Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Front Immunol ; 12: 730128, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34552593

RESUMO

Several types of cancer grow differently depending on the environmental stimuli they receive. In glioma, exposure to an enriched environment (EE) increases the overall survival rate of tumor-bearing mice, acting on the cells that participate to define the tumor microenvironment. In particular, environmental cues increase the microglial production of interleukin (IL)-15 which promotes a pro-inflammatory (antitumor) phenotype of microglia and the cytotoxic activity of natural killer (NK) cells, counteracting glioma growth, thus representing a virtuous mechanism of interaction between NK cells and microglia. To mimic the effect of EE on glioma, we investigated the potential of creating engineered microglia as the source of IL-15 in glioma. We demonstrated that microglia modified with recombinant adeno-associated virus serotype 2 (rAAV2) carrying IL-15 (rAAV2-IL-15), to force the production of IL-15, are able to increase the NK cells viability in coculture. Furthermore, the intranasal delivery of rAAV2-IL-15 microglia triggered the interplay with NK cells in vivo, enhancing NK cell recruitment and pro-inflammatory microglial phenotype in tumor mass of glioma-bearing mice, and ultimately counteracted tumor growth. This approach has a high potential for clinical translatability, highlighting the therapeutic efficacy of forced IL-15 production in microglia: the delivery of engineered rAAV2-IL-15 microglia to boost the immune response paves the way to design a new perspective therapy for glioma patients.


Assuntos
Neoplasias Encefálicas/terapia , Dependovirus/metabolismo , Terapia Genética , Glioma/terapia , Imunoterapia , Interleucina-15/metabolismo , Microglia/transplante , Microambiente Tumoral , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Citotoxicidade Imunológica , Dependovirus/genética , Dependovirus/imunologia , Engenharia Genética , Glioma/genética , Glioma/imunologia , Glioma/metabolismo , Interleucina-15/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/imunologia , Microglia/metabolismo , Fenótipo , Transdução Genética , Carga Tumoral
2.
Mol Neurodegener ; 16(1): 50, 2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34301296

RESUMO

BACKGROUND: Disease-associated microglia (DAMs), that surround beta-amyloid plaques, represent a transcriptionally-distinct microglial profile in Alzheimer's disease (AD). Activation of DAMs is dependent on triggering receptor expressed on myeloid cells 2 (TREM2) in mouse models and the AD TREM2-R47H risk variant reduces microglial activation and plaque association in human carriers. Interestingly, TREM2 has also been identified as a microglial lipid-sensor, and recent data indicates lipid droplet accumulation in aged microglia, that is in turn associated with a dysfunctional proinflammatory phenotype. However, whether lipid droplets (LDs) are present in human microglia in AD and how the R47H mutation affects this remains unknown. METHODS: To determine the impact of the TREM2 R47H mutation on human microglial function in vivo, we transplanted wild-type and isogenic TREM2-R47H iPSC-derived microglial progenitors into our recently developed chimeric Alzheimer mouse model. At 7 months of age scRNA-seq and histological analyses were performed. RESULTS: Here we report that the transcriptome of human wild-type TREM2 and isogenic TREM2-R47H DAM xenografted microglia (xMGs), isolated from chimeric AD mice, closely resembles that of human atherosclerotic foam cells. In addition, much like foam cells, plaque-bound xMGs are highly enriched in lipid droplets. Somewhat surprisingly and in contrast to a recent in vitro study, TREM2-R47H mutant xMGs exhibit an overall reduction in the accumulation of lipid droplets in vivo. Notably, TREM2-R47H xMGs also show overall reduced reactivity to plaques, including diminished plaque-proximity, reduced CD9 expression, and lower secretion of plaque-associated APOE. CONCLUSIONS: Altogether, these results indicate lipid droplet accumulation occurs in human DAM xMGs in AD, but is reduced in TREM2-R47H DAM xMGs, as it occurs secondary to TREM2-mediated changes in plaque proximity and reactivity.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Gotículas Lipídicas/patologia , Glicoproteínas de Membrana , Microglia/patologia , Receptores Imunológicos , Animais , Quimera , Modelos Animais de Doenças , Xenoenxertos , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Microglia/transplante , Receptores Imunológicos/genética
3.
STAR Protoc ; 2(3): 100665, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34308380

RESUMO

Mutations in microglia may cause brain disorders. Replacement of dysfunctional microglia by allogeneic wild-type microglia from bone marrow transplantation (Mr BMT) or peripheral blood can correct the gene deficiency at the brain-wide scale but cannot achieve precise replacement at specific brain regions. Here, we introduce a strategy with potential clinical relevance-microglia replacement by microglia transplantation (Mr MT), combining tamoxifen-induced ablation of Mr BMT cells and intracranial injection of microglia to mouse brain, to achieve region-sepcific microglia replacement. The original abbreviation of this microglia replacement strategy is mrMT. We hereby change the name to Mr MT. For complete details on the use and execution of this protocol, please refer to Xu et al. (2020).


Assuntos
Encéfalo/citologia , Microglia/transplante , Transplante de Tecidos/métodos , Animais , Transplante de Medula Óssea/métodos , Encéfalo/fisiologia , Receptor 1 de Quimiocina CX3C/genética , Feminino , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Tamoxifeno/farmacologia , Transplante de Tecidos/instrumentação , Transplantes
4.
Mol Ther ; 29(4): 1439-1458, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33309882

RESUMO

Contrasting myelin damage through the generation of new myelinating oligodendrocytes represents a promising approach to promote functional recovery after stroke. Here, we asked whether activation of microglia and monocyte-derived macrophages affects the regenerative process sustained by G protein-coupled receptor 17 (GPR17)-expressing oligodendrocyte precursor cells (OPCs), a subpopulation of OPCs specifically reacting to ischemic injury. GPR17-iCreERT2:CAG-eGFP reporter mice were employed to trace the fate of GPR17-expressing OPCs, labeled by the green fluorescent protein (GFP), after permanent middle cerebral artery occlusion. By microglia/macrophages pharmacological depletion studies, we show that innate immune cells favor GFP+ OPC reaction and limit myelin damage early after injury, whereas they lose their pro-resolving capacity and acquire a dystrophic "senescent-like" phenotype at later stages. Intracerebral infusion of regenerative microglia-derived extracellular vesicles (EVs) restores protective microglia/macrophages functions, limiting their senescence during the post-stroke phase, and enhances the maturation of GFP+ OPCs at lesion borders, resulting in ameliorated neurological functionality. In vitro experiments show that EV-carried transmembrane tumor necrosis factor (tmTNF) mediates the pro-differentiating effects on OPCs, with future implications for regenerative therapies.


Assuntos
Senescência Celular/genética , Bainha de Mielina/genética , Receptores Acoplados a Proteínas G/genética , Acidente Vascular Cerebral/terapia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Diferenciação Celular/genética , Linhagem Celular , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/terapia , Macrófagos/metabolismo , Macrófagos/transplante , Masculino , Camundongos , Microglia/metabolismo , Microglia/transplante , Oligodendroglia/transplante , Medicina Regenerativa/métodos , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Fator de Necrose Tumoral alfa/genética
5.
Int J Mol Sci ; 21(17)2020 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-32867222

RESUMO

Stroke remains a major cause of serious disability because the brain has a limited capacity to regenerate. In the last two decades, therapies for stroke have dramatically changed. However, half of the patients cannot achieve functional independence after treatment. Presently, cell-based therapies are being investigated to improve functional outcomes. This review aims to describe conventional cell therapies under clinical trial and outline the novel concept of polarized cell therapies based on protective cell phenotypes, which are currently in pre-clinical studies, to facilitate functional recovery after post-reperfusion treatment in patients with ischemic stroke. In particular, non-neuronal stem cells, such as bone marrow-derived mesenchymal stem/stromal cells and mononuclear cells, confer no risk of tumorigenesis and are safe because they do not induce rejection and allergy; they also pose no ethical issues. Therefore, recent studies have focused on them as a cell source for cell therapies. Some clinical trials have shown beneficial therapeutic effects of bone marrow-derived cells in this regard, whereas others have shown no such effects. Therefore, more clinical trials must be performed to reach a conclusion. Polarized microglia or peripheral blood mononuclear cells might provide promising therapeutic strategies after stroke because they have pleiotropic effects. In traumatic injuries and neurodegenerative diseases, astrocytes, neutrophils, and T cells were polarized to the protective phenotype in pre-clinical studies. As such, they might be useful therapeutic targets. Polarized cell therapies are gaining attention in the treatment of stroke and neurological diseases.


Assuntos
AVC Isquêmico/terapia , Leucócitos Mononucleares/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Doenças do Sistema Nervoso/terapia , Animais , Polaridade Celular , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos , AVC Isquêmico/fisiopatologia , Leucócitos Mononucleares/citologia , Células-Tronco Mesenquimais/citologia , Microglia/citologia , Microglia/transplante , Doenças do Sistema Nervoso/fisiopatologia , Recuperação de Função Fisiológica , Resultado do Tratamento
6.
Cell Rep ; 32(6): 108041, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32783928

RESUMO

Microglia are important immune cells in the central nervous system (CNS). Dysfunctions of gene-deficient microglia contribute to the development and progression of multiple CNS diseases. Microglia replacement by nonself cells has been proposed to treat microglia-associated disorders. However, some attempts have failed due to low replacement efficiency, such as with the traditional bone marrow transplantation approach. In this study, we develop three efficient strategies for microglia replacement: microglia replacement by bone marrow transplantation (mrBMT), microglia replacement by peripheral blood (mrPB), and microglia replacement by microglia transplantation (mrMT). mrBMT and mrPB allow microglia-like cells to efficiently replace resident microglia in the whole CNS. On the other hand, mrMT achieves microglia replacement in brain regions of interest. In summary, the present study offers effective tactics for microglia replacement with diverse application scenarios, which potentially opens up a window on treating microglia-associated CNS disorders.


Assuntos
Sistema Nervoso Central/fisiopatologia , Microglia/transplante , Animais , Sistema Nervoso Central/cirurgia , Humanos , Camundongos
7.
Am J Alzheimers Dis Other Demen ; 35: 1533317520927169, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32536247

RESUMO

Alzheimer's disease (AD) is the most common form of dementia, which is characterized by a progressive cognitive decline and senile plaques formed by amyloid ß (Aß). Microglia are the immune cells of the central nervous system (CNS). Studies have proposed 2 types of microglia, namely, the resident microglia and bone marrow-derived microglia (BMDM). Recent studies suggested that BMDM, not the resident microglia, can phagocytose Aß, which has a great therapeutic potential in AD. Bone marrow-derived microglia can populate the CNS in an efficient manner and their functions can be regulated by some genes. Thus, methods that increase their recruitment and phagocytosis could be used as a new tool that clears Aß and ameliorates cognitive impairment. Herein, we review the neuroprotective functions of BMDM and their therapeutic potential in AD.


Assuntos
Doença de Alzheimer/terapia , Células da Medula Óssea/citologia , Microglia/citologia , Microglia/transplante , Neuroproteção , Fagocitose , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Humanos , Microglia/imunologia , Placa Amiloide/metabolismo
8.
Nat Commun ; 11(1): 1577, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32221280

RESUMO

Microglia, the brain-resident macrophages, exhibit highly dynamic functions in neurodevelopment and neurodegeneration. Human microglia possess unique features as compared to mouse microglia, but our understanding of human microglial functions is largely limited by an inability to obtain human microglia under homeostatic states. Here, we develop a human pluripotent stem cell (hPSC)-based microglial chimeric mouse brain model by transplanting hPSC-derived primitive macrophage progenitors into neonatal mouse brains. Single-cell RNA-sequencing of the microglial chimeric mouse brains reveals that xenografted hPSC-derived microglia largely retain human microglial identity, as they exhibit signature gene expression patterns consistent with physiological human microglia and recapitulate heterogeneity of adult human microglia. Importantly, the engrafted hPSC-derived microglia exhibit dynamic response to cuprizone-induced demyelination and species-specific transcriptomic differences in the expression of neurological disease-risk genes in microglia. This model will serve as a tool to study the role of human microglia in brain development and degeneration.


Assuntos
Encéfalo/citologia , Diferenciação Celular , Quimera/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Microglia/citologia , Animais , Linhagem Celular , Cuprizona , Doenças Desmielinizantes/patologia , Feminino , Humanos , Imageamento Tridimensional , Camundongos , Microglia/transplante , RNA-Seq , Análise de Célula Única , Transcriptoma/genética
10.
CNS Neurosci Ther ; 26(1): 55-65, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31087449

RESUMO

BACKGROUND: Neural stem cells (NSCs) transplantation is considered a promising treatment for Parkinson's disease. But most NSCs are differentiated into glial cells rather than neurons, and only a few of them survive after transplantation due to the inflammatory environment. METHODS: In this study, neural stem cells (NSCs) and microglial cells both forced with the Nurr1 gene were transplanted into the striatum of the rat model of PD. The results were evaluated through reverse transcription polymerase chain reaction (RT-PCR), Western blot, and immunofluorescence analysis. RESULTS: The behavioral abnormalities of PD rats were improved by combined transplantation of NSCs and microglia, both forced with Nurr1. The number of tyrosine hydroxylase+ cells in the striatum of PD rats increased, and the number of Iba1+ cells decreased compared with the other groups. Moreover, the dopamine neurons differentiated from grafted NSCs could still be detected in the striatum of PD rats after 5 months. CONCLUSIONS: The results suggested that transplantation of Nurr1-overexpressing NSCs and microglia could improve the inhospitable host brain environments, which will be  a new potential strategy for the cell replacement therapy in PD.


Assuntos
Terapia Genética/métodos , Microglia/transplante , Células-Tronco Neurais/transplante , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Transtornos Parkinsonianos/terapia , Transplante de Células-Tronco/métodos , Anfetamina , Animais , Comportamento Animal , Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular , Corpo Estriado/cirurgia , Neurônios Dopaminérgicos/transplante , Encefalite/terapia , Feminino , Hidroxidopaminas , Masculino , Proteínas dos Microfilamentos/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/biossíntese , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/psicologia , Ratos , Ratos Sprague-Dawley
11.
Mol Ther ; 28(1): 254-265, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31604678

RESUMO

Despite the poor prognosis of spinal cord injury (SCI), effective treatments are lacking. Diverse factors regulate SCI prognosis. In this regard, microglia play crucial roles depending on their phenotype. The M1 phenotype exacerbates neuroinflammation, whereas the M2 phenotype promotes tissue repair and provides anti-inflammatory effects. Therefore, we compared the effects of M2 and M1 microglia transplantation on SCI. First, we established a method for effective induction of M1 or M2 microglia by exposure to granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin (IL)-4, respectively, to be used for transplantation in a SCI mouse model. In the M2 microglia transplantation group, significant recovery of motor function was observed compared with the control and M1 groups. Elevated transcription of several neuroprotective molecules including mannose receptor C type 1 (Mrc1), arginase 1 (Arg1), and insulin-like growth factor 1 (Igf1) was observed. Moreover, intramuscular injection of FluoroRuby dye revealed recovery of retrograde axonal transport from the neuromuscular junction to upstream of the injured spinal cord only in the M2-transplanted group, although the number of migrated microglia were comparable in both M1 and M2 groups. In conclusion, our results indicated that M2 microglia obtained by IL-4 stimulation may be a promising candidate for cell transplantation therapy for SCI.


Assuntos
Transplante de Células/métodos , Microglia/transplante , Fenótipo , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/terapia , Animais , Animais Recém-Nascidos , Comportamento Animal , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-4/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Atividade Motora , Resultado do Tratamento
12.
J Alzheimers Dis ; 73(1): 413-429, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31796681

RESUMO

Amyloid-ß (Aß) accumulation in the brain triggers the onset of Alzheimer's disease (AD), and its prevention and elimination are high priorities for anti-AD therapeutic strategies. Microglia, the resident immune cells in the brain, promote Aß clearance by phagocytosis. Previously, we demonstrated that injection of primary cultured rat microglia and mouse bone marrow-derived microglia-like cells into the brain decreases the level of Aß and that intrahippocampal injection of these cells ameliorates cognitive impairment in a mouse model of AD. To advance this cell therapeutic strategy to the clinical stage, less invasive ways of preparing autologous microglia-like cells from elderly patients are required. In this study, we demonstrated that hematopoietic stem cells mobilized from the bone marrow to peripheral blood by administering granulocyte colony-stimulating factor and a CXCR4 antagonist to mice differentiated into microglia-like cells upon stimulation with colony-stimulating factor 1 and interleukin-34. The peripheral blood-derived microglia-like (PBDML) cells expressed microglial markers and engaged in Aß phagocytosis. Although PBDML cells were in an anti-inflammatory state under nonstimulated conditions, they expressed mRNAs encoding proinflammatory cytokines following lipopolysaccharide treatment. PBDML cells injected into the hippocampi of a mouse AD model survived for at least 36 days while phagocytosing Aß, contributed to a reduction in brain Aß burden, and ameliorated cognitive impairment in the mice. These results strongly suggest that PBDML cells are a promising source for the development of a novel cell therapy against AD.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Disfunção Cognitiva/terapia , Microglia/transplante , Doença de Alzheimer/psicologia , Animais , Disfunção Cognitiva/psicologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Células-Tronco Hematopoéticas , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Cultura Primária de Células , Ratos , Receptores CXCR4/antagonistas & inibidores , Reconhecimento Psicológico , Análise de Sobrevida
13.
Proc Natl Acad Sci U S A ; 116(50): 25293-25303, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31772018

RESUMO

Microglia are essential for maintenance of normal brain function, with dysregulation contributing to numerous neurological diseases. Protocols have been developed to derive microglia-like cells from human induced pluripotent stem cells (hiPSCs). However, primary microglia display major differences in morphology and gene expression when grown in culture, including down-regulation of signature microglial genes. Thus, in vitro differentiated microglia may not accurately represent resting primary microglia. To address this issue, we transplanted microglial precursors derived in vitro from hiPSCs into neonatal mouse brains and found that the cells acquired characteristic microglial morphology and gene expression signatures that closely resembled primary human microglia. Single-cell RNA-sequencing analysis of transplanted microglia showed similar cellular heterogeneity as primary human cells. Thus, hiPSCs-derived microglia transplanted into the neonatal mouse brain assume a phenotype and gene expression signature resembling that of resting microglia residing in the human brain, making chimeras a superior tool to study microglia in human disease.


Assuntos
Encéfalo/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Microglia/transplante , Animais , Encéfalo/metabolismo , Encéfalo/cirurgia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Microglia/metabolismo , Fenótipo
14.
Nat Neurosci ; 22(12): 2111-2116, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31659342

RESUMO

Although genetics highlights the role of microglia in Alzheimer's disease, one-third of putative Alzheimer's disease risk genes lack adequate mouse orthologs. Here we successfully engraft human microglia derived from embryonic stem cells in the mouse brain. The cells recapitulate transcriptionally human primary microglia ex vivo and show expression of human-specific Alzheimer's disease risk genes. Oligomeric amyloid-ß induces a divergent response in human versus mouse microglia. This model can be used to study the role of microglia in neurological diseases.


Assuntos
Doença de Alzheimer/genética , Células-Tronco Embrionárias/citologia , Microglia/metabolismo , Microglia/transplante , Transcriptoma , Peptídeos beta-Amiloides/farmacologia , Animais , Diferenciação Celular , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos
15.
Brain ; 142(11): 3456-3472, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31529023

RESUMO

The immunological barrier currently precludes the clinical utilization of allogeneic stem cells. Although glial-restricted progenitors have become attractive candidates to treat a wide variety of neurological diseases, their survival in immunocompetent recipients is limited. In this study, we adopted a short-term, systemically applicable co-stimulation blockade-based strategy using CTLA4-Ig and anti-CD154 antibodies to modulate T-cell activation in the context of allogeneic glial-restricted progenitor transplantation. We found that co-stimulation blockade successfully prevented rejection of allogeneic glial-restricted progenitors from immunocompetent mouse brains. The long-term engrafted glial-restricted progenitors myelinated dysmyelinated adult mouse brains within one month. Furthermore, we identified a set of plasma miRNAs whose levels specifically correlated to the dynamic changes of immunoreactivity and as such could serve as biomarkers for graft rejection or tolerance. We put forward a successful strategy to induce alloantigen-specific hyporesponsiveness towards stem cells in the CNS, which will foster effective therapeutic application of allogeneic stem cells.


Assuntos
Tolerância Imunológica , Microglia/imunologia , Microglia/transplante , Bainha de Mielina , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Transferência Adotiva , Aloenxertos , Animais , Citocinas/biossíntese , Rejeição de Enxerto , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Linfócitos T/imunologia , Transplante Homólogo
16.
Glia ; 67(4): 729-740, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30485546

RESUMO

Microglia, which are pathological effectors and amplifiers in the central nervous system, undergo various forms of activation. A well-studied microglial-induced pathological paradigm, spinal microglial activation following peripheral nerve injury (PNI), is a key event for the development of neuropathic pain but the transcription factors contributing to microglial activation are less understood. Herein, we demonstrate that MafB, a dominant transcriptional regulator of mature microglia, is involved in the pathology of a mouse model of neuropathic pain. PNI caused a rapid and marked increase of MafB expression selectively in spinal microglia but not in neurons. We also found that the microRNA mir-152 in the spinal cord which targets MafB expression decreased after PNI, and intrathecal administration of mir-152 mimic suppressed the development of neuropathic pain. Reduced MafB expression using heterozygous Mafb deficient mice and by intrathecal administration of siRNA alleviated the development of PNI-induced mechanical hypersensitivity. Furthermore, we found that intrathecal transfer of Mafb deficient microglia did not induce mechanical hypersensitivity and that conditional Mafb knockout mice did not develop neuropathic pain after PNI. We propose that MafB is a key mediator of the PNI-induced phenotypic alteration of spinal microglia and neuropathic pain development.


Assuntos
Regulação da Expressão Gênica/genética , Fator de Transcrição MafB/metabolismo , Microglia/metabolismo , Neuralgia/patologia , Medula Espinal/patologia , Animais , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hiperalgesia/genética , Hiperalgesia/fisiopatologia , Fator de Transcrição MafB/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Microglia/transplante , Neuralgia/tratamento farmacológico , Limiar da Dor/fisiologia , Fosfopiruvato Hidratase/genética , Fosfopiruvato Hidratase/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/uso terapêutico
17.
Curr Opin Pharmacol ; 44: 8-14, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30472550

RESUMO

Despite multiple efforts to find treatments, prion diseases are still incurable. The currently available therapeutic strategies are mostly based on compounds to inhibit pathological PrP (PrPSc) accumulation, and cellular PrP (PrPC) conversion into PrPSc. However, they cannot reverse the pathological changes already present in the brain. Cell-based therapeutic strategies could promote the repair of the pre-existing brain damage. The few available data come mostly from preclinical studies using neural stem cells, bone marrow-derived microglia and mesenchymal stem cells, as cell sources. Moreover, the benefits of cell-based therapeutic strategies could be linked not only to the replacement of damaged cells, but also to the secretion of trophic factors by the grafted cells that might modulate inflammation, cell death, or endogenous neurogenesis.


Assuntos
Doenças Priônicas/terapia , Animais , Humanos , Transplante de Células-Tronco Mesenquimais , Microglia/transplante , Células-Tronco Neurais/transplante
18.
Braz J Med Biol Res ; 51(10): e7076, 2018 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-30066721

RESUMO

This study evaluated the effect of microglia transplantation on neurological functional recovery in rats subjected to traumatic spinal cord injury (SCI). The rat model of SCI was established using a weight drop device. Forty SCI rats were randomly divided into the microglia group and the saline group. Then, rat-derived microglial cells or normal saline was injected into the injured site 7 days after surgery. The Basso-Beattie-Bresnahan (BBB) score, inclined plate test, and motor-evoked potentials (MEPs) were applied to assess the recovery of motor function. Hematoxylin and eosin (H&E) staining was used to assess the therapeutic effect. Microglia transplantation significantly improved BBB scores and functional scores at 2, 3, 4, 6, and 8 weeks after surgery compared to saline injection (P<0.05). Meanwhile, a prolonged MEP latency and decreased MEP amplitude were observed at 4 and 8 weeks in the microglia group (P<0.05). Histological analysis showed less damage and better prognosis in SCI rats of the microglia group. BrdU+ cell tracing experiments showed that microglia were recruited to the injured area of the spinal cord at 7 and 14 days after transplantation. The intensity of immunofluorescence was increased in CD68+ and OX42+ microglia at 2 days, 1 week, and 2 weeks, and then decreased at 3 and 4 weeks after transplantation in the microglia group. The transplantation of activated microglia played a key role in promoting the recovery of spinal cord function in a rat model of SCI.


Assuntos
Microglia/transplante , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/cirurgia , Animais , Modelos Animais de Doenças , Feminino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Traumatismos da Medula Espinal/patologia , Fatores de Tempo
19.
Neuron ; 99(3): 464-479.e7, 2018 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30033154

RESUMO

Repeated environmental stress has been proposed to induce neural inflammation together with depression and anxiety. Innate immune receptors, such as Toll-like receptors (TLRs), are activated by exogenous or endogenous ligands to evoke inflammation. Here we show that the loss of TLR2 and TLR4 (TLR2/4) abolished repeated social defeat stress (R-SDS)-induced social avoidance and anxiety in mice. TLR2/4 deficiency mitigated R-SDS-induced neuronal response attenuation, dendritic atrophy, and microglial activation in the medial prefrontal cortex (mPFC). Furthermore, mPFC microglia-specific TLR2/4 knockdown blocked social avoidance. Transcriptome analyses revealed that R-SDS induced IL-1α and TNF-α in mPFC microglia in a TLR2/4-dependent manner, and antibody blockade of these cytokines in the mPFC suppressed R-SDS-induced social avoidance. These results identify TLR2/4 as crucial mediators of R-SDS-induced microglial activation in the mPFC, which leads to neuronal and behavioral changes through inflammation-related cytokines, highlighting unexpected pivotal roles of innate immunity in the mPFC in repeated environmental stress-induced behavioral changes. VIDEO ABSTRACT.


Assuntos
Aprendizagem da Esquiva/fisiologia , Microglia/metabolismo , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/metabolismo , Receptor 2 Toll-Like/deficiência , Receptor 4 Toll-Like/deficiência , Animais , Células Cultivadas , Células HEK293 , Humanos , Imunidade Inata/fisiologia , Relações Interpessoais , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/imunologia , Microglia/transplante , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/imunologia , Estresse Psicológico/imunologia , Estresse Psicológico/psicologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia
20.
J Control Release ; 286: 114-124, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30026082

RESUMO

Degeneration of dopamine (DA) neurons in the substantia nigra pars compacta is the pathological hallmark of Parkinson's disease (PD). In PD multiple pathogenic mechanisms initiate and drive this neurodegenerative process, making the development of effective treatments challenging. To date, PD patients are primarily treated with dopaminergic drugs able to temporarily enhance DA levels, therefore relieving motor symptoms. However, the drawbacks of these therapies including the inability to alter disease progression are constantly supporting the search for alternative treatment approaches. Over the past years efforts have been put into the development of new therapeutic strategies based on the delivery of therapeutic genes using viral vectors or transplantation of DA neurons for cell-based DA replacement. Here, past achievements and recent advances in gene- and cell-based therapies for PD are outlined. We discuss how current gene and cell therapy strategies hold great promise for the treatment of PD and how the use of stem cells and recent developments in cellular reprogramming could contribute to open a new avenue in PD therapy.


Assuntos
Técnicas de Reprogramação Celular/métodos , Terapia Genética/métodos , Doença de Parkinson/terapia , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Astrócitos/transplante , Reprogramação Celular , Dopamina/genética , Dopamina/metabolismo , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/transplante , Humanos , Microglia/citologia , Microglia/metabolismo , Microglia/transplante , Doença de Parkinson/genética , Doença de Parkinson/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...