD4(+)CD25(-) Tcon cells) in vivo by transducing the cells with different lentiviruses bearing distinct color signatures. This new technology promises to overcome the limitations of the conventional methods currently available to study lymphocyte survival in vivo. Furthermore, this novel technique has applications not only in autoimmunity and alloimmunity, but also in the wider field of immunology.


Assuntos
Fosfatase Alcalina/metabolismo , Copépodes/enzimologia , Luciferases de Vaga-Lume/metabolismo , Medições Luminescentes , Imagem Molecular/métodos , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Reguladores/citologia , Animais , Células da Medula Óssea/citologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/imunologia , Células HEK293 , Humanos , Lentivirus/genética , Luciferases de Vaga-Lume/genética , Camundongos , Células Precursoras de Linfócitos T/citologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Reguladores/metabolismo , Fatores de Tempo
7.
J Immunol ; 191(5): 2319-27, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23913965

RESUMO

Regulatory T cells (Tregs) play a pivotal role in the maintenance of immune tolerance and hold great promise as cell therapy for a variety of immune-mediated diseases. However, the cellular mechanisms that regulate Treg maintenance and homeostasis have yet to be fully explored. Although Tregs express granzyme-B (GrB) to suppress effector T cells via direct killing, the mechanisms by which they protect themselves from GrB-mediated self-inflicted damage are unknown. To our knowledge, we show for the first time that both induced Tregs and natural Tregs (nTregs) increase their intracellular expression of GrB and its endogenous inhibitor, serine protease inhibitor 6 (Spi6) upon activation. Subcellular fractionation and measurement of GrB activity in the cytoplasm of Tregs show that activated Spi6(-/-) Tregs had significantly higher cytoplasmic GrB activity. We observed an increase in GrB-mediated apoptosis in Spi6(-/-) nTregs and impaired suppression of alloreactive T cells in vitro. Spi6(-/-) Tregs were rescued from apoptosis by the addition of a GrB inhibitor (Z-AAD-CMK) in vitro. Furthermore, adoptive transfer experiments showed that Spi6(-/-) nTregs were less effective than wild type nTregs in suppressing graft-versus-host disease because of their impaired survival, as shown in our in vivo bioluminescence imaging. Finally, Spi6-deficient recipients rejected MHC class II-mismatch heart allografts at a much faster rate and showed a higher rate of apoptosis among Tregs, as compared with wild type recipients. To our knowledge, our data demonstrate, for the first time, a novel role for Spi6 in Treg homeostasis by protecting activated Tregs from GrB-mediated injury. These data could have significant clinical implications for Treg-based therapy in immune-mediated diseases.


Assuntos
Granzimas/imunologia , Homeostase/imunologia , Proteínas de Membrana/imunologia , Serina Endopeptidases/imunologia , Serpinas/imunologia , Linfócitos T Reguladores/imunologia , Animais , Apoptose/imunologia , Citometria de Fluxo , Doença Enxerto-Hospedeiro/imunologia , Granzimas/metabolismo , Transplante de Coração/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Serina Endopeptidases/metabolismo , Serpinas/metabolismo , Linfócitos T Reguladores/metabolismo
8.
J Immunol ; 190(1): 88-96, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23180822

RESUMO

TIM-3 is constitutively expressed on subsets of macrophages and dendritic cells. Its expression on other cells of the innate immune system and its role in fetomaternal tolerance has not yet been explored. In this study, we investigate the role of TIM-3-expressing innate immune cells in the regulation of tolerance at the fetomaternal interface (FMI) using an allogeneic mouse model of pregnancy. Blockade of TIM-3 results in accumulation of inflammatory granulocytes and macrophages at the uteroplacental interface and upregulation of proinflammatory cytokines. Furthermore, TIM-3 blockade inhibits the phagocytic potential of uterine macrophages resulting in a build up of apoptotic bodies at the uteroplacental interface that elicits a local immune response. In response to inflammatory cytokines, Ly-6C(hi)G(neg) monocytic myeloid-derived suppressor cells expressing inducible NO synthase and arginase 1 are induced. However, these suppressive cells fail to downregulate the inflammatory cascade induced by inflammatory granulocytes (Ly-6C(int)G(hi)) and apoptotic cells; the increased production of IFN-γ and TNF-α by inflammatory granulocytes leads to abrogation of tolerance at the FMI and fetal rejection. These data highlight the interplay between cells of the innate immune system at the FMI and their influence on successful pregnancy in mice.


Assuntos
Tolerância Imunológica , Imunidade Celular , Imunidade Inata , Receptores Virais/fisiologia , Animais , Apoptose/imunologia , Linhagem Celular , Técnicas de Cocultura , Feminino , Receptor Celular 2 do Vírus da Hepatite A , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Gravidez , Receptores Virais/biossíntese , Útero/citologia , Útero/imunologia , Útero/metabolismo
9.
J Transplant ; 2012: 896141, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22545201

RESUMO

We encapsulated cyclosporine A (CsA) in poly(ethylene glycol)-b-poly(d,l-lactide-co-glycolide) (PEG-PLGA) nanoparticles (NPs) by nanoprecipitation of CsA and PEG-PLGA. The resulting CsA/PEG-PLGA-NPs were <100 nm in diameter with a narrow particle size distribution. The NP size could be controlled by tuning the polymer concentration, solvent, or water/solvent ratio during formulation. The PEGylated NPs maintained non-aggregated in salt solution. Solid NPs lyoprotected with bovine serum albumin were prepared for the convenience of storage and transportation. The release kinetics of CsA (55.6% released on Day 1) showed potential for maintaining therapeutic CsA concentrations in vivo. In T-cell assays, both free CsA and CsA/PEG-PLGA-NPs suppressed T-cell proliferation and production of inflammatory cytokines dose dependently. In a mixed lymphocyte reaction assay, the IC(50) values for free CsA and CsA/PEG-PLGA-NPs were found to be 30 and 35 ng/mL, respectively. This nanoparticulate CsA delivery technology constitutes a strong basis for future targeted delivery of immunosuppressive drugs with improved efficiency and potentially reduced toxicity.

10.
Diabetes ; 61(6): 1509-18, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22403300

RESUMO

Type 1 diabetes (T1D) remains a major health problem worldwide, with a steadily rising incidence yet no cure. Phosphoinositide 3-kinase-γ (PI3Kγ), a member of a family of lipid kinases expressed primarily in leukocytes, has been the subject of substantial research for its role in inflammatory diseases. However, the role of PI3Kγ inhibition in suppressing autoimmune T1D remains to be explored. We tested the role of the PI3Kγ inhibitor AS605240 in preventing and reversing diabetes in NOD mice and assessed the mechanisms by which this inhibition abrogates T1D. Our data indicate that the PI3Kγ pathway is highly activated in T1D. In NOD mice, we found upregulated expression of phosphorylated Akt (PAkt) in splenocytes. Notably, T regulatory cells (Tregs) showed significantly lower expression of PAkt compared with effector T cells. Inhibition of the PI3Kγ pathway by AS605240 efficiently suppressed effector T cells and induced Treg expansion through the cAMP response element-binding pathway. AS605240 effectively prevented and reversed autoimmune diabetes in NOD mice and suppressed T-cell activation and the production of inflammatory cytokines by autoreactive T cells in vitro and in vivo. These studies demonstrate the key role of the PI3Kγ pathway in determining the balance of Tregs and autoreactive cells regulating autoimmune diabetes.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Quinoxalinas/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Tiazolidinedionas/uso terapêutico , Animais , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinoxalinas/farmacologia , Baço/efeitos dos fármacos , Baço/metabolismo , Linfócitos T Reguladores/metabolismo , Tiazolidinedionas/farmacologia
11.
Adv Exp Med Biol ; 771: 62-75, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23393672

RESUMO

Type 1 diabetes mellitus (T1DM) is an autoimmune disease that involves the progressive destruction of the insulin-producing beta cells in the islets of langerhans. It is a complex process that results from the loss of tolerance to insulin and other beta-cell-specific antigens. Various genetic and environmental factors have been studied so far, but precise causation has yet to be established. Numerous studies in rodents and human subjects have been performed in order to elucidate the role of B and T cells, which determine the risk of development and progression of diabetes. These studies have demonstrated that while T1DM is fundamentally a T-cell-mediated autoimmune response, the development of this disease results from complex interactions between the adaptive and innate immune systems, with numerous cell types thought to contribute to pathogenesis. Like any complex disease, the variation in severity and incidence of T1DM can be attributed to a combination of genetic and environmental factors.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Células Secretoras de Insulina/imunologia , Animais , Autoantígenos/imunologia , Diabetes Mellitus Tipo 1/epidemiologia , Diabetes Mellitus Tipo 1/patologia , Humanos , Infecções/imunologia , Células Secretoras de Insulina/patologia , Leucócitos/imunologia , Roedores , Transdução de Sinais/imunologia
12.
J Immunol ; 187(5): 2252-60, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21795594

RESUMO

Clinical trials using allogeneic mesenchymal stem cells (MSCs) are ongoing for the purpose of providing therapeutic benefit for a variety of human disorders. Pertinent to their clinical use are the accessibility to sufficient quantities of these cells allowing for repetitive administration, as well as a better understanding of the specific mechanisms by which allogeneic MSCs evade host immune responses that in turn influence their life span following administration. In this report, we sought to characterize and compare human peripheral blood MSCs (hPB-MSCs) with bone marrow-derived MSCs. hPB-MSCs met the established criteria to characterize this cellular lineage, including capacity for self-renewal, differentiation into tissues of mesodermal origin, and expression of phenotypic surface markers. In addition, hPB-MSCs suppressed alloreactive proliferation as well as the production of proinflammatory cytokines. Examination of the mechanisms by which allogeneic MSCs evade the host immune response, which is crucial for their therapeutic use, demonstrated that constitutive expression of serine protease inhibitor 9 (PI-9) on hPB-MSCs and bone marrow-derived MSCs is a major defense mechanism against granzyme B-mediated destruction by NK cells. Similarly, MSCs treated with small interfering RNA for PI-9 increased MSC cellular death, whereas expression of transgenic PI-9 following retroviral transduction protected MSCs. These data significantly advance our understanding of the immunomodulatory role for hPB-MSCs as well as the mechanisms by which they evade host immune responses. These findings contribute to the development of MSC-based therapies for diseases.


Assuntos
Citotoxicidade Imunológica/imunologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Serpinas/imunologia , Células da Medula Óssea/citologia , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/métodos , Diferenciação Celular/imunologia , Proliferação de Células , Separação Celular , Citometria de Fluxo , Humanos , Células Matadoras Naturais/imunologia , Transplante de Células-Tronco de Sangue Periférico/métodos , Serpinas/metabolismo
13.
Hemoglobin ; 33 Suppl 1: S107-15, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20001613

RESUMO

Sickle cell disease (SCD), the commonest monogenetic disorder worldwide, represents a major public health burden because of its significant morbidity and mortality. Advances in molecular and cellular biology have resulted in an accumulation of knowledge on sickle cell pathophysiology and broadened our understanding of the complexity of this molecular disease with heterogeneous manifestations. Natural history studies and clinical trials have provided incremental data on clinical features, complications, and predictors of severity in SCD and, above all, have laid important recommendations for prevention and treatment of complications. Disease modifying therapies that have significantly improved survival of SCD patients have been identified over recent years. Despite increasingly successful therapies and better overall survival, patients continue to die especially with increasing age and health providers caring for SCD patients face major challenges. This article will highlight modern management of SCD and its impact on the lives of affected patients.


Assuntos
Anemia Falciforme/terapia , Anemia Falciforme/mortalidade , Ensaios Clínicos como Assunto , Gerenciamento Clínico , Humanos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA