Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
1.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 635-640, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38948290

ABSTRACT

Objective: Intracerebral hemorrhage (ICH), the second most common type of stroke, can cause long-lasting disability in the afflicted patients. The study was conducted to examine the patterns of change in endogenous neural stem cells (eNSCs) and in the regenerative microenvironment after ICH, to observe the relationship between the migration of eNSCs and the pattern of change in the polarization state of immune cells in the microenvironment, and provide a research basis for research on clinical nerve repair. Methods: The collagenase injection method was used for modeling. The ICH model was induced in adult female Sprague-Dawley (SD) rats by injecting type VII collagenase (2 U) into the brain tissue of rats. All the experimental rats weighed 280-300 g. In order to simulate the ICU at different time points, including the acute phase (within 1 week), subacute phase (1-3 weeks), and the chronic phase (over 3 weeks), brain tissues were harvested at 3 day post injection (3 DPI), 10 DPI, 20 DPI, and 30 DPI to evaluate the modeling effect. Immunofluorescence staining of the brain tissue sections was performed with DCX antibody to observe the pattern of change in the migration of eNSCs in the brain tissue at different time points. Immunofluorescence staining of brain tissue sections was performed with CD206 antibody and CD86 antibody for respective observation of the pattern of change in pro-inflammatory (M1-type) and anti-inflammatory (M2-type) immune cells in the regenerative microenvironment of the brain tissue after ICM. Results: Spontaneous ICH was successfully induced by injecting type Ⅶ collagenase into the brain tissue of SD rats. The volume of the hematoma formed started to gradually increase at 3 DPI and reached its maximum at 10 DPI. After that, the hematoma was gradually absorbed and was completely absorbed by 30 DPI. Analysis of the pattern of changes in eNSCs in the brain tissue showed that a small number of eNSCs were activated at 3 DPI, but very soon their number started to decrease. By 10 DPI, eNSCs gradually began to increase. A large number of eNSCs migrated to the hemorrhage site at 20 DPI. Then the number of eNSCs decreased significantly at 30 DPI (P<0.01). Analysis of the immune microenvironment of the brain tissue showed that pro-inflammatory (M1 type) immune cells increased significantly at 10 and 20 DPI (P<0.01) and decreased at 30 DPI. Anti-inflammatory (M2 type) immune cells began to increase gradually at 3 DPI, decreased significantly at 20 DPI (P<0.05), and then showed an increase at 30 DPI. Conclusion: After ICH in rats, eNSCs migrating toward the site of ICH first increase and then decrease. The immune microenvironment demonstrates a pattern of change in which inflammation is suppressed at first, then promoted, and finally suppressed again. Inflammation may have a stimulatory effect on the migration of eNSCs, but excessive inflammatory activation has an inhibitory effect on the differentiation and further activation of eNSCs. After ICH, the early stage of repair and protection (10 d) and the subacute phase (20 d) may provide the best opportunities for intervention.


Subject(s)
Cell Movement , Cerebral Hemorrhage , Doublecortin Protein , Neural Stem Cells , Rats, Sprague-Dawley , Animals , Cerebral Hemorrhage/immunology , Rats , Female , Neural Stem Cells/immunology , Neural Stem Cells/cytology , Disease Models, Animal , Phenotype , Brain/immunology , Brain/pathology , Macrophages/immunology
2.
J Immunol Res ; 2021: 8100298, 2021.
Article in English | MEDLINE | ID: mdl-34337080

ABSTRACT

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.


Subject(s)
Exosomes/transplantation , Fingolimod Hydrochloride/administration & dosage , Neural Stem Cells/cytology , Sphingosine 1 Phosphate Receptor Modulators/administration & dosage , Spinal Cord Injuries/therapy , Animals , Apoptosis/drug effects , Apoptosis/immunology , Disease Models, Animal , Endothelial Cells , Exosomes/immunology , Humans , Male , Neural Stem Cells/immunology , Neurons/drug effects , Neurons/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/immunology , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
3.
Cells ; 10(7)2021 07 06.
Article in English | MEDLINE | ID: mdl-34359882

ABSTRACT

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.


Subject(s)
Fibroins/pharmacology , Hyaluronic Acid/pharmacology , Hydrogels/pharmacology , Neural Stem Cells/drug effects , Spinal Cord/drug effects , Abortion, Legal , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Encapsulation/methods , Cell Proliferation/drug effects , Cell Survival/drug effects , Coculture Techniques , Female , Fetus , Fibroins/chemistry , Humans , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Models, Biological , Neural Stem Cells/cytology , Neural Stem Cells/immunology , Pregnancy , Pregnancy Trimester, First , Primary Cell Culture , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Spinal Cord/cytology , Spinal Cord/immunology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
4.
Viruses ; 13(8)2021 07 27.
Article in English | MEDLINE | ID: mdl-34452333

ABSTRACT

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.


Subject(s)
Central Nervous System Diseases/virology , Neural Stem Cells/virology , Virus Diseases/virology , Virus Physiological Phenomena , Animals , Central Nervous System Diseases/genetics , Central Nervous System Diseases/immunology , Cytokines/genetics , Cytokines/immunology , Humans , Neural Stem Cells/immunology , Viruses/genetics
5.
Cells ; 10(8)2021 08 05.
Article in English | MEDLINE | ID: mdl-34440761

ABSTRACT

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).


Subject(s)
Central Nervous System/physiopathology , Nerve Degeneration , Nerve Regeneration , Neural Stem Cells/transplantation , Neurodegenerative Diseases/surgery , Stem Cell Transplantation , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Alzheimer Disease/surgery , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Amyotrophic Lateral Sclerosis/surgery , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Humans , Neural Stem Cells/immunology , Neural Stem Cells/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neuroimmunomodulation , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Parkinson Disease/surgery , Phenotype , Recovery of Function , Stem Cell Transplantation/adverse effects
6.
Front Immunol ; 12: 697203, 2021.
Article in English | MEDLINE | ID: mdl-34248998

ABSTRACT

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.


Subject(s)
Neural Stem Cells/immunology , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Transgenes , Animals , Astrocytes/immunology , Brain/cytology , Brain/immunology , Brain/surgery , Cell Proliferation/genetics , Cells, Cultured , DNA, Bacterial/genetics , Genes, Bacterial , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Kidney/cytology , Kidney/immunology , Kidney/surgery , Magnetite Nanoparticles , Magnetosomes/genetics , Magnetospirillum/genetics , Microglia/immunology , Neural Stem Cells/cytology , Rats , Recombinant Proteins/genetics , Spleen/cytology , Spleen/immunology , Spleen/surgery , Stem Cell Transplantation/adverse effects , Transplantation, Heterologous
8.
PLoS Negl Trop Dis ; 15(3): e0009183, 2021 03.
Article in English | MEDLINE | ID: mdl-33657175

ABSTRACT

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.


Subject(s)
Immunity, Innate , Neural Stem Cells/virology , Zika Virus Infection/immunology , Zika Virus/physiology , Brain/immunology , Brain/virology , Cell Differentiation , Cell Proliferation , Humans , Neural Stem Cells/immunology , Neurogenesis/immunology , Virus Replication , Zika Virus Infection/virology
9.
Int J Mol Sci ; 22(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572157

ABSTRACT

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.


Subject(s)
Cerebral Cortex/immunology , Inflammation Mediators/metabolism , Microglia/immunology , Platelet-Rich Plasma/immunology , Adult , Age Factors , Aged , Aged, 80 and over , Aging/immunology , Animals , Apoptosis/immunology , Cell Line, Tumor , Cell Proliferation , Cerebral Cortex/cytology , Chemokine CCL11/metabolism , Female , Humans , Male , Mice , Microglia/cytology , Neural Stem Cells/immunology , Neurogenesis/immunology , Neurons/immunology , Platelet-Rich Plasma/cytology , Platelet-Rich Plasma/metabolism , Primary Cell Culture , Rats , Synapses/immunology , Young Adult
10.
Front Immunol ; 12: 784098, 2021.
Article in English | MEDLINE | ID: mdl-34975872

ABSTRACT

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.


Subject(s)
Ischemic Stroke/immunology , Neovascularization, Physiologic/immunology , Neuroinflammatory Diseases/immunology , Neuronal Plasticity/immunology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Ischemic Stroke/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Microglia/immunology , Microglia/metabolism , Neural Stem Cells/immunology , Neural Stem Cells/metabolism , Neuroinflammatory Diseases/pathology , Recovery of Function/immunology
11.
Viruses ; 12(8)2020 08 12.
Article in English | MEDLINE | ID: mdl-32806715

ABSTRACT

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.


Subject(s)
Flavivirus/pathogenicity , Immunity, Innate , Neural Stem Cells/virology , Virus Replication , West Nile virus/pathogenicity , Apoptosis , Cell Survival , Cells, Cultured , Flavivirus/physiology , Humans , Induced Pluripotent Stem Cells , Kinetics , Neural Stem Cells/immunology , Virulence , West Nile virus/physiology
12.
Emerg Microbes Infect ; 9(1): 1580-1589, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32576094

ABSTRACT

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.


Subject(s)
Alphavirus Infections/genetics , Alphavirus/immunology , Neural Stem Cells/virology , RNA, Small Interfering/metabolism , RNA, Viral/immunology , Alphavirus/genetics , Alphavirus Infections/immunology , Alphavirus Infections/virology , Animals , Cell Differentiation , Cell Line , Central Nervous System/immunology , Central Nervous System/virology , Chlorocebus aethiops , HEK293 Cells , Humans , Mice , Muscle, Skeletal/immunology , Muscle, Skeletal/virology , Neural Stem Cells/immunology , RNA, Viral/antagonists & inhibitors , Sindbis Virus/genetics , Sindbis Virus/immunology , Vero Cells , Virus Replication , Zika Virus/genetics , Zika Virus/immunology
13.
J Neuroinflammation ; 17(1): 27, 2020 Jan 18.
Article in English | MEDLINE | ID: mdl-31954398

ABSTRACT

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.


Subject(s)
Hippocampus/cytology , Hippocampus/immunology , Microglia/immunology , Microglia/metabolism , Signal Transduction/immunology , Aminopyridines/pharmacology , Animals , Ethanol/toxicity , Hippocampus/metabolism , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Microglia/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/immunology , Neural Stem Cells/metabolism , Organ Culture Techniques , Pyrroles/pharmacology , Rats , Signal Transduction/drug effects , Toll-Like Receptors/agonists
14.
Int Immunopharmacol ; 81: 106038, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31757678

ABSTRACT

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.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Brain/immunology , Glioblastoma/therapy , Hepatitis B Antibodies/immunology , Hepatitis B Vaccines/administration & dosage , Animals , Astrocytes/immunology , Brain/cytology , Brain/pathology , Carrier State/epidemiology , Carrier State/immunology , Cell Differentiation/immunology , Child , Disease Models, Animal , Glioblastoma/epidemiology , Glioblastoma/immunology , Glioblastoma/pathology , Hepatitis B/epidemiology , Hepatitis B/immunology , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Humans , Immunogenicity, Vaccine , Myelin Sheath/immunology , Neural Stem Cells/immunology , Oligodendroglia/cytology , Oligodendroglia/immunology , Prevalence
15.
Stem Cell Res Ther ; 10(1): 387, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31843025

ABSTRACT

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.


Subject(s)
Interferon Type I/immunology , Neural Stem Cells/immunology , Neural Stem Cells/virology , Zika Virus Infection/immunology , Cell Line , Cells, Cultured , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Encephalitis Viruses, Japanese/immunology , Encephalitis, Japanese/genetics , Encephalitis, Japanese/immunology , Humans , Immunity, Innate , Interferon Type I/biosynthesis , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/immunology , Interferons/genetics , Interferons/immunology , Neural Stem Cells/pathology , Receptors, Immunologic , Transcription, Genetic , Transfection , Zika Virus/immunology , Zika Virus Infection/genetics , Zika Virus Infection/pathology , Interferon Lambda
16.
J Vis Exp ; (151)2019 09 19.
Article in English | MEDLINE | ID: mdl-31609309

ABSTRACT

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.


Subject(s)
5-Methylcytosine/analogs & derivatives , Brain Chemistry , Brain/immunology , Neural Stem Cells/chemistry , Neural Stem Cells/immunology , 5-Methylcytosine/analysis , 5-Methylcytosine/immunology , Age Factors , Animals , Cell Line , Cells, Cultured , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Immunoblotting/methods , Male , Mice , Mice, Inbred C57BL
17.
Brain ; 142(11): 3456-3472, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31529023

ABSTRACT

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.


Subject(s)
Immune Tolerance , Microglia/immunology , Microglia/transplantation , Myelin Sheath , Neural Stem Cells/immunology , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Adoptive Transfer , Allografts , Animals , Cytokines/biosynthesis , Graft Rejection , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , T-Lymphocytes/immunology , Transplantation, Homologous
18.
Arch Toxicol ; 93(9): 2661-2671, 2019 09.
Article in English | MEDLINE | ID: mdl-31332466

ABSTRACT

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.


Subject(s)
Benzhydryl Compounds/toxicity , Environmental Pollutants/toxicity , Neural Stem Cells/drug effects , Neurotoxicity Syndromes/enzymology , Phenols/toxicity , beta-N-Acetylhexosaminidases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzhydryl Compounds/chemistry , Calcium/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/chemistry , Humans , Molecular Docking Simulation , Neural Stem Cells/enzymology , Neural Stem Cells/immunology , Neuronal Outgrowth/drug effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , PC12 Cells , Phenols/chemistry , Rats , Reactive Oxygen Species/metabolism
19.
Immunol Lett ; 212: 6-13, 2019 08.
Article in English | MEDLINE | ID: mdl-31154052

ABSTRACT

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.


Subject(s)
Cell Transdifferentiation/immunology , Colony-Stimulating Factors/pharmacology , Encephalomyelitis, Autoimmune, Experimental/therapy , Mesenchymal Stem Cell Transplantation/methods , Multiple Sclerosis/therapy , Animals , Cell Culture Techniques , Cell Transdifferentiation/drug effects , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Mice , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Neural Stem Cells/immunology , Spheroids, Cellular , Treatment Outcome
20.
J Neuroimmunol ; 332: 73-77, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30959341

ABSTRACT

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.


Subject(s)
Neural Stem Cells/virology , Zika Virus Infection/immunology , Zika Virus/pathogenicity , 2',5'-Oligoadenylate Synthetase/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Female , Hippocampus/cytology , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , Male , Microglia/immunology , Myxovirus Resistance Proteins/biosynthesis , Neural Stem Cells/immunology , Neural Stem Cells/metabolism , Rats , Viral Tropism
SELECTION OF CITATIONS
SEARCH DETAIL
...