Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
J Immunol Res ; 2021: 8100298, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34337080

RESUMO

BACKGROUND: Spinal cord injury (SCI) remains a challenge owing to limited therapies. The exosome of neural stem cells (NSCs-Exos) and FTY720 transplantation could improve SCI effectively. However, the effect and mechanism of NSCs-Exos combined with FTY720 (FTY720-NSCs-Exos) transplantation in the treatment of SCI are not fully understood. METHODS: Sprague Dawley rats (8-week-old) were used to establish the SCI model, followed by the treatment of NSCs-Exos, FTY720, and FTY720-NSCs-Exos. The effect of FTY720, NSCs-Exos, and FTY720-NSCs-Exos combination treatment on hindlimb function, pathological changes, apoptosis activity, and the expression of spinal edema-related proteins and apoptosis-related proteins in SCI models were investigated by BBB scoring, HE staining, TUNEL staining and immunohistochemistry, and Western blotting. Meanwhile, the effect of these treatments on spinal cord microvascular endothelial cells (SCMECs) was detected under hypoxic circumstance. RESULTS: Our results found that FTY720-NSCs-Exos could alleviate pathological alterations and ameliorate the hindlimb function and oxygen insufficiency in model mice after SCI. In addition, exosomes could ameliorate the morphology of neurons, reduce inflammatory infiltration and edema, decrease the expression of Bax and AQP-4, upregulate the expression of claudin-5 and Bcl-2, and inhibit cell apoptosis. At the same time, in vitro experiments showed that FTY720-NSCs-Exos could protect the barrier of SCMECs under hypoxic circumstance, and the mechanism is related to PTEN/AKT pathway. CONCLUSION: FTY720-NSCs-Exos therapy displayed a positive therapeutic effect on SCI by regulating PTEN/AKT pathway and offered a new therapy for SCI.


Assuntos
Exossomos/transplante , Cloridrato de Fingolimode/administração & dosagem , Células-Tronco Neurais/citologia , Moduladores do Receptor de Esfingosina 1 Fosfato/administração & dosagem , Traumatismos da Medula Espinal/terapia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Modelos Animais de Doenças , Células Endoteliais , Exossomos/imunologia , Humanos , Masculino , Células-Tronco Neurais/imunologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Medula Espinal/imunologia , Medula Espinal/patologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/patologia
2.
Cells ; 10(7)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34359882

RESUMO

Scaffolds of recombinant spider silk protein (spidroin) and hyaluronic acid (HA) hydrogel hold promise in combination with cell therapy for spinal cord injury. However, little is known concerning the human immune response to these biomaterials and grafted human neural stem/progenitor cells (hNPCs). Here, we analyzed short- and long-term in vitro activation of immune cells in human peripheral blood mononuclear cells (hPBMCs) cultured with/without recombinant spidroins, HA hydrogels, and/or allogeneic hNPCs to assess potential host-donor interactions. Viability, proliferation and phenotype of hPBMCs were analyzed using NucleoCounter and flow cytometry. hPBMC viability was confirmed after exposure to the different biomaterials. Short-term (15 h) co-cultures of hPBMCs with spidroins, but not with HA hydrogel, resulted in a significant increase in the proportion of activated CD69+ CD4+ T cells, CD8+ T cells, B cells and NK cells, which likely was caused by residual endotoxins from the Escherichia coli expression system. The observed spidroin-induced hPBMC activation was not altered by hNPCs. It is resource-effective to evaluate human compatibility of novel biomaterials early in development of the production process to, when necessary, make alterations to minimize rejection risk. Here, we present a method to evaluate biomaterials and hPBMC compatibility in conjunction with allogeneic human cells.


Assuntos
Fibroínas/farmacologia , Ácido Hialurônico/farmacologia , Hidrogéis/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Aborto Legal , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Encapsulamento de Células/métodos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Feminino , Feto , Fibroínas/química , Humanos , Ácido Hialurônico/química , Hidrogéis/química , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Modelos Biológicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/imunologia , Gravidez , Primeiro Trimestre da Gravidez , Cultura Primária de Células , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Medula Espinal/citologia , Medula Espinal/imunologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/patologia
3.
Viruses ; 13(8)2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34452333

RESUMO

Viruses that infect the central nervous system (CNS) are associated with developmental abnormalities as well as neuropsychiatric and degenerative conditions. Many of these viruses such as Zika virus (ZIKV), cytomegalovirus (CMV), and herpes simplex virus (HSV) demonstrate tropism for neural stem cells (NSCs). NSCs are the multipotent progenitor cells of the brain that have the ability to form neurons, astrocytes, and oligodendrocytes. Viral infections often alter the function of NSCs, with profound impacts on the growth and repair of the brain. There are a wide spectrum of effects on NSCs, which differ by the type of virus, the model system, the cell types studied, and the age of the host. Thus, it is a challenge to predict and define the consequences of interactions between viruses and NSCs. The purpose of this review is to dissect the mechanisms by which viruses can affect survival, proliferation, and differentiation of NSCs. This review also sheds light on the contribution of key antiviral cytokines in the impairment of NSC activity during a viral infection, revealing a complex interplay between NSCs, viruses, and the immune system.


Assuntos
Doenças do Sistema Nervoso Central/virologia , Células-Tronco Neurais/virologia , Viroses/virologia , Fenômenos Fisiológicos Virais , Animais , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/imunologia , Citocinas/genética , Citocinas/imunologia , Humanos , Células-Tronco Neurais/imunologia , Vírus/genética
4.
Cells ; 10(8)2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34440761

RESUMO

Neurodegenerative diseases are characterized by the progressive loss of structure and/or function of both neurons and glial cells, leading to different degrees of pathology and loss of cognition. The hypothesis of circuit reconstruction in the damaged brain via direct cell replacement has been pursued extensively so far. In this context, stem cells represent a useful option since they provide tissue restoration through the substitution of damaged neuronal cells with exogenous stem cells and create a neuro-protective environment through the release of bioactive molecules for healthy neurons, as well. These peculiar properties of stem cells are opening to potential therapeutic strategies for the treatment of severe neurodegenerative disorders, for which the absence of effective treatment options leads to an increasingly socio-economic burden. Currently, the introduction of new technologies in the field of stem cells and the implementation of alternative cell tissues sources are pointing to exciting frontiers in this area of research. Here, we provide an update of the current knowledge about source and administration routes of stem cells, and review light and shadows of cells replacement therapy for the treatment of the three main neurodegenerative disorders (Amyotrophic lateral sclerosis, Parkinson's, and Alzheimer's disease).


Assuntos
Sistema Nervoso Central/fisiopatologia , Degeneração Neural , Regeneração Nervosa , Células-Tronco Neurais/transplante , Doenças Neurodegenerativas/cirurgia , Transplante de Células-Tronco , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/cirurgia , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/fisiopatologia , Esclerose Lateral Amiotrófica/cirurgia , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Humanos , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/fisiopatologia , Neuroimunomodulação , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , Doença de Parkinson/cirurgia , Fenótipo , Recuperação de Função Fisiológica , Transplante de Células-Tronco/efeitos adversos
5.
Front Immunol ; 12: 697203, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248998

RESUMO

Neural stem cell (NSC) therapy is a promising therapeutic strategy for stroke. Researchers have frequently carried out genetic modification or gene editing of stem cells to improve survival or therapeutic function. However, NSC transplantation carries the risk of immune rejection, and genetic modification or gene-editing might further increase this risk. For instance, recent studies have reported on manipulating the stem cell genome and transplantation via the insertion of an exogenous gene derived from magnetotactic bacteria. However, whether transgene-modified stem cells are capable of inducing immunological reactions has not been explored. Although NSCs rarely express the major histocompatibility complex (MHC), they can still cause some immunological issues. To investigate whether transgene-modified NSCs aggravate immunological responses, we detected the changes in peripheral immune organs and intracerebral astrocytes, glial cells, and MHC-I and MHC-II molecules after the injection of GFP-labeled or mms6-GFP-labeled NSCs in a rat model. Xenogeneic human embryonic kidney (HEK-293T) cells were grafted as a positive control group. Our results indicated that xenogeneic cell transplantation resulted in a strong peripheral splenic response, increased astrocytes, enhanced microglial responses, and upregulation of MHC-I and MHC-II expression on the third day of transplantation. But they decreased obviously except Iba-1 positive cells and MHC-II expression. When injection of both mms6-GFP-labeled NSCs and GFP-labeled NSCs also induced similar responses as HEK-293T cells on the third days, but MHC-I and MHC-II expression decreased 3 weeks after transplantation. In addition, mms6 transgene-modified NSCs did not produce peripheral splenic response responses as well as astrocytes, microglial cells, MHC-I and MHC-II positive cells responses when compared with non-modified NSCs. The present study provides preliminary evidence that transgenic modification does not aggravate immunological responses in NSC transplantation.


Assuntos
Células-Tronco Neurais/imunologia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Transgenes , Animais , Astrócitos/imunologia , Encéfalo/citologia , Encéfalo/imunologia , Encéfalo/cirurgia , Proliferação de Células/genética , Células Cultivadas , DNA Bacteriano/genética , Genes Bacterianos , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Rim/citologia , Rim/imunologia , Rim/cirurgia , Nanopartículas de Magnetita , Magnetossomos/genética , Magnetospirillum/genética , Microglia/imunologia , Células-Tronco Neurais/citologia , Ratos , Proteínas Recombinantes/genética , Baço/citologia , Baço/imunologia , Baço/cirurgia , Transplante de Células-Tronco/efeitos adversos , Transplante Heterólogo
7.
PLoS Negl Trop Dis ; 15(3): e0009183, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33657175

RESUMO

Global Zika virus (ZIKV) outbreaks and their strong link to microcephaly have raised major public health concerns. ZIKV has been reported to affect the innate immune responses in neural stem/progenitor cells (NS/PCs). However, it is unclear how these immune factors affect neurogenesis. In this study, we used Asian-American lineage ZIKV strain PRVABC59 to infect primary human NS/PCs originally derived from fetal brains. We found that ZIKV overactivated key molecules in the innate immune pathways to impair neurogenesis in a cell stage-dependent manner. Inhibiting the overactivated innate immune responses ameliorated ZIKV-induced neurogenesis reduction. This study thus suggests that orchestrating the host innate immune responses in NS/PCs after ZIKV infection could be promising therapeutic approach to attenuate ZIKV-associated neuropathology.


Assuntos
Imunidade Inata , Células-Tronco Neurais/virologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Encéfalo/imunologia , Encéfalo/virologia , Diferenciação Celular , Proliferação de Células , Humanos , Células-Tronco Neurais/imunologia , Neurogênese/imunologia , Replicação Viral , Infecção por Zika virus/virologia
8.
Int J Mol Sci ; 22(4)2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33572157

RESUMO

Platelet-rich plasma (PRP) is a biologic therapy that promotes healing responses across multiple medical fields, including the central nervous system (CNS). The efficacy of this therapy depends on several factors such as the donor's health status and age. This work aims to prove the effect of PRP on cellular models of the CNS, considering the differences between PRP from young and elderly donors. Two different PRP pools were prepared from donors 65‒85 and 20‒25 years old. The cellular and molecular composition of both PRPs were analyzed. Subsequently, the cellular response was evaluated in CNS in vitro models, studying proliferation, neurogenesis, synaptogenesis, and inflammation. While no differences in the cellular composition of PRPs were found, the molecular composition of the Young PRP showed lower levels of inflammatory molecules such as CCL-11, as well as the presence of other factors not found in Aged PRP (GDF-11). Although both PRPs had effects in terms of reducing neural progenitor cell apoptosis, stabilizing neuronal synapses, and decreasing inflammation in the microglia, the effect of the Young PRP was more pronounced. In conclusion, the molecular composition of the PRP, conditioned by the age of the donors, affects the magnitude of the biological response.


Assuntos
Córtex Cerebral/imunologia , Mediadores da Inflamação/metabolismo , Microglia/imunologia , Plasma Rico em Plaquetas/imunologia , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/imunologia , Animais , Apoptose/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Córtex Cerebral/citologia , Quimiocina CCL11/metabolismo , Feminino , Humanos , Masculino , Camundongos , Microglia/citologia , Células-Tronco Neurais/imunologia , Neurogênese/imunologia , Neurônios/imunologia , Plasma Rico em Plaquetas/citologia , Plasma Rico em Plaquetas/metabolismo , Cultura Primária de Células , Ratos , Sinapses/imunologia , Adulto Jovem
9.
Front Immunol ; 12: 784098, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975872

RESUMO

Following a cerebral ischemic event, substantial alterations in both cellular and molecular activities occur due to ischemia-induced cerebral pathology. Mounting evidence indicates that the robust recruitment of immune cells plays a central role in the acute stage of stroke. Infiltrating peripheral immune cells and resident microglia mediate neuronal cell death and blood-brain barrier disruption by releasing inflammation-associated molecules. Nevertheless, profound immunological effects in the context of the subacute and chronic recovery phase of stroke have received little attention. Early attempts to curtail the infiltration of immune cells were effective in mitigating brain injury in experimental stroke studies but failed to exert beneficial effects in clinical trials. Neural tissue damage repair processes include angiogenesis, neurogenesis, and synaptic remodeling, etc. Post-stroke inflammatory cells can adopt divergent phenotypes that influence the aforementioned biological processes in both endothelial and neural stem cells by either alleviating acute inflammatory responses or secreting a variety of growth factors, which are substantially involved in the process of angiogenesis and neurogenesis. To better understand the multiple roles of immune cells in neural tissue repair processes post stroke, we review what is known and unknown regarding the role of immune cells in angiogenesis, neurogenesis, and neuronal remodeling. A comprehensive understanding of these inflammatory mechanisms may help identify potential targets for the development of novel immunoregulatory therapeutic strategies that ameliorate complications and improve functional rehabilitation after stroke.


Assuntos
AVC Isquêmico/imunologia , Neovascularização Fisiológica/imunologia , Doenças Neuroinflamatórias/imunologia , Plasticidade Neuronal/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , AVC Isquêmico/patologia , Linfócitos/imunologia , Linfócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Microglia/imunologia , Microglia/metabolismo , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/metabolismo , Doenças Neuroinflamatórias/patologia , Recuperação de Função Fisiológica/imunologia
10.
Viruses ; 12(8)2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32806715

RESUMO

West Nile virus (WNV) and Usutu virus (USUV) are genetically related neurotropic mosquito-borne flaviviruses, which frequently co-circulate in nature. Despite USUV seeming to be less pathogenic for humans than WNV, the clinical manifestations induced by these two viruses often overlap and may evolve to produce severe neurological complications. The aim of this study was to investigate the effects of WNV and USUV infection on human induced pluripotent stem cell-derived neural stem cells (hNSCs), as a model of the neural progenitor cells in the developing fetal brain and in adult brain. Zika virus (ZIKV), a flavivirus with known tropism for NSCs, was used as the positive control. Infection of hNSCs and viral production, effects on cell viability, apoptosis, and innate antiviral responses were compared among viruses. WNV displayed the highest replication efficiency and cytopathic effects in hNSCs, followed by USUV and then ZIKV. In these cells, both WNV and USUV induced the overexpression of innate antiviral response genes at significantly higher levels than ZIKV. Expression of interferon type I, interleukin-1ß and caspase-3 was significantly more elevated in WNV- than USUV-infected hNSCs, in agreement with the higher neuropathogenicity of WNV and the ability to inhibit the interferon response pathway.


Assuntos
Flavivirus/patogenicidade , Imunidade Inata , Células-Tronco Neurais/virologia , Replicação Viral , Vírus do Nilo Ocidental/patogenicidade , Apoptose , Sobrevivência Celular , Células Cultivadas , Flavivirus/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas , Cinética , Células-Tronco Neurais/imunologia , Virulência , Vírus do Nilo Ocidental/fisiologia
11.
Emerg Microbes Infect ; 9(1): 1580-1589, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32576094

RESUMO

The RNA interference (RNAi) pathway directs an important antiviral immunity mechanism in plants and invertebrates. Recently, we and others have demonstrated that the antiviral RNAi response is also conserved in mammals, at least to five distinct RNA viruses, including Zika virus (ZIKV). ZIKV may preferentially infect neuronal progenitor cells (NPCs) in the developing foetal brain. Ex vivo ZIKV infection induces RNAi-mediated antiviral response in human NPCs, but not in the more differentiated NPCs or somatic cells. However, litter is known about the in vivo property or function of the virus-derived small-interfering RNAs (vsiRNAs) targeting ZIKV. Here we report a surprising observation: different from ex vivo observations, viral small RNAs (vsRNAs) targeting ZIKV were produced in vivo upon infection in both central neuron system (CNS) and muscle tissues. In addition, our findings demonstrate the production of canonical vsiRNAs in murine CNS upon antiviral RNAi activation by Sindbis virus (SINV), suggesting the possibility of antiviral immune strategy applied by mammals in the CNS.


Assuntos
Infecções por Alphavirus/genética , Alphavirus/imunologia , Células-Tronco Neurais/virologia , RNA Interferente Pequeno/metabolismo , RNA Viral/imunologia , Alphavirus/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Animais , Diferenciação Celular , Linhagem Celular , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Chlorocebus aethiops , Células HEK293 , Humanos , Camundongos , Músculo Esquelético/imunologia , Músculo Esquelético/virologia , Células-Tronco Neurais/imunologia , RNA Viral/antagonistas & inibidores , Sindbis virus/genética , Sindbis virus/imunologia , Células Vero , Replicação Viral , Zika virus/genética , Zika virus/imunologia
12.
J Neuroinflammation ; 17(1): 27, 2020 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-31954398

RESUMO

BACKGROUND: Microglia are critical mediators of neuroimmune pathology across multiple neurologic disorders. Microglia can be persistently activated or "primed" by Toll-like receptor (TLR) activation, ethanol, stress, and other insults. Thus, strategies to prevent or reverse microglial priming may be beneficial for conditions that involve progressively increasing microglial activation. Microglial depletion with repopulation is emerging as a potential therapy to normalize chronic immune activation. Primary organotypic hippocampal slice culture (OHSC) allows for the study of neuroimmune activation as well as microglial depletion and repopulation without involvement of peripheral immune activation. OHSC undergoes functional maturation and retains cytoarchitecture similar to in vivo. METHODS: OHSC underwent microglial depletion with the CSF1R antagonist PLX3397 with or without repopulation after removal of PLX3397. Immune, trophic, and synaptic gene changes in response to agonists of TLRs 2, 3, 4, 7, and 9 as well as ethanol were assessed in the settings of microglial depletion and repopulation. Gi-DREADD inhibition of microglia was used to confirm select findings seen with depletion. The ability of microglial repopulation to prevent progressive proinflammatory gene induction by chronic ethanol was also investigated. RESULTS: Microglia were depleted (> 90%) by PLX3397 in OHSC. Microglial depletion blunted proinflammatory responses to several TLR agonists as well as ethanol, which was mimicked by Gi-DREADD inhibition of OHSC microglia. Removal of PLX3397 was followed by complete repopulation of microglia. OHSCs with repopulated microglia showed increased baseline expression of anti-inflammatory cytokines (e.g., IL-10), microglial inhibitory signals (e.g., CX3CL1), and growth factors (e.g., BDNF). This was associated with blunted induction (~ 50%) of TNFα and IL-1ß in response to agonists to TLR4 and TLR7. Further, chronic cycled ethanol from 4 days in vitro (DIV) to 16DIV caused immediate 2-fold inductions of TNFα and IL-1ß that grew to ~4-fold of age-matched control slices by 40DIV. This persistent inflammatory gene expression was completely reversed by microglial depletion and repopulation after chronic ethanol. CONCLUSIONS: Microglia in OHSCs mediate proinflammatory responses to TLR agonists and ethanol. Microglial repopulation promoted an anti-inflammatory, trophic neuroenvironment and normalized proinflammatory gene expression. This supports the possibility of microglial depletion with repopulation as a strategy to reverse chronic neuroimmune activation.


Assuntos
Hipocampo/citologia , Hipocampo/imunologia , Microglia/imunologia , Microglia/metabolismo , Transdução de Sinais/imunologia , Aminopiridinas/farmacologia , Animais , Etanol/toxicidade , Hipocampo/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Microglia/efeitos dos fármacos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/metabolismo , Técnicas de Cultura de Órgãos , Pirróis/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Receptores Toll-Like/agonistas
13.
Int Immunopharmacol ; 81: 106038, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31757678

RESUMO

A failure of neurodevelopmental differentiation at the level of oligodendroglial-astrocytic biprogenitors (O2A) is shown to be involved in the pathogenesis of both multiple sclerosis (MS) and glioblastoma multiforme (GBM). In this review article, we suggest that certain antigens of Hepatitis B Virus (HBV) and HBV-Vaccine (HBV-V) could act as immune stimulants in GBM treatment based on several lines of evidence. HBV-Vs may cause rare but prominent neuroimmune side effects including demyelination and multiple sclerosis, which may be associated with HBV-proteins creating antigenic mimicry of oligodendroglial progenitors. The combined prevalance of HBV and Hepatitis C Virus-carrier state is less in patients with brain tumors compared to healthy subjects. Furthermore, within the population of patients with brain tumors, the prevalence is even about two times lesser in GBM in comparison to those with a diagnosis of meningioma. Although indirectly, this epidemiological data may indicate that the immune response triggered against hepadnavirus antigens would eliminate aberrantly differentiating O2A progenitor cells giving rise to GBMs. Moreover, Hepatitis B surface antigen-antibody variable domain is among the top 100 differentially expressed transcripts in fresh frozen and formalin-fixed paraffin-embeded specimens obtained from pediatric GBM tissues in comparison to the control brain tissues. However, the provided evidence is still premature and we think that HBV-V warrants investigation first by epidemiological studies and then by animal experiments to determine whether it reduces the risk of GBM and whether it could slow GBM growth via immune stimulation.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Encéfalo/imunologia , Glioblastoma/terapia , Anticorpos Anti-Hepatite B/imunologia , Vacinas contra Hepatite B/administração & dosagem , Animais , Astrócitos/imunologia , Encéfalo/citologia , Encéfalo/patologia , Portador Sadio/epidemiologia , Portador Sadio/imunologia , Diferenciação Celular/imunologia , Criança , Modelos Animais de Doenças , Glioblastoma/epidemiologia , Glioblastoma/imunologia , Glioblastoma/patologia , Hepatite B/epidemiologia , Hepatite B/imunologia , Antígenos de Superfície da Hepatite B/imunologia , Vacinas contra Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Humanos , Imunogenicidade da Vacina , Bainha de Mielina/imunologia , Células-Tronco Neurais/imunologia , Oligodendroglia/citologia , Oligodendroglia/imunologia , Prevalência
14.
Stem Cell Res Ther ; 10(1): 387, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31843025

RESUMO

BACKGROUND: Neural stem cells (NSCs) residing in the central nervous system play an important role in neurogenesis. Several viruses can infect these neural progenitors and cause severe neurological diseases. The innate immune responses against the neurotropic viruses in these tissue-specific stem cells remain unclear. METHODS: Human NSCs were transfected with viral RNA mimics or infected with neurotropic virus for detecting the expression of antiviral interferons (IFNs) and downstream IFN-stimulated antiviral genes. RESULTS: NSCs are able to produce interferon-ß (IFN-ß) (type I) and λ1 (type III) after transfection with poly(I:C) and that downstream IFN-stimulated antiviral genes, such as ISG56 and MxA, and the viral RNA sensors RIG-I, MDA5, and TLR3, can be expressed in NSCs under poly(I:C) or IFN-ß stimulation. In addition, our results show that the pattern recognition receptors RIG-I and MDA5, as well as the endosomal pathogen recognition receptor TLR3, but not TLR7 and TLR8, are involved in the activation of IFN-ß transcription in NSCs. Furthermore, NSCs infected with the neurotropic viruses, Zika and Japanese encephalitis viruses, are able to induce RIG-I-mediated IFN-ß expression. CONCLUSION: Human NSCs have the ability to activate IFN signals against neurotropic viral pathogens.


Assuntos
Interferon Tipo I/imunologia , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/virologia , Infecção por Zika virus/imunologia , Linhagem Celular , Células Cultivadas , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Vírus da Encefalite Japonesa (Subgrupo)/imunologia , Encefalite Japonesa/genética , Encefalite Japonesa/imunologia , Humanos , Imunidade Inata , Interferon Tipo I/biossíntese , Interferon beta/biossíntese , Interferon beta/genética , Interferon beta/imunologia , Interferons/genética , Interferons/imunologia , Células-Tronco Neurais/patologia , Receptores Imunológicos , Transcrição Gênica , Transfecção , Zika virus/imunologia , Infecção por Zika virus/genética , Infecção por Zika virus/patologia , Interferon lambda
15.
J Vis Exp ; (151)2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31609309

RESUMO

Multiple DNA modifications have been identified in the mammalian genome. Of that, 5-methylcytosine and 5-hydroxymethylcytosine-mediated epigenetic mechanisms have been intensively studied. 5-hydroxymethylcytosine displays dynamic features during embryonic and postnatal development of the brain, plays a regulatory function in gene expression, and is involved in multiple neurological disorders. Here, we describe the detailed methods including immunofluorescence staining and DNA dot-blot to detect 5-hydroxymethylcytosine in cultured cells and brain tissues of mouse.


Assuntos
5-Metilcitosina/análogos & derivados , Química Encefálica , Encéfalo/imunologia , Células-Tronco Neurais/química , Células-Tronco Neurais/imunologia , 5-Metilcitosina/análise , 5-Metilcitosina/imunologia , Fatores Etários , Animais , Linhagem Celular , Células Cultivadas , Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Immunoblotting/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
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
17.
Arch Toxicol ; 93(9): 2661-2671, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31332466

RESUMO

Bisphenol analogues including bisphenol A and its derivatives are ubiquitous environmental contaminants and have been linked to adverse neurodevelopment effects on animals and humans. Most toxicological research focused on estrogen receptor mediated pathways and did not comprehensively clarify the observed toxicity. O-GlcNAcase (OGA), the highest level in brain, plays a critical role in controlling neuronal functions at multi-levels from molecule to animal behaviors. In this work, we intend to investigate the underlying molecular mechanisms for the neurotoxicity of bisphenol analogues by identifying their cellular targets and the resultant effects. The inhibitory actions of seven bisphenol analogues on the OGA activity at molecular level were investigated by our developed electrochemical biosensor. We found that their potency varied with substituent groups, in which tetrabromo bisphenol A (TBBPA) was the strongest. The seven bisphenol analogues (0-100 µM exposure) significantly inhibited OGA activity and up-regulated protein O-GlcNAcylation level in PC12 cells. Inhibition of OGA by bisphenol analogues further induced intracellular calcium, ROS, inflammation, repressed proliferation, interfered with cell cycle, induced apoptosis. And especially, 10 µM tetrabromo bisphenol A (TBBPA) exposure could impair the growth and development of neurite in human neural stem cells (hNSCs). Molecular docking for OGA/bisphenol analogue complexes revealed the hydrophobicity-dominated inhibition potency. OGA, as a new cellular target of bisphenol analogues, would illuminate the molecular mechanism of bisphenol analogues neurotoxicity.


Assuntos
Compostos Benzidrílicos/toxicidade , Poluentes Ambientais/toxicidade , Células-Tronco Neurais/efeitos dos fármacos , Síndromes Neurotóxicas/enzimologia , Fenóis/toxicidade , beta-N-Acetil-Hexosaminidases/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Compostos Benzidrílicos/química , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Poluentes Ambientais/química , Humanos , Simulação de Acoplamento Molecular , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/imunologia , Crescimento Neuronal/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/imunologia , Células PC12 , Fenóis/química , Ratos , Espécies Reativas de Oxigênio/metabolismo
18.
Immunol Lett ; 212: 6-13, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31154052

RESUMO

In the last decade several studies employing stem cells-based therapies have been investigated as an optional treatment for multiple sclerosis. Several preclinical and few clinical studies tested the efficacy of mesenchymal stem cells as a potent candidate for such therapies. Here we suggest the option of "neuralization" of classical mesenchymal stem cells as a cellular structure that resembles neural stem cells as well as there differentiation by a unique procedure towards terminally differentiated neural cells suggesting that this cell population may be appropriate for clinical application in the CNS. We investigated whether neuralized MSC (NMSC) could promote repair and recovery after injection into mice with EAE. Injection of NMSC and differentiated NMSC starting at the onset of the chronic phase of disease improved neurological function compared to controls as well as compared to naïve MSC. Injection of NMSC and mainly differentiated correlated with a reduction in the inflammation as well as in the axonal loss/damage and reduced area of demyelination. These observations suggest that NMSC and differentiated NMSC may suggest a more potent cell-based therapy that naïve MSC in the treatment arsenal of multiple sclerosis.


Assuntos
Transdiferenciação Celular/imunologia , Fatores Estimuladores de Colônias/farmacologia , Encefalomielite Autoimune Experimental/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Esclerose Múltipla/terapia , Animais , Técnicas de Cultura de Células , Transdiferenciação Celular/efeitos dos fármacos , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Feminino , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Esclerose Múltipla/imunologia , Glicoproteína Mielina-Oligodendrócito/administração & dosagem , Glicoproteína Mielina-Oligodendrócito/imunologia , Células-Tronco Neurais/imunologia , Esferoides Celulares , Resultado do Tratamento
19.
J Neuroimmunol ; 332: 73-77, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-30959341

RESUMO

The most notable effect of prenatal Zika virus (ZIKV) infection is severe microcephaly. ZIKV has a selective tropism for neural progenitor cells; however, it is not clear what role the immune cells of the brain, microglia, may have in mitigating or exacerbating neuronal cell death following ZIKV infection. We cultured hippocampal and cortical neural cells from neonatal rat pups and infected them with ZIKV at various multiplicities of infection (MOI). We found that the neuroimmune response to ZIKV infection is composed of both pro-inflammatory and type I interferon responses and is largely dependent upon the viral dose.


Assuntos
Células-Tronco Neurais/virologia , Infecção por Zika virus/imunologia , Zika virus/patogenicidade , 2',5'-Oligoadenilato Sintetase/biossíntese , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , Feminino , Hipocampo/citologia , Interferon beta/biossíntese , Interleucina-6/biossíntese , Masculino , Microglia/imunologia , Proteínas de Resistência a Myxovirus/biossíntese , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/metabolismo , Ratos , Tropismo Viral
20.
Cell Res ; 29(4): 265-273, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30814679

RESUMO

The re-emergence of Zika virus (ZIKV) in the Western Hemisphere has resulted in global public health crisis since 2015. ZIKV preferentially infects and targets human neural progenitor cells (hNPCs) and causes fetal microcephaly upon maternal infection. hNPCs not only play critical roles during fetal brain development, but also persist in adult brain throughout life. Yet the mechanism of innate antiviral immunity in hNPCs remains largely unknown. Here, we show that ZIKV infection triggers the abundant production of virus-derived small interfering RNAs in hNPCs, but not in the more differentiated progenies or somatic cells. Ablation of key RNAi machinery components significantly enhances ZIKV replication in hNPCs. Furthermore, enoxacin, a broad-spectrum antibiotic that is known as an RNAi enhancer, exerts potent anti-ZIKV activity in hNPCs and other RNAi-competent cells. Strikingly, enoxacin treatment completely prevents ZIKV infection and circumvents ZIKV-induced microcephalic phenotypes in brain organoid models that recapitulate human fetal brain development. Our findings highlight the physiological importance of RNAi-mediated antiviral immunity during the early stage of human brain development, uncovering a novel strategy to combat human congenital viral infections through enhancing RNAi.


Assuntos
Encéfalo/imunologia , Células-Tronco Neurais/imunologia , Organoides/imunologia , RNA Viral/imunologia , Infecção por Zika virus/imunologia , Zika virus/genética , Animais , Antivirais/farmacologia , Encéfalo/patologia , Linhagem Celular , Enoxacino/farmacologia , Humanos , Imunidade Inata , Células-Tronco Neurais/patologia , Organoides/patologia , Interferência de RNA , Replicação Viral , Zika virus/imunologia , Zika virus/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...