Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell ; 187(8): 1990-2009.e19, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38513664

RESUMO

Multiple sclerosis (MS) is a neurological disease characterized by multifocal lesions and smoldering pathology. Although single-cell analyses provided insights into cytopathology, evolving cellular processes underlying MS remain poorly understood. We investigated the cellular dynamics of MS by modeling temporal and regional rates of disease progression in mouse experimental autoimmune encephalomyelitis (EAE). By performing single-cell spatial expression profiling using in situ sequencing (ISS), we annotated disease neighborhoods and found centrifugal evolution of active lesions. We demonstrated that disease-associated (DA)-glia arise independently of lesions and are dynamically induced and resolved over the disease course. Single-cell spatial mapping of human archival MS spinal cords confirmed the differential distribution of homeostatic and DA-glia, enabled deconvolution of active and inactive lesions into sub-compartments, and identified new lesion areas. By establishing a spatial resource of mouse and human MS neuropathology at a single-cell resolution, our study unveils the intricate cellular dynamics underlying MS.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Medula Espinal , Animais , Humanos , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Camundongos , Análise da Expressão Gênica de Célula Única , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia , Neuroglia/metabolismo , Neuroglia/patologia
2.
J Neurosci ; 35(22): 8626-39, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26041928

RESUMO

Multiple sclerosis (MS) is a demyelinating disease of the CNS characterized by inflammation and neurodegeneration. Animal models that enable the study of remyelination in the context of ongoing inflammation are greatly needed for the development of novel therapies that target the pathological inhibitory cues inherent to the MS plaque microenvironment. We report the development of an innovative animal model combining cuprizone-mediated demyelination with transfer of myelin-reactive CD4(+) T cells. Characterization of this model reveals both Th1 and Th17 CD4(+) T cells infiltrate the CNS of cuprizone-fed mice, with infiltration of Th17 cells being more efficient. Infiltration correlates with impaired spontaneous remyelination as evidenced by myelin protein expression, immunostaining, and ultrastructural analysis. Electron microscopic analysis further reveals that demyelinated axons are preserved but reduced in caliber. Examination of the immune response contributing to impaired remyelination highlights a role for peripheral monocytes with an M1 phenotype. This study demonstrates the development of a novel animal model that recapitulates elements of the microenvironment of the MS plaque and reveals an important role for T cells and peripheral monocytes in impairing endogenous remyelination in vivo. This model could be useful for testing putative MS therapies designed to enhance remyelination in the setting of active inflammation, and may also facilitate modeling the pathophysiology of denuded axons, which has been a challenge in rodents because they typically remyelinate very quickly.


Assuntos
Sistema Nervoso Central/patologia , Cuprizona/toxicidade , Doenças Desmielinizantes/terapia , Inibidores da Monoaminoxidase/toxicidade , Bainha de Mielina/metabolismo , Células Th17/fisiologia , Transferência Adotiva , Animais , Polaridade Celular/efeitos dos fármacos , Células Cultivadas , Sistema Nervoso Central/ultraestrutura , Doenças Desmielinizantes/induzido quimicamente , Modelos Animais de Doenças , Adjuvante de Freund/toxicidade , Interleucina-17/metabolismo , Antígenos Comuns de Leucócito/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/patologia , Monócitos/ultraestrutura , Proteínas da Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidade , Infiltração de Neutrófilos , Fragmentos de Peptídeos/toxicidade , Regeneração/efeitos dos fármacos , Células Th17/ultraestrutura , Fatores de Tempo
3.
Glia ; 62(9): 1513-29, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24863526

RESUMO

Nerve conduction within the mammalian central nervous system is made efficient by oligodendrocyte-derived myelin. Historically, thyroid hormones have a well described role in regulating oligodendrocyte differentiation and myelination during development; however, it remains unclear which thyroid hormone receptors are required to drive these effects. This is a question with clinical relevance since nonspecific thyroid receptor stimulation can produce deleterious side-effects. Here we report that GC-1, a thyromimetic with selective thyroid receptor ß action and a potentially limited side-effect profile, promotes in vitro oligodendrogenesis from both rodent and human oligodendrocyte progenitor cells. In addition, we used in vivo genetic fate tracing of oligodendrocyte progenitor cells via PDGFαR-CreER;Rosa26-eYFP double-transgenic mice to examine the effect of GC-1 on cellular fate and find that treatment with GC-1 during developmental myelination promotes oligodendrogenesis within the corpus callosum, occipital cortex and optic nerve. GC-1 was also observed to enhance the expression of the myelin proteins MBP, CNP and MAG within the same regions. These results indicate that a ß receptor selective thyromimetic can enhance oligodendrocyte differentiation in vitro and during developmental myelination in vivo and warrants further study as a therapeutic agent for demyelinating models.


Assuntos
Acetatos/farmacologia , Fármacos do Sistema Nervoso Central/farmacologia , Neurogênese/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Fenóis/farmacologia , Receptores beta dos Hormônios Tireóideos/agonistas , Adolescente , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Encéfalo/cirurgia , Células Cultivadas , Criança , Pré-Escolar , Epilepsia/fisiopatologia , Epilepsia/cirurgia , Substância Cinzenta/fisiopatologia , Substância Cinzenta/cirurgia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/crescimento & desenvolvimento , Nervo Óptico/fisiologia , Ratos Sprague-Dawley , Receptores beta dos Hormônios Tireóideos/metabolismo , Adulto Jovem
4.
Artigo em Inglês | MEDLINE | ID: mdl-31467038

RESUMO

OBJECTIVE: To measure the impact of JHU-083, a novel prodrug of the glutamine antagonist 6-diazo-5-oxo-l-norleucine, on immune cell proliferation and activation, along with physical and cognitive impairments associated with the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS: Splenic-derived T cells and bone marrow-derived dendritic cells (DCs) were cultured, activated, and treated daily with vehicle or JHU-083. Proliferation and activation were measured via flow cytometry and IncuCyte live cell analysis. C57BL/6 mice were immunized for EAE. Vehicle or JHU-083 was administered orally every other day either from the time of immunization in the prevention paradigm or from the time of disease onset in the treatment paradigm. Disease scores and body weight were monitored. In the treatment paradigm, cognition was evaluated using the Barnes maze test. RESULTS: JHU-083 selectively inhibits T-cell proliferation and decreases T-cell activation, with no effect on DCs. In vivo, orally administered JHU-083 significantly decreases EAE severity in both prevention and treatment paradigms and reverses EAE-induced cognitive impairment. CONCLUSIONS: JHU-083, a well-tolerated, brain penetrable glutamine antagonist, is a promising novel treatment for both the physical and cognitive deficits of MS.


Assuntos
Compostos Azo/farmacologia , Caproatos/farmacologia , Disfunção Cognitiva/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Esclerose Múltipla/tratamento farmacológico , Animais , Comportamento Animal/efeitos dos fármacos , Células Cultivadas , Disfunção Cognitiva/etiologia , Encefalomielite Autoimune Experimental/complicações , Feminino , Glutamina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/complicações
5.
J Vis Exp ; (108): 53764, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26967760

RESUMO

Efficient oligodendrogenesis is the therapeutic goal of a number of areas of research including spinal cord injury, neonatal hypoxia, and demyelinating diseases such as multiple sclerosis and transverse myelitis. Myelination is required to not only facilitate rapid impulse propagation within the central nervous system, but also to provide trophic support to underlying axons. Oligodendrocyte progenitor cells (OPCs) can be studied in vitro to help identify factors that may promote or inhibit oligodendrocyte differentiation. To date, many of the methods available to evaluate this process have either required large numbers of cells, thus limiting the number of conditions that can be investigated at any one time, or labor-intensive methods of quantification. Herein, we describe a protocol for the isolation of large numbers of highly pure OPCs together with a fast and reliable method to determine oligodendrogenesis from multiple conditions simultaneously. OPCs are isolated from P5-P7 neonatal rat cortices and grown in vitro for three days prior to differentiation. Four days after differentiation, oligodendrogenesis is evaluated using a dual-infrared fluorescence-scanning assay to determine expression of the myelin protein.


Assuntos
Axônios/metabolismo , Doenças Desmielinizantes/patologia , Oligodendroglia/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Fluorescência , Ratos
6.
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
7.
Stem Cell Res ; 9(3): 208-17, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22885102

RESUMO

Engraftment of human embryonic stem cell (hESC)-derived OPCs in animal models of demyelination results in remyelination and clinical recovery, supporting the feasibility of cell replacement therapies in promoting repair of damaged neural tissue. A critical gap in our understanding of the mechanisms associated with repair revolves around the effects of the local microenvironment on transplanted cell survival. We have determined that treatment of human ESC-derived OPCs with the pleiotropic cytokine IFN-γ promotes apoptosis that is associated with mitochondrial cytochrome c released into the cytosol with subsequent caspase 3 activation. IFN-γ-induced apoptosis is mediated, in part, by secretion of the CXC chemokine ligand 10 (CXCL10) from IFN-γ-treated cells. Signaling through the chemokine receptor CXCR2 by the ligand CXCL1 functions in a tonic manner by muting apoptosis and this is associated with reduced levels of cytosolic cytochrome c and impaired cleavage of caspase 3. These findings support a role for both IFN-γ and CXCL10 in contributing to neuropathology by promoting OPC apoptosis. In addition, these data suggest that hOPCs used for therapeutic treatment for human neurologic disease/damage are susceptible to death through exposure to local inflammatory cytokines present within the inflammatory milieu.


Assuntos
Apoptose , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Interferon gama/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Receptores de Interleucina-8B/metabolismo , Transdução de Sinais , Linhagem Celular , Quimiocina CXCL10/metabolismo , Citocromos c/metabolismo , Humanos , Receptores de Interleucina-8B/genética
8.
PLoS One ; 7(6): e39329, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22745733

RESUMO

Canonical Wnt/ß-catenin signaling has been suggested to promote self-renewal of pluripotent mouse and human embryonic stem cells. Here, we show that SB-216763, a glycogen synthase kinase-3 (GSK3) inhibitor, can maintain mouse embryonic stem cells (mESCs) in a pluripotent state in the absence of exogenous leukemia inhibitory factor (LIF) when cultured on mouse embryonic fibroblasts (MEFs). MESCs maintained with SB-216763 for one month were morphologically indistinguishable from LIF-treated mESCs and expressed pluripotent-specific genes Oct4, Sox2, and Nanog. Furthermore, Nanog immunostaining was more homogenous in SB-216763-treated colonies compared to LIF. Embryoid bodies (EBs) prepared from these mESCs expressed early-stage markers for all three germ layers, and could efficiently differentiate into cardiac-like cells and MAP2-immunoreactive neurons. To our knowledge, SB-216763 is the first GSK3 inhibitor that can promote self-renewal of mESC co-cultured with MEFs for more than two months.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Indóis/farmacologia , Maleimidas/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Imuno-Histoquímica , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Reação em Cadeia da Polimerase , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa