Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Acta Neuropathol ; 142(5): 899-915, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34487221

RESUMO

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) characterized by varying degrees of secondary neurodegeneration. Retinal ganglion cells (RGC) are lost in MS in association with optic neuritis but the mechanisms of neuronal injury remain unclear. Complement component C3 has been implicated in retinal and cerebral synaptic pathology that may precede neurodegeneration. Herein, we examined post-mortem MS retinas, and then used a mouse model, experimental autoimmune encephalomyelitis (EAE), to examine the role of C3 in the pathogenesis of RGC loss associated with optic neuritis. First, we show extensive C3 expression in astrocytes (C3+/GFAP+ cells) and significant RGC loss (RBPMS+ cells) in post-mortem retinas from people with MS compared to retinas from non-MS individuals. A patient with progressive MS with a remote history of optic neuritis showed marked reactive astrogliosis with C3 expression in the inner retina extending into deeper layers in the affected eye more than the unaffected eye. To study whether C3 mediates retinal degeneration, we utilized global C3-/- EAE mice and found that they had less RGC loss and partially preserved neurites in the retina compared with C3+/+ EAE mice. C3-/- EAE mice had fewer axonal swellings in the optic nerve, reflecting reduced axonal injury, but had no changes in demyelination or T cell infiltration into the CNS. Using a C3-tdTomato reporter mouse line, we show definitive evidence of C3 expression in astrocytes in the retina and optic nerves of EAE mice. Conditional deletion of C3 in astrocytes showed RGC protection replicating the effects seen in the global knockouts. These data implicate astrocyte C3 expression as a critical mediator of retinal neuronal pathology in EAE and MS, and are consistent with recent studies showing C3 gene variants are associated with faster rates of retinal neurodegeneration in human disease.


Assuntos
Complemento C3/metabolismo , Esclerose Múltipla/patologia , Doenças Neuroinflamatórias/patologia , Células Ganglionares da Retina/patologia , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Camundongos , Esclerose Múltipla/imunologia , Degeneração Neural/imunologia , Degeneração Neural/patologia , Doenças Neuroinflamatórias/imunologia , Nervo Óptico/patologia , Neurite Óptica/imunologia , Neurite Óptica/patologia
2.
Nat Commun ; 12(1): 652, 2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33510160

RESUMO

Injury and loss of oligodendrocytes can cause demyelinating diseases such as multiple sclerosis. To improve our understanding of human oligodendrocyte development, which could facilitate development of remyelination-based treatment strategies, here we describe time-course single-cell-transcriptomic analysis of developing human stem cell-derived oligodendrocyte-lineage-cells (hOLLCs). The study includes hOLLCs derived from both genome engineered embryonic stem cell (ESC) reporter cells containing an Identification-and-Purification tag driven by the endogenous PDGFRα promoter and from unmodified induced pluripotent (iPS) cells. Our analysis uncovers substantial transcriptional heterogeneity of PDGFRα-lineage hOLLCs. We discover sub-populations of human oligodendrocyte progenitor cells (hOPCs) including a potential cytokine-responsive hOPC subset, and identify candidate regulatory genes/networks that define the identity of these sub-populations. Pseudotime trajectory analysis defines developmental pathways of oligodendrocytes vs astrocytes from PDGFRα-expressing hOPCs and predicts differentially expressed genes between the two lineages. In addition, pathway enrichment analysis followed by pharmacological intervention of these pathways confirm that mTOR and cholesterol biosynthesis signaling pathways are involved in maturation of oligodendrocytes from hOPCs.


Assuntos
Heterogeneidade Genética , Variação Genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Análise de Célula Única/métodos , Transcriptoma/genética , Astrócitos/citologia , Astrócitos/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Colesterol/biossíntese , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Redes Reguladoras de Genes/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células Precursoras de Oligodendrócitos/citologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
3.
PLoS One ; 15(6): e0233980, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32511247

RESUMO

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS) that results in variable severities of neurodegeneration. The understanding of MS has been limited by the inaccessibility of the affected cells and the lengthy timeframe of disease development. However, recent advances in stem cell technology have facilitated the bypassing of some of these challenges. Towards gaining a greater understanding of the innate potential of stem cells from people with varying degrees of disability, we generated induced pluripotent stem cells (iPSCs) from peripheral blood mononuclear cells derived from stable and progressive MS patients, and then further differentiated them into oligodendrocyte (OL) lineage cells. We analyzed differentiation under both homeostatic and inflammatory conditions via sustained exposure to low-dose interferon gamma (IFNγ), a prominent cytokine in MS. We found that all iPSC lines differentiated into mature myelinating OLs, but chronic exposure to IFNγ dramatically inhibited differentiation in both MS groups, particularly if exposure was initiated during the pre-progenitor stage. Low-dose IFNγ was not toxic but led to an early upregulation of interferon response genes in OPCs followed by an apparent redirection in lineage commitment from OL to a neuron-like phenotype in a significant portion of the treated cells. Our results reveal that a chronic low-grade inflammatory environment may have profound effects on the efficacy of regenerative therapies.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Esclerose Múltipla Crônica Progressiva/patologia , Oligodendroglia/citologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Homeostase , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/fisiologia , Inflamação/patologia , Interferon gama/farmacologia , Leucócitos Mononucleares/citologia , Regeneração
4.
J Neuroimmunol ; 339: 577115, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31778849

RESUMO

Multiple sclerosis (MS) is an autoimmune disease that remains in need of effective therapies. Plant-derived medicines have appealing properties for the treatment of autoimmune diseases. MYMD-1 is a synthetic plant alkaloid that has been shown to ameliorate the course of autoimmune thyroiditis. The goal of the present study was to determine whether MYMD-1 would produce similar beneficial effects in a mouse model of MS, experimental autoimmune encephalomyelitis (EAE) induced by immunization with myelin oligodendrocyte glycoprotein. MYMD-1 improved the course of EAE and suppressed activation of effector T cells without causing global immunosuppression or toxicity. These results suggest that MYMD-1 may be of interest for evaluating for the treatment of autoimmune diseases.


Assuntos
Alcaloides/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Alcaloides/síntese química , Alcaloides/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
PLoS One ; 14(9): e0221747, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31490950

RESUMO

Multiple sclerosis (MS) is characterized by demyelinated lesions in the central nervous system. Destruction of myelin and secondary damage to axons and neurons leads to significant disability, particularly in people with progressive MS. Accumulating evidence suggests that the potential for myelin repair exists in MS, although for unclear reasons this process fails. The cells responsible for producing myelin, the oligodendrocytes, and their progenitors, oligodendrocyte precursor cells (OPCs), have been identified at the site of lesions, even in adults. Their presence suggests the possibility that endogenous remyelination without transplantation of donor stem cells may be a mechanism for myelin repair in MS. Strategies to develop novel therapies have focused on induction of signaling pathways that stimulate OPCs to mature into myelin-producing oligodendrocytes that could then possibly remyelinate lesions. We have been investigating pharmacological approaches to enhance OPC differentiation, and have identified that the combination of two agents, triiodothyronine (T3) and quetiapine, leads to an additive effect on OPC differentiation and consequent myelin production via both overlapping and distinct signaling pathways. While the ultimate production of myelin requires cholesterol biosynthesis, we identified that quetiapine enhances gene expression in this pathway more potently than T3. Two blockers of cholesterol production, betulin and simvastatin, reduced OPC differentiation into myelin producing oligodendrocytes. Elucidating the nature of agents that lead to complementary and additive effects on oligodendrocyte differentiation and myelin production may pave the way for more efficient induction of remyelination in people with MS.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colesterol/biossíntese , Células Precursoras de Oligodendrócitos/citologia , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Fumarato de Quetiapina/farmacologia , Tri-Iodotironina/farmacologia , Animais , Sinergismo Farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Básica da Mielina/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , Triterpenos/farmacologia
6.
Brain ; 142(9): 2722-2736, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31289819

RESUMO

Multiple sclerosis is a heterogeneous disease with an unpredictable course and a wide range of severity; some individuals rapidly progress to a disabled state whereas others experience only mild symptoms. Though genetic studies have identified variants that are associated with an increased risk of developing multiple sclerosis, no variants have been consistently associated with multiple sclerosis severity. In part, the lack of findings is related to inherent limitations of clinical rating scales; these scales are insensitive to early degenerative changes that underlie disease progression. Optical coherence tomography imaging of the retina and low-contrast letter acuity correlate with and predict clinical and imaging-based outcomes in multiple sclerosis. Therefore, they may serve as sensitive phenotypes to discover genetic predictors of disease course. We conducted a set of genome-wide association studies of longitudinal structural and functional visual pathway phenotypes in multiple sclerosis. First, we assessed genetic predictors of ganglion cell/inner plexiform layer atrophy in a discovery cohort of 374 patients with multiple sclerosis using mixed-effects models adjusting for age, sex, disease duration, optic neuritis and genetic ancestry and using a combination of single-variant and network-based analyses. For candidate variants identified in discovery, we conducted a similar set of analyses of ganglion cell/inner plexiform layer thinning in a replication cohort (n = 376). Second, we assessed genetic predictors of sustained loss of 5-letters in low-contrast letter acuity in discovery (n = 582) using multivariable-adjusted Cox proportional hazards models. We then evaluated candidate variants/pathways in a replication cohort. (n = 253). Results of both studies revealed novel subnetworks highly enriched for connected genes in early complement activation linked to measures of disease severity. Within these networks, C3 was the gene most strongly associated with ganglion cell/inner plexiform layer atrophy (P = 0.004) and C1QA and CR1 were top results in analysis of sustained low-contrast letter acuity loss. Namely, variant rs158772, linked to C1QA, and rs61822967, linked to CR1, were associated with 71% and 40% increases in risk of sustained LCLA loss, respectively, in meta-analysis pooling discovery and replication cohorts (rs158772: hazard ratio: 1.71; 95% confidence interval 1.30-2.25; P = 1.3 × 10-4; rs61822967: hazard ratio: 1.40; 95% confidence interval: 1.16-1.68; P = 4.1 × 10-4). In conclusion, early complement pathway gene variants were consistently associated with structural and functional measures of multiple sclerosis severity. These results from unbiased analyses are strongly supported by several prior reports that mechanistically implicated early complement factors in neurodegeneration.


Assuntos
Proteínas do Sistema Complemento/genética , Redes Reguladoras de Genes/genética , Esclerose Múltipla/genética , Degeneração Neural/genética , Vias Visuais/fisiopatologia , Adulto , Ensaios Clínicos Fase III como Assunto , Método Duplo-Cego , Feminino , Seguimentos , Heterogeneidade Genética , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Polimorfismo de Nucleotídeo Único , Modelos de Riscos Proporcionais , Estudos Prospectivos , Ensaios Clínicos Controlados Aleatórios como Assunto , Retina/patologia , Tomografia de Coerência Óptica
7.
J Leukoc Biol ; 105(5): 829-841, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30762897

RESUMO

Myeloid-derived suppressor cells (MDSCs) are a diverse group of cells that are recognized for their remarkable suppressive effects on pro-inflammatory T cells. The pleiotropic nature of these cells, however, has been demonstrated by their differential effects on immune responses in different settings. Our and others' work has demonstrated suppressive effects of these cells. We previously demonstrated that these cells were mobilized to the lungs during experimental autoimmune encephalomyelitis (EAE), which is a murine model of multiple sclerosis, and potently inhibited CD8+ T cell responses against influenza infection. Interestingly, they appeared to have a lesser effect on CD4+ T cells, and in fact, others have demonstrated that spleen-derived MDSCs could actually promote Th17 differentiation. We sought to determine the role of lung-derived MDSCs on EAE pathogenesis, as excursion through the lungs by pathologic CNS-Ag targeted T cells was shown to be critical for EAE induction. Our results indicate a robust accumulation of granulocytic MDSCs in the lungs of mice during EAE, which could promote Th17 polarization, and which coincided with the trafficking of autoimmune-targeted T cells through the lungs. These studies underscore the pleiotropic effect of MDSCs on T cells and their potential pro-inflammatory phenotypes in neuro-inflammatory disease. Understanding both the intrinsic multifunctional nature of these cells and the ability to influence organ-specific targets such as the CNS from remote organs such as lungs will help to elucidate both mechanisms of disease and possible new therapeutic approaches.


Assuntos
Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Pulmão/imunologia , Células Supressoras Mieloides/imunologia , Células Th17/imunologia , Animais , Autoimunidade , Comunicação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Movimento Celular , Sistema Nervoso Central/patologia , Técnicas de Cocultura , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Feminino , Expressão Gênica , Humanos , Interleucina-17/genética , Interleucina-17/imunologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Glicoproteína Mielina-Oligodendrócito/administração & dosagem , Células Supressoras Mieloides/efeitos dos fármacos , Células Supressoras Mieloides/patologia , Fragmentos de Peptídeos/administração & dosagem , Toxina Pertussis/administração & dosagem , Transdução de Sinais/imunologia , Células Th17/efeitos dos fármacos , Células Th17/patologia , Fator de Crescimento Transformador beta/farmacologia
8.
Biomaterials ; 190-191: 24-37, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30391800

RESUMO

Microvessels of the blood-brain barrier (BBB) regulate transport into the brain. The highly specialized brain microvascular endothelial cells, a major component of the BBB, express tight junctions and efflux transporters which regulate paracellular and transcellular permeability. However, most existing models of BBB microvessels fail to exhibit physiological barrier function. Here, using (iPSC)-derived human brain microvascular endothelial cells (dhBMECs) within templated type I collagen channels we mimic the cylindrical geometry, cell-extracellular matrix interactions, and shear flow typical of human brain post-capillary venules. We characterize the structure and barrier function in comparison to non-brain-specific microvessels, and show that dhBMEC microvessels recapitulate physiologically low solute permeability and quiescent endothelial cell behavior. Transcellular permeability is increased two-fold using a clinically relevant dose of a p-glycoprotein inhibitor tariquidar, while paracellular permeability is increased using a bolus dose of hyperosmolar agent mannitol. Lastly, we show that our human BBB microvessels are responsive to inflammatory cytokines via upregulation of surface adhesion molecules and increased leukocyte adhesion, but no changes in permeability. Human iPSC-derived blood-brain barrier microvessels support quantitative analysis of barrier function and endothelial cell dynamics in quiescence and in response to biologically- and clinically-relevant perturbations.


Assuntos
Barreira Hematoencefálica/citologia , Células Endoteliais/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Microvasos/citologia , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar , Diferenciação Celular , Linhagem Celular , Células Endoteliais/metabolismo , Desenho de Equipamento , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Microvasos/metabolismo , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos
9.
J Neuroimmunol ; 316: 7-16, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29274729

RESUMO

Pro-inflammatory T cells are critical to the pathogenesis of multiple sclerosis (MS). We investigated the potential for the anti-proliferative, pro-apoptotic drug gemcitabine to affect development of MS-relevant effector TH1, TH17, and Treg cells. Gemcitabine directly suppressed proliferation, activation, and induced apoptosis of all effector subsets in subtype and dose-dependent fashion. This drug also prevented development of disease in the MS model experimental autoimmune encephalomyelitis (EAE) and significantly reduced the abundance of TH1 and TH17 cells. Our results indicate that pathogenic CD4+ T cells may be viable targets by gemcitabine for therapeutic benefit in MS.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Encefalomielite Autoimune Experimental/imunologia , Imunossupressores/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Animais , Linfócitos T CD4-Positivos/imunologia , Desoxicitidina/farmacologia , Feminino , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Gencitabina
10.
J Exp Med ; 214(2): 297-307, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28057805

RESUMO

The discovery that central nervous system (CNS)-targeted autoreactive T cells required a process of licensing in the lung revealed an unexpected relationship between these organs. The clinical and immunological significance of this finding is bidirectional in that it showed not only a mechanism by which T cells become pathogenic before entering the CNS, but also the potential for this process to influence lung immunity as well. Epidemiological studies have shown that people with multiple sclerosis (MS) suffer from increased morbidity and mortality from infectious diseases, independent of immunosuppressive therapies. Respiratory infections account for a large percentage of deaths of people with MS. In this study, to investigate the mechanisms responsible for this enhanced susceptibility, we established a comorbid model system in which mice with experimental autoimmune encephalomyelitis (EAE) were administered a sublethal dose of influenza. Whereas mice with either EAE alone or influenza alone survived, 70% of comorbid mice died as a result of uncontrolled viral replication. Immunological analyses revealed that the induction of EAE led to a surprising alteration of the lung milieu, converting an effective stimulatory influenza-reactive environment into a suppressive one. These results provide mechanistic information that may help to explain the unexpected immunological interactions.


Assuntos
Autoimunidade , Encéfalo/imunologia , Infecções por Orthomyxoviridae/mortalidade , Animais , Movimento Celular , Comorbidade , Encefalomielite Autoimune Experimental/mortalidade , Feminino , Pulmão/virologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/fisiologia , Óxido Nítrico Sintase Tipo II/fisiologia , Replicação Viral
11.
PLoS One ; 10(9): e0139008, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26407166

RESUMO

Mesenchymal stem cells (MSCs) are pleiotropic cells with potential therapeutic benefits for a wide range of diseases. Because of their immunomodulatory properties they have been utilized to treat autoimmune diseases such as multiple sclerosis (MS), which is characterized by demyelination. The microenvironment surrounding MSCs is thought to affect their differentiation and phenotype, which could in turn affect the efficacy. We thus sought to dissect the potential for differential impact of MSCs on central nervous system (CNS) disease in T cell mediated and non-T cell mediated settings using the MOG35-55 experimental autoimmune encephalomyelitis (EAE) and cuprizone-mediated demyelination models, respectively. As the pathogeneses of MS and EAE are thought to be mediated by IFNγ-producing (TH1) and IL-17A-producing (TH17) effector CD4+ T cells, we investigated the effect of MSCs on the development of these two key pathogenic cell groups. Although MSCs suppressed the activation and effector function of TH17 cells, they did not affect TH1 activation, but enhanced TH1 effector function and ultimately produced no effect on EAE. In the non- T cell mediated cuprizone model of demyelination, MSC administration had a positive effect, with an overall increase in myelin abundance in the brain of MSC-treated mice compared to controls. These results highlight the potential variability of MSCs as a biologic therapeutic tool in the treatment of autoimmune disease and the need for further investigation into the multifaceted functions of MSCs in diverse microenvironments and the mechanisms behind the diversity.


Assuntos
Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/terapia , Encefalomielite Autoimune Experimental/terapia , Células-Tronco Mesenquimais/citologia , Animais , Linfócitos T CD4-Positivos/imunologia , Morte Celular , Proliferação de Células , Corpo Caloso/patologia , Cuprizona , Citocinas/biossíntese , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/imunologia , Feminino , Subpopulações de Linfócitos/imunologia , Transplante de Células-Tronco Mesenquimais , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/imunologia , Oligodendroglia/patologia , Fragmentos de Peptídeos/imunologia
12.
Crit Rev Oncog ; 20(5-6): 509-18, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27279244

RESUMO

Gene therapy for cancer is a still evolving approach that resulted from a long history of studies into genetic modification of organisms. The fascination with manipulating gene products has spanned hundreds if not thousands of years, beginning with observations of the hereditary nature of traits in plants and culminating to date in the alteration of genetic makeup in humans via modern technology. From early discoveries noting the potential for natural mobility of genetic material to the culmination of clinical trials in a variety of disease, gene transfer has had an eventful and sometimes tumultuous course. Within the present review is a brief history of the biology of gene transfer, how it came to be applied to genetic diseases, and its early applications to cancer therapies. Some of the different types of methods used to modify cells, the theories behind the approaches, and some of the limitations encountered along the way are reviewed.


Assuntos
Terapia Genética/métodos , Sequências Repetitivas Dispersas , Neoplasias/terapia , Animais , Feminino , Humanos , Masculino
13.
World J Stem Cells ; 6(5): 526-39, 2014 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-25426250

RESUMO

Mesenchymal stem cells (MSCs) are a pleiotropic population of cells that are self-renewing and capable of differentiating into canonical cells of the mesenchyme, including adipocytes, chondrocytes, and osteocytes. They employ multi-faceted approaches to maintain bone marrow niche homeostasis and promote wound healing during injury. Biomedical research has long sought to exploit their pleiotropic properties as a basis for cell therapy for a variety of diseases and to facilitate hematopoietic stem cell establishment and stromal reconstruction in bone marrow transplantation. Early results demonstrated their usage as safe, and there was little host response to these cells. The discovery of their immunosuppressive functions ushered in a new interest in MSCs as a promising therapeutic tool to suppress inflammation and down-regulate pathogenic immune responses in graft-versus-host and autoimmune diseases such as multiple sclerosis, autoimmune diabetes, and rheumatoid arthritis. MSCs produce a large number of soluble and membrane-bound factors, some of which inhibit immune responses. However, the full range of MSC-mediated immune-modulation remains incompletely understood, as emerging reports also reveal that MSCs can adopt an immunogenic phenotype, stimulate immune cells, and yield seemingly contradictory results in experimental animal models of inflammatory disease. The present review describes the large body of literature that has been accumulated on the fascinating biology of MSCs and their complex effects on immune responses.

14.
Stem Cells ; 32(10): 2744-55, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24911892

RESUMO

Mesenchymal stem cells (MSC) have emerged as a promising candidate for inflammatory suppression and disease amelioration, especially of neuro-inflammatory diseases such as multiple sclerosis (MS). Auto-reactive CD4+ and CD8+ T cells acquire pathogenic IFNγ-producing- (Type I) and IL-17A-producing- (Type 17) effector phenotypes in MS and its animal model experimental autoimmune encephalomyelitis (EAE). Although MSC have been extensively demonstrated to suppress pathogenic effector CD4+ T cells and CD4+ T cell-mediated EAE, surprisingly few studies have addressed their modulation of effector CD8+ T cells represented in MS or their impact on CD8+ T cell-mediated EAE. We find that MSC differentially modulate CD8+ T cell development depending on effector T cell subtype. MSC drive activated low-IFNγ producers toward an enhanced high-IFNγ Tc1-like phenotype but strongly inhibit the production of IL-17A and Tc17 polarization in vitro. These observations are underscored by differential MSC modulation of T cell activation, proliferation, and signature transcription factor up-regulation. In addition, effector CD8+ T cells co-cultured with MSC exhibited increased production of IL-2, a molecule known to enhance IFNγ, yet suppress IL-17A, production. Based on these in vitro effects on CD8+ T cells, we next evaluated their impact on the severity of EAE. To better evaluate CD8+ T cells, we immunized mice with MOG37-50 , which is a CD8-targeted epitope. Our results revealed a worsening of disease, consistent with their in vitro stimulation of Tc1 cells. These findings highlight the emerging duality of MSC in immune modulation and provide implications for their future use in immune-related diseases.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Progressão da Doença , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Células-Tronco Mesenquimais/citologia , Subpopulações de Linfócitos T/imunologia , Animais , Apoptose , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Técnicas de Cocultura , Feminino , Interferon gama/biossíntese , Interleucina-2/biossíntese , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/imunologia , Proteínas com Domínio T/metabolismo , Subpopulações de Linfócitos T/citologia
15.
J Biol Chem ; 288(33): 23868-74, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23846700

RESUMO

The initiation and maintenance of the immune response require a coordinated regulation of signal transduction pathways. Identifying the mechanisms by which these pathways are controlled and modulated is a significant goal of immunology. In the present report, we show a novel role for the zinc finger transcription factor Kruppel-like factor 4 (KLF4) in the modulation of the inflammatory immune response via its regulation of IL-6. We analyzed the role of KLF4 in the production of IL-6 by dendritic cells. Our data indicate that KLF4 can act in a dual function manner. It acts as a transcription factor in that it can bind to and activate the IL-6 promoter at specific binding sites. KLF4 also has a role in the chromatin remodeling of the IL-6 promoter in that cells deficient in KLF4 exhibited a relative hypoacetylation. These results indicate a molecular role for KLF4 in modulating the intensity of the inflammatory response and help to explain its pleiotropic role in different settings.


Assuntos
Epigênese Genética , Interleucina-6/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Regiões Promotoras Genéticas , Acetilação , Animais , Sequência de Bases , Sítios de Ligação , Sequência Consenso/genética , Células Dendríticas/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Interleucina-6/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/deficiência , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , NF-kappa B/metabolismo , Ligação Proteica/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Transcrição Gênica
16.
PLoS One ; 8(1): e54267, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23382885

RESUMO

Increased expression of the voltage-gated potassium channel Kν1.3 on activated effector memory T cells (T(EM)) is associated with pathology in multiple sclerosis (MS). To date, most studies of Kν1.3 channels in MS have focused on CD4+ T(EM) cells. Much less is known about the functional relevance of Kv1.3 on CD8+ T(EM) cells. Herein, we examined the effects of Kν1.3 blockade on CD8+ T cell proliferation, differentiation into cytotoxic effector cells, and release of granzyme B (GrB), a key effector of CD8+ T cell-mediated cytotoxicity. We confirmed the expression of Kv1.3 channels on activated human CD8+ T lymphocytes by immunofluorescent staining. To test the functional relevance of the Kv1.3 channel in CD8+ T cells, we inhibited this channel via pharmacological blockers or a lentiviral-dominant negative (Kv1.xDN) approach and determined the effects of the blockade on critical pathogenic parameters of CD8+ T cells. We found that blockade of Kv1.3 with both lentivirus and pharmacologic agents effectively inhibited cytotoxic effector memory cells' proliferation, secretion of GrB, and their ability to kill neural progenitor cells. Intriguingly, the KvDN transduced T cells exhibited arrested differentiation from central memory (T(CM)) to effector memory (T(EM)) states. Transduction of cells that had already differentiated into T(EM) with KvDN led to their conversion into T(CM). CD8+ T(EM) have a critical role in MS and other autoimmune diseases. Our present results indicate a critical role for Kv1.3 in the conversion of CD8+ T cells into potential pathogenic effector cells with cytotoxic function.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , Granzimas/metabolismo , Canal de Potássio Kv1.3/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Granzimas/genética , Granzimas/imunologia , Humanos , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/genética , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Ativação Linfocitária/imunologia , Bloqueadores dos Canais de Potássio/farmacologia
17.
J Immunol ; 188(12): 5877-86, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22581856

RESUMO

Increasing evidence suggests ion channels have critical functions in the differentiation and plasticity of T cells. Kv1.3, a voltage-gated K(+) channel, is a functional marker and a pharmacological target for activated effector memory T cells. Selective Kv1.3 blockers have been shown to inhibit proliferation and cytokine production by human and rat effector memory T cells. We used Kv1.3 knockout (KO) mice to investigate the mechanism by which Kv1.3 blockade affects CD4(+) T cell differentiation during an inflammatory immune-mediated disease. Kv1.3 KO animals displayed significantly lower incidence and severity of myelin oligodendrocyte glycoprotein (MOG) peptide-induced experimental autoimmune encephalomyelitis. Kv1.3 was the only K(V) channel expressed in MOG 35-55-specific CD4(+) T cell blasts, and no K(V) current was present in MOG-specific CD4(+) T cell-blasts from Kv1.3 KO mice. Fewer CD4(+) T cells migrated to the CNS in Kv1.3 KO mice following disease induction, and Ag-specific proliferation of CD4(+) T cells from these mice was impaired with a corresponding cell-cycle delay. Kv1.3 was required for optimal expression of IFN-γ and IL-17, whereas its absence led to increased IL-10 production. Dendritic cells from Kv1.3 KO mice fully activated wild-type CD4(+) T cells, indicating a T cell-intrinsic defect in Kv1.3 KO mice. The loss of Kv1.3 led to a suppressive phenotype, which may contribute to the mechanism by which deletion of Kv1.3 produces an immunotherapeutic effect. Skewing of CD4(+) T cell differentiation toward Ag-specific regulatory T cells by pharmacological blockade or genetic suppression of Kv1.3 might be beneficial for therapy of immune-mediated diseases such as multiple sclerosis.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Canal de Potássio Kv1.3/metabolismo , Ativação Linfocitária/imunologia , Linfócitos T Reguladores/imunologia , Animais , Western Blotting , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Eletrofisiologia , Encefalomielite Autoimune Experimental/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/citologia
18.
J Biol Chem ; 287(2): 1261-8, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22110135

RESUMO

The maintenance of T cell memory is critical for the development of rapid recall responses to pathogens, but may also have the undesired side effect of clonal expansion of T effector memory (T(EM)) cells in chronic autoimmune diseases. The mechanisms by which lineage differentiation of T cells is controlled have been investigated, but are not completely understood. Our previous work demonstrated a role of the voltage-gated potassium channel Kv1.3 in effector T cell function in autoimmune disease. In the present study, we have identified a mechanism by which Kv1.3 regulates the conversion of T central memory cells (T(CM)) into T(EM). Using a lentiviral-dominant negative approach, we show that loss of function of Kv1.3 mediates reversion of T(EM) into T(CM), via a delay in cell cycle progression at the G2/M stage. The inhibition of Kv1.3 signaling caused an up-regulation of SMAD3 phosphorylation and induction of nuclear p21(cip1) with resulting suppression of Cdk1 and cyclin B1. These data highlight a novel role for Kv1.3 in T cell differentiation and memory responses, and provide further support for the therapeutic potential of Kv1.3 specific channel blockers in T(EM)-mediated autoimmune diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Inibidor de Quinase Dependente de Ciclina p21/imunologia , Memória Imunológica , Canal de Potássio Kv1.3/imunologia , Transdução de Sinais/imunologia , Proteína Smad3/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/imunologia , Proteína Quinase CDC2/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/imunologia , Divisão Celular/genética , Divisão Celular/imunologia , Células Cultivadas , Ciclina B1/genética , Ciclina B1/imunologia , Ciclina B1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fase G2/genética , Fase G2/imunologia , Humanos , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Fosforilação/genética , Fosforilação/imunologia , Transdução de Sinais/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo
19.
J Neuroimmunol ; 235(1-2): 70-6, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21550672

RESUMO

The goal of this study was to investigate the effects of natalizumab therapy on the immune cell composition and phenotype in the blood of relapsing MS patients treated over the course of 12 months. We collected peripheral blood from 26 RRMS patients before treatment onset, and then 6 and 12 months after therapy. PBMC was isolated and then analyzed for phenotypic characteristics by FACS and for cytokine production by ELISA. The results of our studies showed changes in both numbers and activation states of immune cells following therapy. These changes were observed at the 6 month timepoint and generally persisted through the 12 month timepoint. The proportions of NK cells (CD3⁻CD56+) and hematopoetic stem cells (CD34+lin⁻) were increased after natalizumab treatment. Decreases were noted in numbers of CD14+ monocytes, and possibly their migratory potential, since their expression levels of α4ß1 were decreased. Relative numbers of CD20+ B cells were increased, but the proportion of CD20+ cells expressing high levels of α4ß1 integrin was decreased. While proportions of CD4+ and CD8+ T cells did not change, the percentage of cells expressing α4ß1 integrin was significantly decreased for both subsets. Natalizumab therapy produces a number of phenotypic changes in the immune composition of peripheral blood. These changes may help to explain both the mechanisms of action of natalizumab and also shed light on the potential for the observed increase in PML in these patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Imunomodulação/imunologia , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Adulto , Idoso , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/imunologia , Humanos , Imunomodulação/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Natalizumab
20.
J Immunol ; 185(12): 7161-4, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21076063

RESUMO

Th17 cells play a significant role in inflammatory and autoimmune responses. Although a number of molecular pathways that contribute to the lineage differentiation of T cells have been discovered, the mechanisms by which lineage commitment occurs are not fully understood. Transcription factors play a key role in driving T cells toward specific lineages. We have identified a role for the transcription factor Kruppel-like factor (KLF) 4 in the development of IL-17-producing CD4(+) T cells. KLF4 was required for the production of IL-17, and further, chromatin immunoprecipitation analysis demonstrated binding of KLF4 to the IL-17 promoter, indicating a direct effect on the regulation of IL-17. Further, KLF4-deficient T cells upregulated expression of retinoic acid-related orphan receptor γt similar to wild-type during the polarization process toward Th17, suggesting that these two transcription factors are regulated independently.


Assuntos
Diferenciação Celular/imunologia , Fatores de Transcrição Kruppel-Like/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Regiões Promotoras Genéticas/imunologia , Células Th17/imunologia , Regulação para Cima/imunologia , Animais , Diferenciação Celular/genética , Interleucina-17/genética , Interleucina-17/imunologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Regiões Promotoras Genéticas/genética , Células Th17/citologia , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA