Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Neuroscience ; 463: 272-287, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33811940

ABSTRACT

Opioid use by women during pregnancy has risen dramatically since 2004, accompanied by a striking increase in the prevalence of neonatal opioid withdrawal syndrome (NOWS) and other long-term neurological deficits. However, the mechanisms underlying the impact of prenatal opioid exposure on fetal neurodevelopment are largely unknown. To translate from the clinical presentation, we developed a novel mouse model to study the neurodevelopmental consequences of maternal opioid use and management. Female mice were treated with oxycodone (OXY) before mating to mimic opioid use disorder (OUD) in humans. Following pregnancy confirmation, dams were switched to buprenorphine (BUP) via oral administration, simulating medication management of OUD (MOUD) in pregnant women. Here, we document critical changes in fetal brain development including reduced cortical thickness, altered corticogenesis, and ventriculomegaly in embryos from dams that were treated with opioids before and throughout pregnancy. Maternal care giving behavior was slightly altered without affecting gross growth of offspring. However, adolescent offspring exposed to maternal opioid use during pregnancy exhibited hyperactivity in late adolescence. Remarkably, we also show increased generation of dopaminergic neurons within the ventral tegmental area (VTA) of mice exposed to prenatal opioids. These data provide critical evidence of teratogenic effects of opioid use during pregnancy and suggest a causal relationship between maternal opioid use and neurodevelopmental/behavioral anomalies in adolescence.


Subject(s)
Buprenorphine , Neonatal Abstinence Syndrome , Opioid-Related Disorders , Prenatal Exposure Delayed Effects , Adolescent , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/toxicity , Animals , Buprenorphine/therapeutic use , Female , Humans , Infant, Newborn , Mice , Neonatal Abstinence Syndrome/drug therapy , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy
3.
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
4.
PLoS Negl Trop Dis ; 14(7): e0008413, 2020 07.
Article in English | MEDLINE | ID: mdl-32628667

ABSTRACT

Global Zika virus (ZIKV) outbreaks and their link to microcephaly have raised major public health concerns. However, the mechanism of maternal-fetal transmission remains largely unknown. In this study, we determined the role of yolk sac (YS) microglial progenitors in a mouse model of ZIKV vertical transmission. We found that embryonic (E) days 6.5-E8.5 were a critical window for ZIKV infection that resulted in fetal demise and microcephaly, and YS microglial progenitors were susceptible to ZIKV infection. Ablation of YS microglial progenitors significantly reduced the viral load in both the YS and the embryonic brain. Taken together, these results support the hypothesis that YS microglial progenitors serve as "Trojan horses," contributing to ZIKV fetal brain dissemination and congenital brain defects.


Subject(s)
Fetus/pathology , Microcephaly/pathology , Microglia/virology , Pregnancy Complications, Infectious/pathology , Zika Virus Infection/pathology , Zika Virus/isolation & purification , Animals , Brain/virology , Disease Models, Animal , Female , Fetus/virology , Humans , Infectious Disease Transmission, Vertical , Male , Mice , Mice, Inbred C57BL , Microcephaly/embryology , Microcephaly/virology , Microglia/metabolism , Pregnancy , Pregnancy Complications, Infectious/virology , Viral Load , Zika Virus/physiology , Zika Virus Infection/transmission , Zika Virus Infection/virology
5.
Brain Res ; 1723: 146425, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31473223

ABSTRACT

Cocaine and ethanol are two commonly co-abused substances; however, the neuropathology following chronic dual consumption is poorly understood. Neural stem cells (NSCs) are a subpopulation of cells within the adult brain that are integral to brain maintenance and repair making them an appealing target to reverse neurodegeneration associated with abused substances. Yet, knowledge about NSC response to chronic poly-drug administration of ethanol and cocaine is minimal. Here, we developed a novel chronic poly-drug administration paradigm of ethanol and cocaine using a transgenic mouse model to trace endogenous NSC survival and differentiation in three brain regions from both male and female mice. We report significant and distinct patterns of NSC survival and differentiation among brain regions, as well as between sexes. Additionally, poly-drug administration had synergistic effects on NSC survival. Altered cognitive and hedonic behaviors were also observed, however the extent of these behavioral changes was not proportional to the NSC changes. With this mouse model we can effectively examine cognitive and behavioral changes and correlate them with pathological changes in the brain in response to chronic poly-drug administration, which is of great value in understanding the progression of neurodegeneration in polysubstance use disorders and evaluation potential therapeutics on neuroregeneration.


Subject(s)
Cocaine/adverse effects , Ethanol/adverse effects , Neural Stem Cells/drug effects , Adult Stem Cells/drug effects , Age Factors , Animals , Brain/pathology , Cell Differentiation/drug effects , Cocaine/metabolism , Cocaine/pharmacology , Disease Models, Animal , Ethanol/metabolism , Ethanol/pharmacology , Female , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Regeneration/drug effects , Neurogenesis/physiology , Sex Factors
6.
Neurochem Int ; 120: 21-32, 2018 11.
Article in English | MEDLINE | ID: mdl-30041015

ABSTRACT

Human neural stem cells (hNSCs) can differentiate into an oligodendrocyte lineage to facilitate remyelination in patients. Molecular mechanisms underlying oligodendrocyte fate specification remains unknown, hindering the development of efficient methods to generate oligodendrocytes from hNSCs. We have found that Neurobasal-A medium (NB) is capable of inducing hNSCs to oligodendrocyte progenitor cells (OPCs). We identified several signaling molecules are altered after cultivation in NB medium, including Akt, ERK1/2 and c-Src. While sustained activation of Akt and ERK1/2 during both NB induction and subsequent differentiation was required for OPC differentiation, c-Src phosphorylation was increased temporally during the period of NB induction. Both pharmacological inhibition and RNA interference confirmed that a transient elevation of phospho-c-Src is critical for OPC induction. Furthermore, inactivation of c-Src inhibited phosphorylation of Akt and ERK1/2. In summary, we identified a novel and critical role of c-Src in guiding hNSC differentiation to an oligodendrocyte lineage.


Subject(s)
Cell Differentiation/physiology , Neural Stem Cells/cytology , Oligodendroglia/cytology , src-Family Kinases/metabolism , CSK Tyrosine-Protein Kinase , Cell Lineage/physiology , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Myelin Sheath/metabolism , Neurogenesis/physiology
7.
Stem Cell Reports ; 9(6): 1916-1930, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29129682

ABSTRACT

Chronic alcohol abuse results in alcohol-related neurodegeneration, and critical gaps in our knowledge hinder therapeutic development. Neural stem cells (NSCs) are a subpopulation of cells within the adult brain that contribute to brain maintenance and recovery. While it is known that alcohol alters NSCs, little is known about how NSC response to alcohol is related to sex, brain region, and stage of differentiation. Understanding these relationships will aid in therapeutic development. Here, we used an inducible transgenic mouse model to track the stages of differentiation of adult endogenous NSCs and observed distinct NSC behaviors in three brain regions (subventricular zone, subgranular zone, and tanycyte layer) after long-term alcohol consumption. Particularly, chronic alcohol consumption profoundly affected the survival of NSCs in the subventricular zone and altered NSC differentiation in all three regions. Significant differences between male and female mice were further discovered.


Subject(s)
Alcohol Drinking/physiopathology , Lateral Ventricles/physiopathology , Nerve Degeneration/physiopathology , Neural Stem Cells/drug effects , Adult Stem Cells/drug effects , Alcohol Drinking/genetics , Alcohols/toxicity , Animals , Brain Mapping , Cell Differentiation/drug effects , Disease Models, Animal , Female , Lateral Ventricles/drug effects , Male , Mice , Mice, Transgenic , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Neural Stem Cells/pathology , Neurons/drug effects , Neurons/pathology
8.
Stem Cell Reports ; 8(3): 715-727, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28216147

ABSTRACT

Zika virus (ZIKV) infection causes microcephaly in a subset of infants born to infected pregnant mothers. It is unknown whether human individual differences contribute to differential susceptibility of ZIKV-related neuropathology. Here, we use an Asian-lineage ZIKV strain, isolated from the 2015 Mexican outbreak (Mex1-7), to infect primary human neural stem cells (hNSCs) originally derived from three individual fetal brains. All three strains of hNSCs exhibited similar rates of Mex1-7 infection and reduced proliferation. However, Mex1-7 decreased neuronal differentiation in only two of the three stem cell strains. Correspondingly, ZIKA-mediated transcriptome alterations were similar in these two strains but significantly different from that of the third strain with no ZIKV-induced neuronal reduction. This study thus confirms that an Asian-lineage ZIKV strain infects primary hNSCs and demonstrates a cell-strain-dependent response of hNSCs to ZIKV infection.


Subject(s)
Brain/metabolism , Brain/virology , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Astrocytes , Brain/immunology , Cell Differentiation , Cell Proliferation , Cell Survival , Chlorocebus aethiops , Cluster Analysis , Fetus , Gene Expression , Gene Expression Profiling , Humans , Immunity, Innate , Male , Mice , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons , Transcriptome , Vero Cells , Zika Virus/classification , Zika Virus Infection/genetics , Zika Virus Infection/immunology , Zika Virus Infection/metabolism
9.
Cell Transplant ; 25(10): 1863-1877, 2016 10.
Article in English | MEDLINE | ID: mdl-26980267

ABSTRACT

Neural stem cells (NSCs) promote recovery from brain trauma, but neuronal replacement is unlikely the sole underlying mechanism. We hypothesize that grafted NSCs enhance neural repair at least partially through modulating the host immune response after traumatic brain injury (TBI). C57BL/6 mice were intracerebrally injected with primed human NSCs (hNSCs) or vehicle 24 h after a severe controlled cortical impact injury. Six days after transplantation, brain tissues were collected for Western blot and immunohistochemical analyses. Observations included indicators of microglia/macrophage activation, M1 and M2 phenotypes, axonal injury detected by amyloid precursor protein (APP), lesion size, and the fate of grafted hNSCs. Animals receiving hNSC transplantation did not show significant decreases of brain lesion volumes compared to transplantation procedures with vehicle alone, but did show significantly reduced injury-dependent accumulation of APP. Furthermore, intracerebral transplantation of hNSCs reduced microglial activation as shown by a diminished intensity of Iba1 immunostaining and a transition of microglia/macrophages toward the M2 anti-inflammatory phenotype. The latter was represented by an increase in the brain M2/M1 ratio and increases of M2 microglial proteins. These phenotypic switches were accompanied by the increased expression of anti-inflammatory interleukin-4 receptor α and decreased proinflammatory interferon-γ receptor ß. Finally, grafted hNSCs mainly differentiated into neurons and were phagocytized by either M1 or M2 microglia/macrophages. Thus, intracerebral transplantation of primed hNSCs efficiently leads host microglia/macrophages toward an anti-inflammatory phenotype that presumably contributes to stem cell-mediated neuroprotective effects after severe TBI in mice.


Subject(s)
Brain Injuries, Traumatic/therapy , Macrophages/metabolism , Microglia/metabolism , Neural Stem Cells/transplantation , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , B7-2 Antigen/metabolism , Brain/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cell Differentiation , Cells, Cultured , Humans , Interleukin-4 Receptor alpha Subunit/genetics , Interleukin-4 Receptor alpha Subunit/metabolism , Lectins, C-Type/metabolism , Macrophages/cytology , Macrophages/immunology , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/immunology , Neural Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , Phagocytosis , Phenotype , Receptors, Cell Surface/metabolism , Receptors, IgG/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism
10.
J Neurorestoratology ; 1: 1-12, 2013.
Article in English | MEDLINE | ID: mdl-35663360

ABSTRACT

The efficacy of stem cell-based therapy for neurological diseases depends highly on cell survival post-transplantation. One of the key factors affecting cell survival is the grafting procedure. The current study aims to determine whether needle insertion into intact rat spinal cords creates a hypoxic environment that is prone to lipid peroxidation damage upon reperfusion, and whether an antioxidant protects human neural stem cells (hNSCs) both in vitro and post-transplantation into rat spinal cords. We show here that a single needle injection creates a hypoxic environment within the rat spinal cord that peaks at approximately 12 hours before reperfusion occurs. Lipid peroxidation damage at the transplantation site is evident by 48 hours post-needle insertion. In an in vitro model, hypoxia-reperfusion results in apoptotic death of hNSCs. Pretreatment with the antioxidant, α-lipoic acid, protects hNSCs against hypoxia-reperfusion injury and oxidative stress-mediated cell death. Increasing glutathione, but not Akt signaling, contributes to the protective effect of lipoic acid. Pretreating hNSCs with lipoic acid also increases the cell survival rate 1 month post-transplantation. Further investigation is warranted to develop improved techniques to maximize the survival of transplanted stem cells.

11.
Am J Stem Cells ; 1(1): 2-21, 2012.
Article in English | MEDLINE | ID: mdl-23671793

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease. Human neural stem cells (hNSCs) may have the potential to replace lost motor neurons. The therapeutic efficacy of stem cell therapy depends greatly on the survival of grafted stem cell-derived motor neurons in the microenvironment of the spinal cord in ALS. After transplantation of hNSCs into the spinal cords of transgenic ALS rats, morphological analysis reveals that grafted hNSCs differentiate into motor neurons. However, hNSCs degenerate and show signs of nitroxidative damage at the disease end-stage. Using an in vitro coculture system, we systematically assess interactions between microglia and astroglia derived from both nontransgenic rats and transgenic rats expressing human mutant SOD1(G93A) before and after symptomatic disease onset, and determine the effects of such microglia-astroglia interactions on the survival of hNSC-derived motor neurons. We found that ALS microglia, specifically isolated after symptomatic disease onset, are directly toxic to hNSC-derived motor neurons. Furthermore, nontransgenic astrocytes not only lose their protective role in hNSC-derived motor neuron survival in vitro, but also exhibit toxic features when cocultured with mutant SOD1(G93A) microglia. Using inhibitors of inducible nitric oxide synthase and NADPH oxidase, we show that microglia-generated nitric oxide and superoxide partially contribute to motor neuron loss and astrocyte dysfunction in this coculture paradigm. In summary, reactive oxygen/nitrogen species released from overactivated microglia in ALS directly eliminate human neural stem cell-derived motor neurons and reduce the neuroprotective capacities of astrocytes.

12.
J Neurotrauma ; 29(2): 295-312, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22077363

ABSTRACT

Transplantation of neural stem cells (NSCs) improves functional outcomes following traumatic brain injury (TBI). Previously we demonstrated that human NSCs (hNSCs) via releasing glial cell line-derived neurotrophic factor (GDNF), preserved cognitive function in rats following parasagittal fluid percussion. However, the underlying mechanisms remain elusive. In this study, we report that NSC grafts significantly reduce TBI-induced axonal injury in the fimbria and other brain regions by blocking abnormal accumulation of amyloid precursor protein (APP). A preliminary mass spectrometry proteomics study revealed the opposite effects of TBI and NSCs on many of the cytoskeletal proteins in the CA3 region of the hippocampus, including α-smooth muscle actin (α-SMA), the main stress fiber component. Further, Western blot and immunostaining studies confirmed that TBI significantly increased the expression of α-SMA in hippocampal neurons, whereas NSC grafts counteracted the effect of TBI. In an in vitro model, rapid stretch injury significantly shortened lengths of axons and dendrites, increased the expression of both APP and α-SMA, and induced actin aggregation, effects offset by GDNF treatment. These GDNF protective effects were reversed by a GDNF-neutralizing antibody or a specific calcineurin inhibitor, and were mimicked by a specific Rho inhibitor. In summary, we demonstrate for the first time that hNSC grafts and treatment with GDNF acutely reduce traumatic axonal injury and promote neurite outgrowth. Possible mechanisms underlying GDNF-mediated neurite protection include balancing the activity of calcineurin, whereas GDNF-induced neurite outgrowth may result from the reduction of the abnormal α-SMA expression and actin aggregation via blocking Rho signals. Our study also suggests the necessity of further exploring the roles of α-SMA in the central nervous system (CNS), which may lead to a new avenue to facilitate recovery after TBI and other injuries.


Subject(s)
Diffuse Axonal Injury/pathology , Diffuse Axonal Injury/physiopathology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Recovery of Function/physiology , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Line , Cells, Cultured , Diffuse Axonal Injury/metabolism , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Humans , Male , Neural Stem Cells/cytology , Rats , Rats, Sprague-Dawley
13.
PLoS One ; 6(8): e23414, 2011.
Article in English | MEDLINE | ID: mdl-21887250

ABSTRACT

Fibroblast growth factor (FGF) and epidermal growth factor (EGF) are critical for the development of the nervous system. We previously discovered that FGF2 and EGF had opposite effects on motor neuron differentiation from human fetal neural stem cells (hNSCs), but the underlying mechanisms remain unclear. Here, we show that FGF2 and EGF differentially affect the temporal patterns of Akt and glycogen synthase kinase 3 beta (GSK3ß) activation. High levels of phosphatidylinositol 3-kinase (PI3K)/Akt activation accompanied with GSK3ß inactivation result in reduction of the motor neuron transcription factor HB9. Inhibition of PI3K/Akt by chemical inhibitors or RNA interference or overexpression of a constitutively active form of GSK3ß enhances HB9 expression. Consequently, PI3K inhibition increases hNSCs differentiation into HB9(+)/microtubule-associated protein 2 (MAP2)(+) motor neurons in vitro. More importantly, blocking PI3K not only enhances motor neuron differentiation from hNSCs grafted into the ventral horn of adult rat spinal cords, but also permits ectopic generation of motor neurons in the dorsal horn by overriding environmental influences. Our data suggest that FGF2 and EGF affect the motor neuron fate decision in hNSCs differently through a fine tuning of the PI3K/AKT/GSK3ß pathway, and that manipulation of this pathway can enhance motor neuron generation.


Subject(s)
Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Glycogen Synthase Kinase 3/metabolism , Motor Neurons/cytology , Neural Stem Cells/cytology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3 beta , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Motor Neurons/drug effects , Motor Neurons/enzymology , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neural Stem Cells/transplantation , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Kinase C/metabolism , RNA Interference/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...