Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Front Biosci (Landmark Ed) ; 27(8): 239, 2022 08 11.
Article in English | MEDLINE | ID: mdl-36042179

ABSTRACT

BACKGROUND: Residual undifferentiated induced pluripotent stem cells (iPSCs) detection is essential for both Embryonic Stem Cells (ESCs) and iPSCs application in final cell therapy products. However, specific differentiated cells require specific genes for residual detection; identifying the suitable marker is costly and time-consuming. Thus, a universal marker for iPSCs residue detection for all three germline cells would greatly benefit PSC-derived cellular therapies. METHODS: Next-generation sequencing (NGS) was performed on total RNAs isolated from the iPSC cell lines and embryonic stem cells (H9), the top 30 expressed genes were selected as candidates. By analysis expression fold change comparing iPSC cells to the differentiated cells, seven genes were highly expressed in iPSCs but showed minimal background expression in differentiated cells. Tissue expression pattern of the candidate genes were explored in the Genotype-Tissue Expression (GTEx) project database, candidate genes were narrowed down to two genes. Spike-in experiments were performed to determine the detection limit and correlation with the number of iPSCs and gene expression by ddPCR. RESULTS: By next-generation sequencing (NGS), we identified two marker genes (ESRG and ZSCAN10) suitable for universal undifferentiated iPSC detection. Both ESRG and ZSCAN10 are highly expressed in iPSCs. ZSCAN10 is slightly expressed in the testis, pituitary, and cerebellum; ESRG is highly expressed in the vagina and scarcely expressed in the other tissues. Furthermore, the ddPCR method with a probe and primers for ESRG and ZSCAN10 detected a trace of undifferentiated hiPSCs to a spiked level of 0.0001%. CONCLUSIONS: These results suggest that targeting ESRG/ZSCAN10 transcripts is highly sensitive, quantitative, and could be broadly applied to quality control of almost all iPSC-derived cell therapy products.


Subject(s)
Induced Pluripotent Stem Cells , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Line , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male
2.
Clin Transl Med ; 12(6): e901, 2022 06.
Article in English | MEDLINE | ID: mdl-35696531

ABSTRACT

BACKGROUND: The chimeric antigen receptor NK-92 (CAR NK-92) cell targeting the prostate-specific membrane antigen (PSMA) has shown antitumour effects in castration-resistant prostate cancer (CRPC). However, the expression changes of programmed death ligand 1 (PD-L1) and its mechanisms on CAR NK-92 and CRPC cells and the effect of the anti-PD-L1 monoclonal antibody (mAb) on PD-L1 expressed on CAR NK-92 cells remain unknown. METHODS: Human dendritic cells and CD8+ T cells were acquired from blood samples of healthy donors and cocultured with C4-2 cells. Changes in PD-L1 expression were detected by flow cytometry. Differential gene expressions were investigated by RNA sequence analysis, while the regulation of PD-L1 molecular signaling was explored using western blotting. In vitro cytotoxicity was evaluated using the Cell Counting Kit-8 assay and the bioluminescent intensity (BLI) of green fluorescent protein-labelled C4-2 cells. CRPC growth in vivo was monitored using callipers and BLI in male NOD/SCID mice subcutaneously injected with C4-2 cells and treated intravenously with anti-PD-L1/PD-1 mAb, CAR NK-92 or cocultured CD8+ T cells. RESULTS: Significantly upregulated expression of PD-L1k was observed in cocultured C4-2 and CAR NK-92 cells. In addition, upregulation of PD-L1 expression was dependent on interferon-γ in C4-2 cells, while it was dependent on direct cell-to-cell interaction via the NK group 2 member D/ phosphatidylinositol 3-kinase/AKT pathway in CAR NK-92 cells. The anti-PD-L1 mAb directly acted on PD-L1 expressed on CAR NK-92 cells and augmented the cytotoxicity of CAR NK-92 cells against C4-2 and CRPC cells from one patient in vitro. Anti-PD-L1 mAb significantly enhanced the antitumour effect of CAR NK-92 cells against CRPC cells in vivo when compared to treatment with CAR NK-92 cells or combined with anti-PD-1 mAb in the absence or presence of cocultured CD8+ T cells. CONCLUSION: Combined treatment with CAR NK-92 and anti-PD-L1 mAb improved the antitumour efficacy against CRPC, which is of extraordinary translational value in the clinical treatment of CRPC.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Chimeric Antigen , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Humans , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/therapeutic use
3.
Cancer Commun (Lond) ; 42(8): 768-783, 2022 08.
Article in English | MEDLINE | ID: mdl-35706368

ABSTRACT

BACKGROUND: The mortality of castration-resistant prostate cancer (CRPC) is high due to lack of an effective treatment. Chimeric antigen receptor (CAR)-based therapy is a promising immunotherapeutic strategy. Here, we aimed to design a novel CAR-natural killer (NK) cells with a clinically significant tumoricidal effect on CRPC. METHODS: We constructed novel CAR-NK92MI cells with a CD244-based recombinant lentiviral vector. Different intracellular segments (CD244, NKG2D, or CD3ζ) were screened to identify the best candidate according to cell lysis assay and CD107a expression levels. To enhance the affinity of the CAR to the tumor antigen, we compared an antibody specific for prostate-specific membrane antigen (anti-PSMA) with PSMA-targeted polypeptide (p-PSMA), which was screened by phage display combinatorial library. Then, CAR-NK92MI cells with both a high affinity for PSMA and a strong tumoricidal capacity were generated. In addition, we verified their tumor-killing effect in vitro and in vivo. The release of cytokine by NK92MI cells was compared with that by CAR-NK92MI cells through flow cytometry and enzyme-linked immunosorbent assay. Moreover, ferroptosis-related cell death was explored as a possible underlying mechanism. RESULTS: Three different CAR intracellular regions CAR1 (CD244), CAR2 (CD244, NKG2D) and CAR3 (CD244, NKG2D, and CD3ζ) were constructed. CAR2 was chosen to confer a stronger tumoricidal ability on CAR-NK92MI cells. Compared with anti-PSMA, p-PSMA exhibited enhanced affinity for the tumor antigen. Thus, p-PSMA-CAR-NK92MI cells, which expressed CAR with a polypeptide-based antigen-binding region, an intracellular CD244 and a NKG2D costimulatory domain, were generated. They could selectively and successfully kill PSMA+ target cells and exhibited specific lysis rate of 73.19% for PSMA-positive C4-2 cells and 33.04% for PSMA-negative PC3 cells. Additionally, p-PSMA-CAR-NK92MI cells had significantly higher concentrations of IFN-γ, TNF-α and granzyme B than NK92MI cells. In a CRPC cancer xenograft model, p-PSMA-CAR-NK92MI cells significantly inhibited tumor growth and exerted a more consistent killing effect than NK92MI cells. Moreover, ferroptosis is a potential mechanism through which CAR-NK92MI cells attack cancer cells, and is triggered by IFN-γ. CONCLUSIONS: p-PSMA-CAR-NK92MI cells can effectively kill CRPCPSMA+ cells in vitro and in vivo. This strategy may provide additional treatment options for patients with CRPC.


Subject(s)
Ferroptosis , Prostatic Neoplasms, Castration-Resistant , Receptors, Chimeric Antigen , Antigens, Neoplasm , Cell Line, Tumor , Humans , Killer Cells, Natural , Male , NK Cell Lectin-Like Receptor Subfamily K/genetics , Peptides , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/therapy , Receptors, Chimeric Antigen/genetics
5.
Front Cell Dev Biol ; 9: 630654, 2021.
Article in English | MEDLINE | ID: mdl-33659254

ABSTRACT

Cystic Fibrosis (CF) is an autosomal recessive disorder caused by mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene. CF-related diabetes (CFRD) is one of the most prevalent comorbidities of CF. Altered glucose homeostasis has been reported in CF patients. The mechanism has not been fully elucidated. Besides the consequence of pancreatic endocrine dysfunction, we focus on insulin-responsive tissues and glucose transportation to explain glucose homeostasis alteration in CFRD. Herein, we found that CFTR knockout mice exhibited insulin resistance and glucose tolerance. Furthermore, we demonstrated insulin-induced glucose transporter 4 (GLUT4) translocation to the cell membrane was abnormal in the CFTR knockout mice muscle fibers, suggesting that defective intracellular GLUT4 transportation may be the cause of impaired insulin responses and glucose homeostasis. We further demonstrated that PI(4,5)P2 could rescue CFTR related defective intracellular GLUT4 transportation, and CFTR could regulate PI(4,5)P2 cellular level through PIP5KA, suggesting PI(4,5)P2 is a down-stream signal of CFTR. Our results revealed a new signal mechanism of CFTR in GLUT4 translocation regulation, which helps explain glucose homeostasis alteration in CF patients.

6.
Biochem Biophys Res Commun ; 534: 1020-1025, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33131771

ABSTRACT

Significant cellular morphology changes in renal tubules were observed in diabetes patients and animal models. However, the interaction between insulin and tubular epithelial cells microvillar structure remains obscure. To understand microvillar dynamics, we used Scanning Ion Conductance Microscope to visualize microvillar in the living cell. Here, we found two layers of microvilli on the tubular epithelial cell surface: short compact microvilli and netlike long microvilli. Insulin treatment could increase microvilli length and density. This process was mediated by the PI3K/PLCγ signaling pathway, other than the PI3K/Arp2/3 signal pathway. In conclusion, our findings present a novel insulin signaling transduction mechanism, which contributes to understanding renal tubular epithelial cell microvilli dynamic regulation.


Subject(s)
Epithelial Cells/metabolism , Insulin/metabolism , Kidney Tubules/metabolism , Microvilli/metabolism , Phospholipase C gamma/metabolism , Signal Transduction , Animals , Anura , Cell Line , Epithelial Cells/cytology , Humans , Kidney Tubules/cytology
7.
Int J Mol Sci ; 21(20)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33050503

ABSTRACT

Apart from its ion channel properties, the Kir2.1 channel has been found in tumors and cancer cells to facilitate cancer cell motility. It is assumed that Kir2.1 might be associated with cell actin filament dynamics. With the help of structured illumination microscopy (SIM), we show that Kir2.1 overexpression promotes actin filament dynamics, cell invasion, and adhesion. Mutated Kir2.1 channels, with impaired membrane expression, present much weaker actin regulatory effects, which indicates that precise Kir2.1 membrane localization is key to its actin filament remolding effect. It is found that Kir2.1 membrane expression and anchoring are associated with PIP2 affinity, and PIP2 depletion inhibits actin filament dynamics. We also report that membrane-expressed Kir2.1 regulates redistribution and phosphorylation of FLNA (filamin A), which may be the mechanism underlying Kir2.1 and actin filament dynamics. In conclusion, Kir2.1 membrane localization regulates cell actin filaments, and not the ion channel properties. These data indicate that Kir2.1 may have additional cellular functions distinct from the regulation of excitability, which provides new insight into the study of channel proteins.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Ion Channel Gating , Potassium Channels, Inwardly Rectifying/metabolism , Aquaporins/metabolism , Cell Adhesion/genetics , Cell Line , Cell Membrane/metabolism , Cell Movement/genetics , Fluorescent Antibody Technique , Gene Expression , Humans , Phosphorylation , Potassium Channels, Inwardly Rectifying/genetics , Protein Binding
8.
Oncogene ; 37(19): 2545-2558, 2018 05.
Article in English | MEDLINE | ID: mdl-29459713

ABSTRACT

Warburg effect has been recognized as a hallmark of cancer cells for many years, but its modulation mechanism remains a great focus. Our current study found a member of solute carrier family 25 (SLC25A29), the main arginine transporter on mitochondria, significantly elevated in various cancer cells. Knockout of SLC25A29 by CRISPR/Cas9 inhibited proliferation and migration of cancer cells both in vitro and in vivo. SLC25A29-knockout cells also showed an altered metabolic status with enhanced mitochondrial respiration and reduced glycolysis. All of above impacts could be reversed after rescuing SLC25A29 expression in SLC25A29-knockout cells. Arginine is transported into mitochondria partly for nitric oxide (NO) synthesis. Deletion of SLC25A29 resulted in severe decrease of NO production, indicating that the mitochondria is a significant source of NO. SLC25A29-knockout cells dramatically altered the variation of metabolic processes, whereas addition of arginine failed to reverse the effect, highlighting the necessity of transporting arginine into mitochondria by SLC25A29. In conclusion, aberrant elevated SLC25A29 in cancer functioned to transport more arginine into mitochondria, improved mitochondria-derived NO levels, thus modulated metabolic status to facilitate increased cancer progression.


Subject(s)
Carnitine Acyltransferases/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Neoplasms/metabolism , Nitric Oxide/metabolism , Up-Regulation , Animals , Arginine/metabolism , Carnitine Acyltransferases/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Glycolysis , HeLa Cells , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mitochondrial Proteins/metabolism , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/pathology
9.
PLoS Genet ; 14(1): e1007165, 2018 01.
Article in English | MEDLINE | ID: mdl-29370161

ABSTRACT

Intellectual disability (ID), one of the most common human developmental disorders, can be caused by genetic mutations in Cullin 4B (Cul4B) and cereblon (CRBN). CRBN is a substrate receptor for the Cul4A/B-DDB1 ubiquitin ligase (CRL4) and can target voltage- and calcium-activated BK channel for ER retention. Here we report that ID-associated CRL4CRBN mutations abolish the interaction of the BK channel with CRL4, and redirect the BK channel to the SCFFbxo7 ubiquitin ligase for proteasomal degradation. Glioma cell lines harbouring CRBN mutations record density-dependent decrease of BK currents, which can be restored by blocking Cullin ubiquitin ligase activity. Importantly, mice with neuron-specific deletion of DDB1 or CRBN express reduced BK protein levels in the brain, and exhibit similar impairment in learning and memory, a deficit that can be partially rescued by activating the BK channel. Our results reveal a competitive targeting of the BK channel by two ubiquitin ligases to achieve exquisite control of its stability, and support changes in neuronal excitability as a common pathogenic mechanism underlying CRL4CRBN-associated ID.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Learning/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Proteolysis , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/physiology , Adaptor Proteins, Signal Transducing , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
10.
Curr Drug Targets ; 19(3): 279-298, 2018 02 08.
Article in English | MEDLINE | ID: mdl-28356028

ABSTRACT

BACKGROUND: Stem cell therapy provides great promising therapeutic benefits for various neurological disorders. Cell transplantation has emerged as cell replacement application for nerve damage. Recently, nanomaterials obtain wide development in various industrial and medical fields, and nanoparticles have been applied in the neurological field for tracking and treating nervous system diseases. Combining stem cells with nanotechnology has raised more and more attentions; and it has demonstrated that the combination has huge effects on clinical diagnosis and therapeutics in multiple central nervous system diseases, meanwhile, improves prognosis. OBJECTIVE: The aim of this review was to give a brief overview of the application of nanomaterials in stem cell therapy for neurological diseases. RESULTS: Nanoparticles not only promote stem cell proliferation and differentiation in vitro or in vivo, but also play dominant roles on stem cell imaging and tracking. Furthermore, via delivering genes or drugs, nanoparticles can participate in stem cell therapeutic applications for various neurological diseases, such as ischemic stroke, spinal cord injury (SCI), multiple sclerosis (MS), Parkinson's disease (PD), Alzheimer's disease (AD) and gliomas. However, nanoparticles have potential cytotoxic effects on nerve cells, which are related to their physicochemical properties. CONCLUSION: Nano-stem cell-based therapy as a promising strategy has the ability to affect neuronal repair and regeneration in the central nervous system.


Subject(s)
Nanotechnology/methods , Nervous System Diseases/therapy , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Cell Tracking , Humans , Nanostructures
11.
Sci Rep ; 7(1): 14749, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116138

ABSTRACT

Peripheral vascular occlusive disease (PVOD) is a common manifestation of atherosclerosis, and it has a high rate of morbidity. Therapeutic angiogenesis would re-establish blood perfusion and rescue ischemic tissue. Vascular endothelial growth factor (VEGF) induces angiogenesis and can potentially be used to treat ischemic diseases, yet in clinical trials VEGF has not fulfilled its full potential with side effects. Whether amino acids promote angiogenesis and the molecular mechanisms are largely unknown. Here we showed that (1) Glycine significantly promoted angiogenesis both in vitro and in vivo and effectively protected mitochondrial function. (2) Activation of glycine transporter 1(GlyT1) induced by VEGF led to an increase in intracellular glycine. (3) Glycine directly bounded to voltage dependent anion channel 1 (VDAC1) on the mitochondrial outer membrane and inhibited its opening. These original results highlight glycine as a necessary mediator in VEGF signalling via the GlyT1-glycine-mTOR-VDAC1 axis pathway. Therefore, the findings in this study are of significance providing new mechanistic insights into angiogenesis and providing better understanding of glycine function in angiogenesis, which may provide valuable information for development of novel therapeutic targets for the treatment of angiogenic vascular disorders.


Subject(s)
Glycine/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Animals , Glycine Plasma Membrane Transport Proteins/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism
12.
Mol Med Rep ; 16(2): 2259-2263, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28656309

ABSTRACT

Nifedipine is widely used to treat high blood pressure and angina. Were nifedipine able to promote the proliferation and migration of breast cancer, it would pose a significant threat for patients. Thus, it is important to determine the effects of nifedipine on breast cancer and the mechanism involved. The present study identified that nifedipine significantly promoted the proliferation and migration of breast cancer cells in vitro. The mechanism of nifedipine on different breast cancer cells was investigated and it was identified that the effects of nifedipine on MCF­7 cells were via the protein kinase B­endothelial constitutive nitric oxide synthase­nitric oxide axis, and on MDA­MB­231 cells via activation of the extracellular signal­regulated kinase pathway. These results identified the distinct pathways in the activation of cell proliferation and migration presented in different cell lines by nifedipine. The present study advises that nifedipine can promote breast cancer and should be avoided for women who suffer from breast cancer and hypertension.


Subject(s)
Cell Proliferation/drug effects , Nifedipine/pharmacology , Breast Neoplasms , Calcium Channels/chemistry , Calcium Channels/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , MCF-7 Cells , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
13.
Sci Rep ; 7: 43546, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28272473

ABSTRACT

Serotonin or 5-hydroxytryptamine (5-HT) has been shown to be essential in lots of physiological and pathological processes. It is well known that 5-HT and 5-HT transporter (5-HTT) play important roles in the pulmonary artery in pulmonary hypertension. However, little is known about the function of 5-HTT in other arteries. In this study we found that the expression of 5-HTT was elevated in injured carotid arteries and over-expression of 5-HTT induced proliferation of smooth muscle cells (SMCs); however, this phenotype could be reversed by knocking-down of 5-HTT or endothelial cells conditional medium (EC-CM). A 5-HTT inhibitor, fluoxetine, treated animals also exhibited reduced restenosis after injury. We identified that miR-195 was packaged in the extracellular vesicles from EC-CM. We further confirmed that extracellular vesicles could transfer miR-195 from ECs to SMCs to inhibit the expression of 5-HTT in SMCs and the proliferation of SMCs. These results provide the first evidence that ECs communicate with SMCs via micro-RNA195 in the regulation of the proliferation of SMCs through 5-HTT, which will contribute to a better understanding of communications between ECs and SMCs via micro-RNA. Our findings suggest a potential target for the control of vessel restenosis.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , RNA Interference , Serotonin Plasma Membrane Transport Proteins/genetics , Animals , Biological Transport , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Gene Expression Regulation/drug effects , Humans , Myocytes, Smooth Muscle/drug effects , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Rats
14.
Vascul Pharmacol ; 88: 11-20, 2017 01.
Article in English | MEDLINE | ID: mdl-27884759

ABSTRACT

Vascular endothelial growth factor (VEGF) signaling is tightly regulated by specific VEGF receptors (VEGF-R). Recently, we identified heterodimerisation between VEGFR-1 and VEGFR-2 (VEGFR1-2) to regulate VEGFR-2 function. However, both the mechanism of action and the relationship with VEGFR-1 homodimers remain unknown. The current study shows that activation of VEGFR1-2, but not VEGFR-1 homodimers, inhibits VEGFR-2 receptor phosphorylation under VEGF stimulation in human endothelial cells. Furthermore, inhibition of phosphatidylinositol 3-kinase (PI3K) increases VEGFR-2 phosphorylation under VEGF stimulation. More importantly, inhibition of PI3K pathway abolishes the VEGFR1-2 mediated inhibition of VEGFR-2 phosphorylation. We further demonstrate that inhibition of PI3K pathway promotes capillary tube formation. Finally, the inhibition of PI3K abrogates the inhibition of in vitro angiogenesis mediated by VEGFR1-2 heterodimers. These findings demonstrate that VEGFR1-2 heterodimers and not VEGFR-1 homodimers inhibit VEGF-VEGFR-2 signaling by suppressing VEGFR-2 phosphorylation via PI3K pathway.


Subject(s)
Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neovascularization, Physiologic/physiology , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Protein Multimerization , Signal Transduction , Swine
15.
Sci Rep ; 6: 37336, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27874067

ABSTRACT

5-HT has been reported to possess significant effects on cardiac activities, but activation of 5-HTR on the cell membrane failed to illustrate the controversial cardiac reaction. Because 5-HT constantly comes across the cell membrane via 5-HT transporter (5-HTT) into the cytoplasm, whether 5-HTR is functional present on the cellular organelles is unknown. Here we show 5-HTR3 and 5-HTR4 were located in cardiac mitochondria, and regulated mitochondrial activities and cellular functions. Knock down 5-HTR3 and 5-HTR4 in neonatal cardiomyocytes resulted in significant increase of cell damage in response to hypoxia, and also led to alternation in heart beating. Activation of 5-HTR4 attenuated mitochondrial Ca2+ uptake under the both normoxic and hypoxic conditions, whereas 5-HTR3 augmented Ca2+ uptake only under hypoxia. 5-HTR3 and 5-HTR4 exerted the opposite effects on the mitochondrial respiration: 5-HTR3 increased RCR (respiration control ratio), but 5-HTR4 reduced RCR. Moreover, activation of 5-HTR3 and 5-HTR4 both significantly inhibited the opening of mPTP. Our results provided the first evidence that 5-HTR as a GPCR and an ion channel, functionally expressed in mitochondria and participated in the mitochondria function and regulation to maintain homeostasis of mitochondrial [Ca2+], ROS, and ATP generation efficiency in cardiomyocytes in response to stress and O2 tension.


Subject(s)
Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Cell Hypoxia , Cells, Cultured , Mice, Inbred BALB C , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA Interference , Receptors, Serotonin, 5-HT3/genetics , Receptors, Serotonin, 5-HT4/genetics
16.
Sci Rep ; 6: 35475, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27762288

ABSTRACT

Puerarin, a known isoflavone, is commonly found as a Chinese herb medicine. It is widely used in China to treat cardiac diseases such as angina, cardiac infarction and arrhythmia. However, its cardioprotective mechanism remains unclear. In this study, puerarin significantly prolonged ventricular action potential duration (APD) with a dosage dependent manner in the micromolar range on isolated rat ventricular myocytes. However, submicromolar puerarin had no effect on resting membrane potential (RMP), action potential amplitude (APA) and maximal velocity of depolarization (Vmax) of action potential. Only above the concentration of 10 mM, puerarin exhibited more aggressive effect on action potential, and shifted RMP to the positive direction. Millimolar concentrations of puerarin significantly inhibited inward rectified K+ channels in a dosage dependent manner, and exhibited bigger effects upon Kir2.1 vs Kir2.3 in transfected HEK293 cells. As low as micromolar range concentrations of puerarin significantly inhibited Kv7.1 and IKs. These inhibitory effects may due to the direct inhibition of puerarin upon channels not via the PKA-dependent pathway. These results provided direct preclinical evidence that puerarin prolonged APD via its inhibitory effect upon Kv7.1 and IKs, contributing to a better understanding the mechanism of puerarin cardioprotection in the treatment of cardiovascular diseases.


Subject(s)
Action Potentials/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Isoflavones/pharmacology , Vasodilator Agents/pharmacology , Ventricular Function/drug effects , Adenosine Diphosphate/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Signal Transduction/drug effects
17.
Stem Cells Int ; 2016: 6129579, 2016.
Article in English | MEDLINE | ID: mdl-27656217

ABSTRACT

Objective. Systematic review and meta-analysis to observe the efficacy and safety of stem cell transplantation therapy in patients with brain ischemia. Methods. We searched Cochrane Library, PubMed, Ovid, CBM, CNKI, WanFang, and VIP Data from its inception to December 2015, to collect randomized controlled trials (RCT) of stem cell transplantation for the ischemic stroke. Two authors independently screened the literature according to the inclusion and exclusion criteria, extracted data, and assessed the risk of bias. Thereafter, meta-analysis was performed. Results. Sixteen studies and eighteen independent treatments were included in the current meta-analysis. The results based upon the pooled mean difference from baseline to follow-up points showed that the stem cell transplantation group was superior to the control group with statistical significance in the neurologic deficits score (NIHSS, MD = 1.57; 95% CI, 0.64-2.51; I (2) = 57%; p = 0.001), motor function (FMA, MD = 4.23; 95% CI, 3.08-5.38; I (2) = 0%; p < 0.00001), daily life ability (Barthel, MD = 8.37; 95% CI, 4.83-11.91; I (2) = 63%; p < 0.00001), and functional independence (FIM, MD = 8.89; 95% CI, 4.70-13.08; I (2) = 79%; p < 0.0001). Conclusions. It is suggested that the stem cell transplantation therapy for patients with brain ischemic stroke can significantly improve the neurological deficits and daily life quality, with no serious adverse events. However, higher quality and larger data studies are required for further investigation to support clinical application of stem cell transplantation.

18.
Sci Rep ; 6: 32291, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27554433

ABSTRACT

To evaluate the preclinical studies using NSCs transplantation therapy for experimental ischemic stroke, and determine the effect size of NSCs therapy and the correlations between different clinical measures. We firstly searched literatures to identify studies of NSCs therapy in animal cerebral ischemia models, and then calculated the quality score of studies, assessed the effect size of NSCs therapy relative to behavioral and histologic endpoints by meta-analysis. A total of 37 studies and 54 independent treated interventions were used for systematic review and meta-analysis. The median quality score was 5 of 10. 36 studies (53 intervention arms) reported functional outcome, 22 studies (34 intervention arms) reported structural outcome. After adjusted by subgroup and sensitivity analysis, the mean effect sizes were improved by 1.35 for mNSS, 1.84 for rotarod test, 0.61 for cylinder test, and 0.84 for infarct volume. Furthermore, effect size had a certain interaction with clinical variables, for example early NSCs therapy etc. In this preclinical studies, we demonstrated that transplanted NSCs significantly improved outcomes (both functional and structural outcome) in ischemic stroke. It is suggested that future preclinical animal model studies of stroke should improve study quality validity and reduce potentially confounded publication bias.


Subject(s)
Brain Ischemia/therapy , Neural Stem Cells/transplantation , Stem Cell Transplantation , Stroke/therapy , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Neural Stem Cells/metabolism , Stroke/metabolism , Stroke/pathology
19.
Zhonghua Nan Ke Xue ; 22(2): 110-5, 2016 Feb.
Article in Chinese | MEDLINE | ID: mdl-26939393

ABSTRACT

OBJECTIVE: To study the impact of the chloride channel dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) on the cytoskeleton of Sertoli cells in the mouse. METHODS: TM4 Sertoli cells were cultured and treated with CFTR(inh)-172 at the concentrations of 1, 5, 10 and 20 µmol/L for 48 hours. Then the cytotoxicity of CFT(inh)-172 was assessed by CCK-8 assay, the expressions of F-actin and Ac-tub in the TM4 Sertoli cells detected by immunofluorescence assay, and those of N-cadherin, vimentin and vinculin determined by qPCR. RESULTS: CFTR(inh)-172 produced cytotoxicity to the TM4 Sertoli cells at the concentration of 20 µmol/L. The expressions of F-actin and Ac-tub were decreased gradually in the TM4 Sertoli cells with the prolonging of treatment time and increasing concentration of CFTR(inh)-172 (P < 0.05). The results of qPCR showed that different concentrations of CFTR(inh)-172 worked no significant influence on the mRNA expressions of N-cadherin, vimentin and vinculin in the Sertoli cells. CONCLUSION: The CFTR chloride channel plays an important role in maintaining the normal cytoskeleton of Sertoli cells. The reduced function and expression of the CFTR chloride channel may affect the function of Sertoli cells and consequently spermatogenesis of the testis.


Subject(s)
Benzoates/pharmacology , Chloride Channels/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cytoskeleton/drug effects , Sertoli Cells/drug effects , Thiazolidines/pharmacology , Actins/metabolism , Animals , Male , Mice , Sertoli Cells/metabolism , Spermatogenesis , Time Factors
20.
J Mol Cell Cardiol ; 91: 179-87, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26774701

ABSTRACT

AIMS: Pulmonary arterial hypertension [1] is a proliferative disorder associated with enhanced proliferation and suppressed apoptosis of pulmonary artery smooth muscle cells (PASMCs). Reactive oxygen species (ROS) is implicated in the development of PAH and regulates the vascular tone and functions. However, which cellular signaling mechanisms are triggered by ROS in PAH is still unknown. Hence, here we wished to characterize the signaling mechanisms triggered by ROS. METHODS AND RESULTS: By Western blots, we showed that increased intracellular ROS caused inhibition of the glycolytic pyruvate kinase M2 (PKM2) activity through promoting the phosphorylation of PKM2. Monocrotaline (MCT)-induced rats developed severe PAH and right ventricular hypertrophy, with a significant increase in the P-PKM2 and decrease in pyruvate kinase activity which could be attenuated with the treatments of PKM2 activators, FBP and l-serine. The antioxidant NAC, apocynin and MnTBAP had the similar protective effects in the development of PAH. In vitro assays confirmed that inhibition of PKM2 activity could modulate the flux of glycolytic intermediates in support of cell proliferation through the increased pentose phosphate pathway (PPP). Increased ROS and decreased PKM2 activity also promoted the Cav1.2 expression and intracellular calcium. CONCLUSION: Our data provide new evidence that PKM2 makes a critical regulatory contribution to the PAHs for the first time. Decreased pyruvate kinase M2 activity confers additional advantages to rat PASMCs by allowing them to sustain anti-oxidant responses and thereby support cell survival in PAH. It may become a novel treatment strategy in PAH by using of PKM2 activators.


Subject(s)
Hypertension, Pulmonary/enzymology , Hypertrophy, Right Ventricular/enzymology , Myocytes, Smooth Muscle/enzymology , Pulmonary Artery/enzymology , Pyruvate Kinase/genetics , Reactive Oxygen Species/metabolism , Acetophenones/pharmacology , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/pathology , Male , Metalloporphyrins/pharmacology , Monocrotaline , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Organ Culture Techniques , Pentose Phosphate Pathway , Phosphorylation , Primary Cell Culture , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pyruvate Kinase/metabolism , Rats , Rats, Sprague-Dawley , Serine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...