Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Drug Des Devel Ther ; 18: 2715-2727, 2024.
Article in English | MEDLINE | ID: mdl-38974122

ABSTRACT

Hepatic fibrosis (HF) is a pathological process of structural and functional impairment of the liver and is a key component in the progression of chronic liver disease. There are no specific anti-hepatic fibrosis (anti-HF) drugs, and HF can only be improved or prevented by alleviating the cause. Autophagy of hepatic stellate cells (HSCs) is closely related to the development of HF. In recent years, traditional Chinese medicine (TCM) has achieved good therapeutic effects in the prevention and treatment of HF. Several active ingredients from TCM (AITCM) can regulate autophagy in HSCs to exert anti-HF effects through different pathways, but relevant reviews are lacking. This paper reviewed the research progress of AITCM regulating HSCs autophagy against HF, and also discussed the relationship between HSCs autophagy and HF, pointing out the problems and limitations of the current study, in order to provide references for the development of anti-HF drugs targeting HSCs autophagy in TCM. By reviewing the literature in PubMed, Web of Science, Embase, CNKI and other databases, we found that the relationship between autophagy of HSCs and HF is currently controversial. HSCs autophagy may promote HF by consuming lipid droplets (LDs) to provide energy for their activation. However, in contrast, inducing autophagy in HSCs can exert the anti-HF effect by stimulating their apoptosis or senescence, reducing type I collagen accumulation, inhibiting the extracellular vesicles release, degrading pro-fibrotic factors and other mechanisms. Some AITCM inhibit HSCs autophagy to resist HF, with the most promising direction being to target LDs. While, others induce HSCs autophagy to resist HF, with the most promising direction being to target HSCs apoptosis. Future research needs to focus on cell targeting research, autophagy targeting research and in vivo verification research, and to explore the reasons for the contradictory effects of HSCs autophagy on HF.


Subject(s)
Autophagy , Drugs, Chinese Herbal , Hepatic Stellate Cells , Liver Cirrhosis , Medicine, Chinese Traditional , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Autophagy/drug effects , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Animals
2.
Ann Transl Med ; 11(2): 74, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36819507

ABSTRACT

Background: This study sought to explore the mechanism underlying the therapeutic effects of electroacupuncture (EA) on spatial memory deficits caused by surgery. Methods: Hepatic apex resection was performed under propofol-based total intravenous anesthesia. Male Sprague-Dawley rats were subjected to EA treatment or EA + mitochondrial division inhibitor-1 (mdivi-1) treatment once a day for three consecutive days after surgery. The Morris water maze test was used to evaluate the spatial memory of the rats after surgery. Tissue from the hippocampus of each rat was frozen and used for transcriptomic and proteomic analyses to identify potential targets for EA treatment. Western blotting was used to confirm the protein expression levels. The levels of reactive oxygen species (ROS) and adenosine triphosphate (ATP) were detected using commercial kits. The rat mitochondria were then isolated, and the activity of mitochondrial complex V was assessed. Results: EA attenuated surgery-induced spatial memory deficits on postoperative day 3, while these effects were reversed by treatment with the mdivi-1 (P<0.05). Ribonucleic acid (RNA)-sequencing revealed that EA upregulated multiple metabolic pathways and the phosphatidylinositol 3­kinas/protein kinase B signaling pathway. The proteomic and western blotting results suggested that the EA treatment substantially downregulated coiled-coil-helix-coiled-coil-helix domain containing 3 (ChChd3) expression in the hippocampus. The EA treatment significantly increased the autophagy-related protein levels, including phosphatase and tensin homolog-induced kinase 1, Parkin, MAP1LC3 (LC3), and Beclin1, and inhibited the production of ROS and inflammatory cytokine interleukin-1ß in the hippocampus (P<0.05). Conclusions: These results suggest that EA ameliorates postoperative spatial memory deficits and protects hippocampus from oxidative stress and inflammation through enhanced autophagy in an animal model of perioperative neurocognitive disorders (PNDs).

3.
Sleep Biol Rhythms ; 21(3): 347-357, 2023 Jul.
Article in English | MEDLINE | ID: mdl-38476312

ABSTRACT

The study was attempted to investigate the effect on and mechanisms of action of dexmedetomidine with regard to learning and memory impairment in rats with chronic rapid eye movement (REM) sleep deprivation. A total of 50 male Sprague Dawley rats were randomly divided into five groups. Modified multiple platform method was conducted to cause the sleep deprivation of rats. Dexmedetomidine and midazolam were administered by intraperitoneal injection. Learning and memory ability was assessed through Morris water maze. Morphological changes of rat hippocampal neurons and synaptic were detected by transmission electron microscope and Golgi staining. The gene expression in hippocampus of each group was detected by RNA-seq and verified by RT-PCR and western blot. REM Sleep-deprived rats exhibited spatial learning and memory deficits. Furthermore, there was decreased density of synaptic spinous in the hippocampal CA1 region of the sleep deprivation group compared with the control. Additionally, transmission electron microscopy showed that the synaptic gaps of hippocampal neurons in REM sleep deprivation group were loose and fuzzy. Interestingly, dexmedetomidine treatment normalized these events to control levels following REM sleep deprivation. Molecular biological methods showed that Alox15 expression increased significantly after REM sleep deprivation as compared to control, while dexmedetomidine administration reversed the expression of Alox15. Dexmedetomidine alleviated the spatial learning and memory dysfunction induced with chronic REM sleep deprivation in rats. This protective effect may be related to the down-regulation of Alox15 expression and thereby the enhancement of synaptic structural plasticity in the hippocampal CA1 area of rats. Supplementary Information: The online version contains supplementary material available at 10.1007/s41105-023-00450-8.

4.
Pain Physician ; 25(4): E629-E640, 2022 07.
Article in English | MEDLINE | ID: mdl-35793187

ABSTRACT

BACKGROUND: Thalamic pain (TP), also known as central post-stroke pain, is a chronic neuropathic pain syndrome that follows a stroke and is a severe pain that is usually intractable. No universally applicable and effective therapies have been proposed. Emerging studies have reported that electroacupuncture (EA) can potentially be used as an effective therapy for the treatment of neuropathic pain. However, whether EA influences TP and if so, by what potential mechanism, remains poorly understood. OBJECTIVE: The aim of this study was to detect the efficacy of EA and explore possible mechanisms for treating TP. STUDY DESIGN: Controlled animal study. SETTING: The laboratory at the Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine. METHODS: Male Sprague Dawley rats were randomly divided into 3 groups (n = 15 / group): sham-operated (SH) group, thalamic pain model (TP) group, EA treatment (EA) group. After the TP rat model was successfully established, EA was used for intervention. During the experiment, the mechanical pain thresholds of rats were detected among the groups. The right thalamus of the rats was extracted on postoperative day 28 for RNA-sequencing (RNA-Seq) analysis to find the changes in gene expression in different groups of rats. The key genes were screened using reverse transcription-polymerase chain reaction (RT-PCR) detection and subsequently identified with western blotting and immunofluorescence. RESULTS: The mechanical withdrawal threshold (MWT) value of the right facial skin in the TP group and the EA group decreased significantly on the 3rd day after surgery, compared to the SH group (P < 0.01). From 7 to 28 days, the MWT value increased continually in the EA group; however, there was no significant change in the TP group. The results of RNA-seq showed that compared to the TP group, 377 genes changed in the EA group. Moreover, ADCY1 expression increased significantly in the TP group as compared to the SH group, while EA treatment reversed the expression of ADCY1. LIMITATIONS: In addition to ADCY1, the mechanism(s) of other signaling pathways in TP need to be explored in future research. CONCLUSIONS: EA treatment may promote the recovery of TP model rat by regulating ADCY1 expression.


Subject(s)
Electroacupuncture , Neuralgia , Animals , Electroacupuncture/methods , Humans , Male , Neuralgia/metabolism , Pain Threshold , Rats , Rats, Sprague-Dawley , Up-Regulation
5.
Neuroreport ; 25(10): 782-7, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-24918459

ABSTRACT

Nestin knockout leads to embryonic lethality and self-renewal deficiency in neural stem cells (NSCs). However, how nestin maintains self-renewal remains uncertain. Here, we used the dosage effect of nestin in heterozygous mice (Nes+/-) to study self-renewal of NSCs. With existing extracellular signaling in vivo or in vitro, nestin levels do not affect proliferation ability or apoptosis when compared between Nes+/- and Nes+/+ NSCs. However, self-renewal ability of Nes+/- NSCs is impaired when plated at a low cell density and completely lost at a clonal density. This deficiency in self-renewal at a clonal density is rescued using a medium conditioned by Nes+/+ NSCs. In addition, the Akt signaling pathway is altered at low density and reversed by conditioned medium. Our data show that secreted factors contribute toward maintaining self-renewal of NSCs by nestin, potentially through Akt signaling.


Subject(s)
Cell Proliferation/genetics , Nestin/genetics , Neural Stem Cells/physiology , Animals , Apoptosis , Cell Count , Cells, Cultured , Extracellular Space/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
6.
PLoS One ; 8(5): e63876, 2013.
Article in English | MEDLINE | ID: mdl-23667683

ABSTRACT

We have recently demonstrated that adeno-associated virus serotype 9 (AAV9)-mediated human erythropoietin (hEPO) gene delivery into the brain protects dopaminergic (DA) neurons in the substantia nigra in a rat model of Parkinson's disease. In the present study, we examined whether pre-exposure to AAV9-hEPO vectors with an intramuscular or intrastriatal injection would reduce AAV9-mediated hEPO transduction in rat brain. We first characterized transgene expression and immune responses against AAV9-hEPO vectors in rat striatum at 4 days, 3 weeks and 6 months, and with doses ranging from 10(11) to 10(13) viral genomes. To sensitize immune system, rats received an injection of AAV9-hEPO into either the muscle or the left striatum, and then sequentially an injection of AAV9-hEPO into the right striatum 3 weeks later. We observed that transgene expression exhibited in a time course and dose dependent manner, and inflammatory and immune responses displayed in a time course manner. Intramuscular, but not intrastriatal injections of AAV9-hEPO resulted in reduced levels of hEPO transduction and increased levels of the major histocompatibility complex (MHC) class I and class II antigen expression in the striatum following AAV9-hEPO re-administration. There were infiltration of the cluster of differentiation 4 (CD4)-and CD8-lymphacytes, and accumulation of activated microglial cells and astrocytes in the virally injected striatum. In addition, the sera from the rats with intramuscular injections of AAV9-hEPO contained greater levels of antibodies against both AAV9 capsid protein and hEPO protein than the other treatment groups. hEPO gene expression was negatively correlated with the levels of circulating antibodies against AAV9 capsid protein. Intramuscular and intrastriatal re-administration of AAV9-hEPO led to increased numbers of red blood cells in peripheral blood. Our results suggest that pre-immunization with an intramuscular injection can lead to the reduction of transgene expression in the striatal re-administration.


Subject(s)
Corpus Striatum/metabolism , Erythropoietin/metabolism , Genetic Vectors/immunology , Immunization/methods , Transduction, Genetic/methods , Animals , Dependovirus/genetics , Erythropoietin/administration & dosage , Erythropoietin/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Injections, Intramuscular , Major Histocompatibility Complex/genetics , Neutralization Tests , Rats , Transgenes/genetics
7.
Glia ; 60(11): 1761-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22855415

ABSTRACT

Increasing evidence indicates the functional expression of ionotropic γ-aminobutyric acid receptor (GABA(A) -R) in astrocytes. However, it remains controversial in regard to the intracellular Cl(-) concentration ([Cl(-) ](i) ) and the functional role of anion-selective GABA(A) -R in astrocytes. In gramicidin perforated-patch recordings from rat hippocampal CA1 astrocytes, GABA and GABA(A) -R-specific agonist THIP depolarized astrocyte membrane potential (V(m) ), and the THIP-induced currents reversed at the voltages between -75.3 and -78.3 mV, corresponding to a [Cl(-) ](i) of 3.1-3.9 mM that favored a passive distribution of Cl(-) anions across astrocyte membrane. Further analysis showed that GABA(A) -R-induced V(m) depolarization was ascribed to HCO(3) (-) efflux, while a passively distributed Cl(-) mediated no net flux or influx of Cl(-) that leads to an unchanged or hyperpolarized V(m) . In addition to a rapidly activated GABA(A) -R current component, GABA and THIP also induced a delayed inward current (DIC) in 63% of astrocytes. The DIC became manifest after agonist withdrawal and enhanced in amplitude with increasing agonist application duration or concentrations. Astrocytic two-pore domain K(+) channels (K2Ps), especially TWIK-1, appeared to underlie the DIC, because (1) acidic intracellular pH, as a result of HCO(3) (-) efflux, inhibited TWIK-1, (2) the DIC remained in the Cs(+) recording solutions that inhibited conventional K(+) channels, and (3) the DIC was completely inhibited by 1 mM quinine but not by blockers for other cation/anion channels. Altogether, HCO(3) (-) efflux through activated GABA(A) -R depolarizes astrocyte V(m) and induces a delayed inhibition of K2Ps K(+) channels via intracellular acidification.


Subject(s)
Astrocytes/physiology , Bicarbonates/metabolism , Hippocampus/physiology , Membrane Potentials/physiology , Potassium Channels/physiology , Receptors, GABA-A/metabolism , Animals , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
8.
Stem Cells ; 28(12): 2162-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20963821

ABSTRACT

The intermediate filament protein, nestin, is a widely employed marker of multipotent neural stem cells (NSCs). Recent in vitro studies have implicated nestin in a number of cellular processes, but there is no data yet on its in vivo function. Here, we report the construction and functional characterization of Nestin knockout mice. We found that these mice show embryonic lethality, with neuroepithelium of the developing neural tube exhibiting significantly fewer NSCs and much higher levels of apoptosis. Consistent with this in vivo observation, NSC cultures derived from knockout embryos show dramatically reduced self-renewal ability that is associated with elevated apoptosis but no overt defects in cell proliferation or differentiation. Unexpectedly, nestin deficiency has no detectable effect on the integrity of the cytoskeleton. Furthermore, the knockout of Vimentin, which abolishes nestin's ability to polymerize into intermediate filaments in NSCs, does not lead to any apoptotic phenotype. These data demonstrate that nestin is important for the proper survival and self-renewal of NSCs, and that this function is surprisingly uncoupled from nestin's structural involvement in the cytoskeleton.


Subject(s)
Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Cell Death , Cell Proliferation , Cell Survival , Cells, Cultured , Cytoskeleton/metabolism , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gene Deletion , Gene Expression Regulation , Gene Targeting , Intermediate Filament Proteins/deficiency , Intermediate Filament Proteins/genetics , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nestin , Neural Tube/metabolism , Neural Tube/pathology , Phenotype
9.
Neuroreport ; 19(4): 419-24, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-18287939

ABSTRACT

Successful isolation and expansion of neural stem/progenitor cells from cynomolgus monkey (cm-NSPCs), may not only help to increase our understanding of NSPCs, but also provide an important translational tool for preclinical trials. Here we initially isolated NSPCs from aborted fetal cynomolgus monkey brain, and expanded them in adherent culture system. Then we demonstrated that cultured cm-NSPCs were almost bipolar cells proliferated in clump-like structure, expressed typical markers for NSPCs, and could differentiate into neurons, astrocytes, and oligodendrocytes. After transduction with lentivirus, 70-80% of cm-NSPCs expressed enhanced green fluorescent protein and the stemness was unaffected. This study provided basis for obtaining large numbers of cm-NSPCs, and efficient transduction of them with exogenous genes, which would facilitate cell-based therapies in nonhuman primate models, and might help to investigate the mechanism of central nervous system development and/or controlling neural regeneration.


Subject(s)
Aborted Fetus/cytology , Brain/cytology , Brain/embryology , Macaca fascicularis/embryology , Stem Cells/cytology , Animals , Biomarkers , Cell Culture Techniques , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Separation , Cells, Cultured , Genetic Therapy/methods , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Lentivirus/genetics , Nerve Regeneration/genetics , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Stem Cell Transplantation/methods , Stem Cells/metabolism , Transfection/methods
10.
Hum Mol Genet ; 17(1): 27-37, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17913699

ABSTRACT

The full potential of embryonic stem (ES) cells to generate precise cell lineages and complex tissues can be best realized when they are differentiated in vivo-i.e. in developing blastocysts. Owing to various practical and ethical constraints, however, it is impossible to introduce ES cells of certain species into blastocysts of the same species. One solution is to introduce ES cells into blastocysts of a different species. However, it is not known whether ES cells can contribute extensively to chimerism when placed into blastocysts of a distantly related species. Here, we address this question using two divergent species, Apodemus sylvaticus and Mus musculus, whose genome sequence differs by approximately 18% from each other. Despite this considerable evolutionary distance, injection of Apodemus ES cells into Mus blastocysts led to viable chimeras bearing extensive Apodemus contributions to all major organs, including the germline, with Apodemus contribution reaching approximately 40% in some tissues. Immunostaining showed that Apodemus ES cells have differentiated into a wide range of cell types in the chimeras. Our results thus provide a proof of principle for the feasibility of differentiating ES cells into a wide range of cell types and perhaps even complex tissues by allowing them to develop in vivo in an evolutionarily divergent host-a strategy that may have important applications in research and therapy. Furthermore, our study demonstrates that mammalian evolution can proceed at two starkly contrasting levels: significant divergence in genome and proteome sequence, yet striking conservation in developmental programs.


Subject(s)
Embryonic Stem Cells/cytology , Transplantation Chimera/genetics , Animals , Animals, Genetically Modified , Base Sequence , Biological Evolution , Blastocyst/cytology , Cell Differentiation , DNA Primers/genetics , Embryonic Development/genetics , Embryonic Stem Cells/transplantation , Female , Germ Cells , Green Fluorescent Proteins/genetics , Male , Mice , Murinae/embryology , Murinae/genetics , Organ Specificity , Phylogeny , Polymerase Chain Reaction , Pregnancy , Recombinant Proteins/genetics , Species Specificity , Teratoma/genetics , Teratoma/pathology , Transplantation Chimera/embryology , Transplantation, Heterologous
11.
Rejuvenation Res ; 10(2): 205-14, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17518693

ABSTRACT

Neurospheres are free-floating heterogeneous spheroid structures maintained in culture and have been used for in vitro expansion of neural stem/progenitor cells. But the growth characteristics and proliferation kinetics of cell population in neurospheres are poorly understood. Using clonogenic culture and immunocytochemistry, we observed the growth dynamics of cell populations in neurospheres and identified key parameters relating to the growth dynamics of cells in neurospheres, including the fraction of dividing cells, cell cycle duration, time delay of first division, and survival rate of cells. Based on these parameters, we established two mathematical models that describe kinetic features of neural stem/progenitor cells under clonogenic conditions. These models provide a powerful tool to explain and predict the experimental outcome and clarify the cell growth characteristics of neural stem/progenitor cells.


Subject(s)
Models, Biological , Neurons/cytology , Spheroids, Cellular , Stem Cells/cytology , Animals , Cell Division , Cell Survival , Clone Cells , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stem Cells/physiology
12.
Mol Cell Biochem ; 304(1-2): 167-79, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17530189

ABSTRACT

Human mesenchymal stem cells (hMSC) are a population of multipotent cells that can differentiate into osteoblasts, chondrocytes, adipocytes, and other cells. The exact mechanism governing the differentiation of hMSC into osteoblasts remains largely unknown. Here, we analyzed protein expression profiles of undifferentiated as well as osteogenic induced hMSC using 2-D gel electrophoresis (2-DE), mass spectrometry (MS), and peptide mass fingerprinting (PMF) to investigate the early gene expression in osteoblast differentiation. We have generated proteome maps of undifferentiated hMSC and osteogenic induced hMSC on day 3 and day 7. 2-DE revealed 102 spots with at least 2.0-fold changes in expression and 52 differently expressed proteins were successfully identified by MALDI-TOF-MS. These proteins were classified into 7 functional categories: metabolism, signal transduction, transcription, calcium-binding protein, protein degradation, protein folding and others. The expression of some identified proteins was confirmed by further RT-PCR analyses. This study clarifies the global proteome during osteoblast differentiation. Our results will play an important role in better elucidating the underlying molecular mechanism in hMSC differentiation into osteoblasts.


Subject(s)
Cell Differentiation , Gene Expression Profiling/methods , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteoblasts/metabolism , Proteomics , Cells, Cultured , Cluster Analysis , Electrophoresis, Gel, Two-Dimensional , Humans , Mesenchymal Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Neuroreport ; 17(10): 981-6, 2006 Jul 17.
Article in English | MEDLINE | ID: mdl-16791088

ABSTRACT

Embryonic stem cells can proliferate indefinitely and are capable of differentiating into derivatives of all three embryonic germ layers in vitro, including the neural lineage. The main objective of this study is to test the effects of neural stem cell conditioned medium on the neural differentiation of mouse embryonic stem cells. When cultured in neural stem cell conditioned medium, mouse embryonic stem cells can form floating cell spheres composed of many nestin-positive cells. After trypsinization and growth on gelatin, these embryonic stem cell-derived neural progenitor cells can be expanded for more than 3 months without loss of neural progenitor characteristics. Both neuronal and glial cells can be readily generated from these cells under differentiation conditions. Thus, neural stem cell conditioned medium is a highly potent reagent for inducing the development of mouse embryonic stem cells into the neural lineage, especially neural progenitor cells.


Subject(s)
Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Neurons/drug effects , Stem Cells/drug effects , Animals , Cell Count/methods , Cell Proliferation/drug effects , Cell Separation/methods , Cells, Cultured , Embryo, Mammalian , Immunohistochemistry/methods , Intermediate Filament Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/drug effects , Neurons/cytology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells/physiology , Time Factors
14.
Neuroreport ; 17(4): 377-81, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16514362

ABSTRACT

Neural stem cells are multipotent and self-renewing cells with important potential application in cell replacement therapy in brain damage. Many studies have shown that nestin-positive cells represent neural stem and progenitor cells in the central neural system. Here, we derived neural stem cells from the subventricular zone of a newborn nestin-promoter-driven green fluorescent protein mouse, and found that the percentage of nestin-positive cells decreased continuously at each passage in neurosphere culture. Using the relative proliferation ratio and relative division ratio analysis, we concluded that the slower cycling of nestin-positive cells was responsible for this decrease.


Subject(s)
Cell Culture Techniques/methods , Cell Cycle/physiology , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Stem Cell Transplantation/methods , Stem Cells/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Count , Cell Division/physiology , Cell Proliferation , Cell Separation/methods , Cells, Cultured , Green Fluorescent Proteins , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nestin , Neurons/cytology , Spheroids, Cellular , Stem Cells/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...