Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Stroke ; 55(6): 1660-1671, 2024 Jun.
Article En | MEDLINE | ID: mdl-38660789

BACKGROUND: Activation of the acid-sensing ion channels (ASICs) by tissue acidosis, a common feature of brain ischemia, contributes to ischemic brain injury, while blockade of ASICs results in protection. Cholestane-3ß,5α,6ß-triol (Triol), a major cholesterol metabolite, has been demonstrated as an endogenous neuroprotectant; however, the mechanism underlying its neuroprotective activity remains elusive. In this study, we tested the hypothesis that inhibition of ASICs is a potential mechanism. METHODS: The whole-cell patch-clamp technique was used to examine the effect of Triol on ASICs heterogeneously expressed in Chinese hamster ovary cells and ASICs endogenously expressed in primary cultured mouse cortical neurons. Acid-induced injury of cultured mouse cortical neurons and middle cerebral artery occlusion-induced ischemic brain injury in wild-type and ASIC1 and ASIC2 knockout mice were studied to examine the protective effect of Triol. RESULTS: Triol inhibits ASICs in a subunit-dependent manner. In Chinese hamster ovary cells, it inhibits homomeric ASIC1a and ASIC3 without affecting ASIC1ß and ASIC2a. In cultured mouse cortical neurons, it inhibits homomeric ASIC1a and heteromeric ASIC1a-containing channels. The inhibition is use-dependent but voltage- and pH-independent. Structure-activity relationship analysis suggests that hydroxyls at the 5 and 6 positions of the A/B ring are critical functional groups. Triol alleviates acidosis-mediated injury of cultured mouse cortical neurons and protects against middle cerebral artery occlusion-induced brain injury in an ASIC1a-dependent manner. CONCLUSIONS: Our study identifies Triol as a novel ASIC inhibitor, which may serve as a new pharmacological tool for studying ASICs and may also be developed as a potential drug for treating stroke.


Acid Sensing Ion Channels , Acidosis , Cricetulus , Mice, Knockout , Animals , Acid Sensing Ion Channels/metabolism , Acid Sensing Ion Channels/genetics , Mice , CHO Cells , Acidosis/metabolism , Acidosis/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , Neurons/drug effects , Neurons/metabolism , Cricetinae , Neuroprotective Agents/pharmacology , Cholestanols/pharmacology , Mice, Inbred C57BL , Acid Sensing Ion Channel Blockers/pharmacology , Male , Cells, Cultured
2.
Vet Sci ; 10(7)2023 Jul 11.
Article En | MEDLINE | ID: mdl-37505861

Free radicals, including reactive oxygen species (ROS) and reactive nitrogen species (RNS), play critical roles in various physiological activities such as cell differentiation, apoptosis, and vascular tension when existing in cells at low levels. However, excessive amounts of free radicals are harmful, causing DNA damage, lipid peroxidation, protein degeneration, and abnormal cell death. Certain viral infections induce cells to produce excessive free radicals, which in multiple ways help the virus to replicate, mature, and exit. Iron is a necessary element for many intracellular enzymes, involved in both cellular activities and viral replication. Ferroptosis, a programmed cell death mode distinct from apoptosis, necrosis, and pyroptosis, is characterized by lipid peroxide accumulation and damage to the antioxidant system, affecting many cellular processes. Viral infection commonly manifests as decreased glutathione (GSH) content and down-regulated glutathione peroxidase 4 (GPX4) activity, similar to ferroptosis. Recent studies have suggested a possible relationship among free radicals, viral infections and ferroptosis. This review aims to elucidate the molecular mechanism linking free radicals and ferroptosis during viral infections and provide a new theoretical basis for studying viral pathogenesis and control.

3.
Biomolecules ; 13(3)2023 03 10.
Article En | MEDLINE | ID: mdl-36979442

KB-R7943, an isothiourea derivative, is widely used as a pharmacological inhibitor of reverse sodium-calcium exchanger (NCX). It has been shown to have neuroprotective and analgesic effects in animal models; however, the detailed molecular mechanisms remain elusive. In the current study, we investigated whether KB-R7943 modulates acid-sensing ion channels (ASICs), a group of proton-gated cation channels implicated in the pathophysiology of various neurological disorders, using the whole-cell patch clamp techniques. Our data show that KB-R7943 irreversibly inhibits homomeric ASIC1a channels heterologously expressed in Chinese Hamster Ovary (CHO) cells in a use- and concentration-dependent manner. It also reversibly inhibits homomeric ASIC2a and ASIC3 channels in CHO cells. Both the transient and sustained current components of ASIC3 are inhibited. Furthermore, KB-R7943 inhibits ASICs in primary cultured peripheral and central neurons. It inhibits the ASIC-like currents in mouse dorsal root ganglion (DRG) neurons and the ASIC1a-like currents in mouse cortical neurons. The inhibition of the ASIC1a-like current is use-dependent and unrelated to its effect on NCX since neither of the other two well-characterized NCX inhibitors, including SEA0400 and SN-6, shows an effect on ASIC. Our data also suggest that the isothiourea group, which is lacking in other structurally related analogs that do not affect ASIC1a-like current, may serve as a critical functional group. In summary, we characterize KB-R7943 as a new ASIC inhibitor. It provides a novel pharmacological tool for the investigation of the functions of ASICs and could serve as a lead compound for developing small-molecule drugs for treating ASIC-related disorders.


Acid Sensing Ion Channels , Sodium-Calcium Exchanger , Cricetinae , Mice , Animals , Cricetulus , Sodium-Calcium Exchanger/genetics , CHO Cells
4.
Front Vet Sci ; 8: 640067, 2021.
Article En | MEDLINE | ID: mdl-33681335

Mycoplasma synoviae is an important pathogen of poultry, causing significant economic losses in this industry. Analysis of the unique genes and shared genes among different M. synoviae strains and among related species is helpful for studying the molecular pathogenesis of M. synoviae and provides valuable molecular diagnostic targets to facilitate the identification of M. synoviae species. We selected a total of 46 strains, including six M. synoviae strains, from 25 major animal (including avian) Mycoplasma species/subspecies that had complete genome sequences and annotation information published in GenBank, and used them for comparative genomic analysis. After analysis, 16 common genes were found in the 46 strains. Thirteen single-copy core genes and the 16s rRNA genes were used for genetic evolutionary analysis. M. synoviae was found to have a distant evolutionary relationship not only with other arthritis-causing mycoplasmas, but also with another major avian pathogen, Mycoplasma gallisepticum, that shares the major virulence factor vlhA with M. synoviae. Subsequently, six unique coding genes were identified as shared among these M. synoviae strains that are absent in other species with published genome sequences. Two of the genes were found to be located in the genetically stable regions of the genomes of M. synoviae and were determined to be present in all M. synoviae isolated strains (n = 20) and M. synoviae-positive clinical samples (n = 48) preserved in our laboratory. These two genes were used as molecular diagnostic targets for which SYBR green quantitative PCR detection methods were designed. The two quantitative PCR methods exhibited good reproducibility and high specificity when tested on positive plasmid controls and genomic DNA extracted from different M. synoviae strains, other major avian pathogenic bacteria/mycoplasmas, and low pathogenic Mycoplasma species. The detection limit for the two genes was 10 copies or less per reaction. The clinical sensitivity and specificity of the quantitative PCR methods were both 100% based on testing chicken hock joint samples with positive or negative M. synoviae infection. This research provides a foundation for the study of species-specific differences and molecular diagnosis of M. synoviae.

5.
Microb Pathog ; 154: 104829, 2021 May.
Article En | MEDLINE | ID: mdl-33727170

Mycoplasma synoviae (MS) infection causes infectious synovitis and arthritis with hyperplasia of synovial cells in the chicken joint. However, its mechanism is unknown. We used primary chicken synovial fibroblast (CSF) as the research object to study the role of MS in the proliferation of MS-infected CSF and determine the mechanisms involved. Using integrated transcriptomic and proteomic analyses of the interaction between CSF and MS, we screened a proliferation-regulated factor, serum amyloid A (SAA), that may regulate proliferation of MS-infected CSF. SAA appears to be associated with MS-induced CSF proliferation. To study the role of SAA in MS-induced CSF proliferation, a eukaryotic expression vector overexpressing SAA and a small interfering RNA (siRNA) targeting Saa were constructed to manipulate the expression of SAA. Cell proliferation and apoptosis were detected via cell counting kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU), or terminal deoxyribonucleotidyl transferase-mediated dUTP nick-dnd labeling (TUNEL) assays, respectively. Western blot analysis was used to examine the protein expression level of SAA, cyclin E1, and cyclin-dependent kinase 2 (CDK2). In vitro, MS significantly promoted the proliferation of CSF and increased the production of SAA. Overexpression of SAA accelerated the proliferative ability of CSF, whereas knockdown of SAA depressed the proliferative ability of CSF. A TUNEL assay indicated that MS did not induce apoptosis. Silencing of SAA suppressed the expression of cyclin E1 and CDK2. These results suggest that MS may upregulate the expression of SAA, accelerate the cell cycle, and promote proliferation of CSF.


Arthritis, Rheumatoid , Mycoplasma synoviae , Animals , Cell Proliferation , Chickens/metabolism , Fibroblasts/metabolism , Mycoplasma synoviae/metabolism , Proteomics , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , Synovial Membrane , Up-Regulation
6.
Poult Sci ; 99(11): 5366-5377, 2020 Nov.
Article En | MEDLINE | ID: mdl-33142453

Mycoplasma synoviae (MS) is an important avian pathogen causing considerable economic hardship in the poultry industry. A major inflammation caused by MS is synovitis that occurs in the synovial tendon sheath and joint synovium. However, the overall appearance of pathological changes in the tendon sheath and surrounding tissues caused by MS infection at the level of pathological tissue sections was poor. Studies on the role of MS and synovial sheath cells (SSCs) interaction in the development of synovitis have not been carried out. Through histopathological observation, our study found that a major MS-induced pathological change of the tendon sheath synovium was extensive scattered and focal inflammatory cell infiltration of the tendon sheath synovial layer. In vitro research experiments revealed that the CFU numbers of MS adherent and invading SSC, the levels of expression of various pattern recognition receptors, inflammatory cytokines, and chemokines coding genes, such as IL-1ß, IL-6, IL-8, CCL-20, RANTES, MIP-1ß, TLR7, and TLR15 in SSCs, and chemotaxis of macrophages were significantly increased when the multiplicity of infection (MOI) of MS to SSC were increased tenfold. The expression level of IL-12p40 in SSC was significantly higher when the MOIs of MS to SSC were increased by a factor of 100. The interaction between MS and SSC can activate macrophages, which was manifested by a significant increase in the expression of IL-1ß, IL-6, IL-8, CCL-20, RANTES, MIP-1ß, and CXCL-13. This study systematically demonstrated that the interaction of MS with chicken SSC contributes to the inflammatory response caused by the robust expression of related cytokines and macrophage chemotaxis. These findings are helpful in elucidating the molecular mechanism of MS-induced synovitis in chickens.


Chickens , Host-Pathogen Interactions , Joint Capsule , Mycoplasma Infections , Mycoplasma synoviae , Animals , Cytokines/genetics , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Inflammation/veterinary , Joint Capsule/cytology , Joint Capsule/microbiology , Macrophages/cytology , Macrophages/microbiology , Mycoplasma Infections/physiopathology , Mycoplasma Infections/veterinary
7.
Am J Transl Res ; 12(9): 5563-5574, 2020.
Article En | MEDLINE | ID: mdl-33042438

INTRODUCTION: Hyperoxia-induced injury is a common form of damage in lung tissues, which could lead to bronchopulmonary dysplasia (BPD) in newborns. Recent studies have discovered that FOXA2 played a substantial role in protecting lung tissues from various injuries and lncRNA-NEF could activate the expression of FOXA2. However, it is unclear whether lncRNA-NEF could alleviate hyperoxia-caused damage of lung tissues by activating FOXA2. MATERIAL AND METHODS: In this study, we used the lentivirus to establish the lncRNA-NEF overexpression RLE-6TN and MLE-12 cells. After that, the lentivirus was also used to knockdown the expression of FOXA2 in the two lncRNA-NEF overexpression cells. ELISA was performed to detect the levels of TNF-α, IL-1ß and IL-6. The production of ROS, SOD, MDA and LDH was determined with the commercial kits. The apoptosis rates of these cells were measured with the flow cytometry. RESULTS: The secretion of TNF-α, IL-1ß and IL-6 was inhibited in RLE-6TN and MLE-12 cells after the overexpression of lncRNA-NEF. Furthermore, the production of ROS, MDA and LDH was also suppressed after the upregulation of lncRNA-NEF. The promotion of lncRNA-NEF also restricted the hyperoxia-induced apoptosis. However, the knockdown of FOXA2 abolished all the inhibitory effects exerted by lncRNA-NEF. CONCLUSION: LncRNA-NEF regulated hyperoxia-caused inflammatory response, oxidative damage and apoptosis of RLE-6TN and MLE-12 cells by affecting the expression of FOXA2.

8.
Vaccine ; 38(30): 4695-4703, 2020 06 19.
Article En | MEDLINE | ID: mdl-32446833

Duck hepatitis A virus type 3 (DHAV-3) is an important pathogen that causes substantial losses in the Chinese duck industry. DHAV-3 is highly fatal to ducklings and there is no licensed vaccine in China available to reduce DHAV-3 infection. Our goal was to develop a live attenuated vaccine candidate against DHAV-3. A field isolated strain, SD, was attenuated by serially passaging in specific-pathogen-free (SPF) chicken embryos, and it lost its pathogenicity after 40 passages. The 70th passaged strain (SD70), which achieved good growth capacity in chicken embryos with a viral titer of 107.5 ELD50/mL, was chosen to be the live attenuated vaccine candidate. The SD70 strain did not cause clinical signs of disease or mortality in 1-day-old ducklings and showed no virulence reversion after seven rounds of in vivo back passages. The minimum effective dose of SD70 was determined to be 102.5 ELD50 via the vaccination route of subcutaneous inoculation. A single dose of the SD70 provided good protection to susceptible ducklings against the lethal DHAV-3 strain. Compared with the genomic sequence of the parent SD strain, the SD70 had 12 amino acid substitutions, some of which may play a role in virulence attenuation. This study demonstrated that the attenuated SD70 strain is a promising vaccine candidate for the prevention of DHAV-3 infection in China. It exhibited safety, good stability and excellent protection.


Hepatitis Virus, Duck , Hepatitis, Viral, Animal , Picornaviridae Infections , Poultry Diseases , Animals , Chick Embryo , China , Ducks , Hepatitis, Viral, Animal/prevention & control , Picornaviridae Infections/prevention & control , Picornaviridae Infections/veterinary , Poultry Diseases/prevention & control , Vaccines, Attenuated
9.
Front Microbiol ; 11: 576, 2020.
Article En | MEDLINE | ID: mdl-32318048

Mycoplasma synoviae (MS), which causes respiratory disease, eggshell apex abnormalities, infectious synovitis, and arthritis in avian species, has become an economically detrimental poultry pathogen in recent years. In China, the disease is characterized by infectious synovitis and arthritis. However, the mechanism by which MS causes infectious synovitis and arthritis remains unknown. Increasing evidence suggests that synovial fibroblasts (SF) play a key role in the pathogenesis of arthritis. Here, both RNA sequencing and tandem mass tag analyses are utilized to compare the response of primary chicken SF (CSF) following infection with and without MS. The host response between non-infected and infected cells was remarkably different at both the mRNA and protein levels. In total, 2,347 differentially expressed genes (DEGs) (upregulated, n = 1,137; downregulated, n = 1,210) and 221 differentially expressed proteins (DEPs) (upregulated, n = 129; downregulated, n = 92) were detected in the infected group. A correlation analysis indicated a moderate positive correlation between the mRNA and protein level changes in MS-infected CSF. At both the transcriptomic and proteomic levels, 149 DEGs were identified; 88 genes were upregulated and 61 genes were downregulated in CSF. Additionally, part of these regulated genes and their protein products were grouped into seven categories: proliferation-related and apoptosis-related factors, inflammatory mediators, proangiogenic factors, antiangiogenic factors, matrix metalloproteinases, and other arthritis-related proteins. These proteins may be involved in the pathogenesis of MS-induced arthritis in chickens. To our knowledge, this is the first integrated analysis on the mechanism of CSF-MS interactions that combined transcriptomic and proteomic technologies. In this study, many key candidate genes and their protein products related to MS-induced infectious synovitis and arthritis were identified.

10.
Microbiol Resour Announc ; 9(6)2020 Feb 06.
Article En | MEDLINE | ID: mdl-32029555

Here, we report the complete genome sequence of Mycoplasma synoviae HN01, a virulent epidemic strain isolated from a sick chicken with synovitis in Henan Province, China. HN01 is the Asian source of an M. synoviae strain that is completely sequenced, genome annotated, and published with relevant data.

11.
BMC Genomics ; 21(1): 123, 2020 Feb 04.
Article En | MEDLINE | ID: mdl-32019511

BACKGROUND: Duck hepatitis A virus type 3 (DHAV-3) is one of the most harmful pathogens in the duck industry. However, the molecular mechanism underlying DHAV-3 infection in ducklings remains poorly understood. To study the genetic regulatory network for miRNA-mRNA and the signaling pathways involved in DHAV-3 infection in ducklings, we conducted global miRNA and mRNA expression profiling of duckling liver tissues infected with lethal DHAV-3 by high-throughput sequencing. RESULTS: We found 156 differentially expressed miRNAs (DEMs) and 7717 differentially expressed genes (DEGs) in livers of mock-infected and DHAV-3-infected duckling. A total of 19,606 miRNA-mRNA pairs with negatively correlated expression patterns were identified in miRNA-mRNA networks constructed on the basis of these DEMs and DEGs. Moreover, immune-related pathways, including the cytokine-cytokine receptor interaction, apoptosis, Toll-like receptor, Jak-STAT, and RIG-I-like receptor signaling pathway, were significantly enriched through analyzing functions of mRNAs in the network in response to DHAV-3 infection. Furthermore, apl-miR-32-5p, apl-miR-125-5p, apl-miR-128-3p, apl-miR-460-5p, and novel-m0012-3p were identified as potential regulators in the immune-related signaling pathways during DHAV-3 infection. And some host miRNAs were predicted to target the DHAV-3 genome. CONCLUSIONS: This is the first integrated analysis of miRNA and mRNA in DHAV-3-infected ducklings. The results indicated the important roles of miRNAs in regulating immune response genes and revealed the immune related miRNA-mRNA regulation network in the DHAV-3-infected duckling liver. These findings increase our knowledge of the roles of miRNAs and their target genes in DHAV-3 replication and pathogenesis. They also aid in the understanding of host-virus interactions.


Ducks/genetics , Ducks/virology , Gene Regulatory Networks/genetics , Hepatitis Virus, Duck/genetics , Liver/virology , MicroRNAs/genetics , RNA, Messenger/genetics , Animals , Cytokines/genetics , Genome, Viral/genetics , Poultry Diseases/virology , Receptors, Cytokine/genetics , Signal Transduction/genetics
12.
Medicine (Baltimore) ; 98(24): e15945, 2019 Jun.
Article En | MEDLINE | ID: mdl-31192931

BACKGROUND: Fluoxetine has been reported to treat anorexia nervosa (AN) caused by chemotherapy in patients with cholangiocarcinoma effectively. However, no study systematically investigated its efficacy and safety. Thus, this study will systematically assess its efficacy and safety for AN caused by chemotherapy in patients with cholangiocarcinoma. METHODS: A comprehensive literature search for relevant studies will be conducted from the following databases from inception to the present: MEDILINE, EMBASE, Cochrane Library, Web of Science, PSYCINFO, Allied and Complementary Medicine Database, Chinese Biomedical Literature Database, and China National Knowledge Infrastructure. All randomized controlled trials on assessing the efficacy and safety of fluoxetine for AN caused by chemotherapy in patients with cholangiocarcinoma will be considered for inclusion in this study. RevMan V.5.3 software will be used for risk of bias assessment and statistical analysis. RESULTS: This study will summarize the latest evidence of fluoxetine for AN caused by chemotherapy in patients with cholangiocarcinoma through assessing outcomes of weight, depression, anxiety, and quality of life. Additionally, any adverse events will also be analyzed. CONCLUSION: The findings of this study will provide most recent evidence of fluoxetine for AN caused by chemotherapy in patients with cholangiocarcinoma. SYSTEMATIC REVIEW REGISTRATION: PROSPERO CRD42019131583.


Anorexia Nervosa/drug therapy , Antidepressive Agents, Second-Generation/therapeutic use , Antineoplastic Agents/adverse effects , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Fluoxetine/therapeutic use , Anorexia Nervosa/chemically induced , Bile Duct Neoplasms/psychology , China , Cholangiocarcinoma/psychology , Female , Humans , Male , Quality of Life , Randomized Controlled Trials as Topic , Research Design , Treatment Outcome
13.
Int Immunopharmacol ; 62: 114-119, 2018 Sep.
Article En | MEDLINE | ID: mdl-30005226

Thyroid cancer is one of the malignancies with better clinical outcomes. However, a minority of patients develops an aggressive anaplastic thyroid carcinoma. Development of innovative and multimodal therapeutic strategies is urgently needed. Here, we investigated the role of CXCR5+ CD8 T cells in the peripheral blood, tumor-involved lymph nodes (TILN), and tumor mass of thyroid cancer patients. In peripheral blood mononuclear cells, CXCR5+ cells represented 1.4% ±â€¯0.84% (mean ±â€¯s.d.) of total CD8 T cells, while in TILN and in tumor, the frequencies of CXCR5+ CD8 T cells were significantly higher at 27.7% ±â€¯7.8% and 15.5% ±â€¯2.9%, respectively. Compared to CXCR5- CD8 T cells, CXCR5+ CD8 T cells presented significantly higher PD-1 expression and lower or comparable TIM-3 and CTLA-4 expression. To compare and contrast the functional characteristics of CXCR5+ CD8 T cells and CXCR5- CD8 T cells, these cells were separated from TILNs and were TCR-stimulated via anti-CD3/CD28. Upon stimulation, CXCR5+ CD8 T cells presented stronger downregulation of CD27, higher expression of proinflammatory cytokines IL-2, IFN-γ, and TNF-α, and higher proliferation capacity than CXCR5- CD8 T cells. Moreover, CXCR5+ CD8 T cells presented higher expression of cytotoxic molecules Gzm-A, Gzm-B, and perforin. Overall, these results demonstrated that in thyroid cancer patients CXCR5+ CD8 T cells infiltrated the TILNs and the tumors, and were functionally more potent compared to their CXCR5- counterpart.


CD8-Positive T-Lymphocytes/immunology , Lymph Nodes/immunology , Lymphocyte Activation , Programmed Cell Death 1 Receptor/genetics , Receptors, CXCR5/immunology , Thyroid Cancer, Papillary/immunology , Adult , CTLA-4 Antigen/metabolism , Cells, Cultured , Female , Hepatitis A Virus Cellular Receptor 2 , Humans , Lymph Nodes/blood supply , Lymphatic Metastasis , Male , Middle Aged , Receptors, CXCR5/metabolism , Thyroid Cancer, Papillary/blood , Thyroid Cancer, Papillary/pathology , Transcription, Genetic/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
14.
Biomed Pharmacother ; 103: 167-173, 2018 Jul.
Article En | MEDLINE | ID: mdl-29653361

Over the past decades, lncRNAs have attracted more and more attentions of researchers. It has been verified that lncRNAs can modulate multiple biological behaviors in various human cancers. LncRNA ASAP1-IT1 has been certified to be a tumor facilitator in several malignant tumors. This study aims to investigate the effects of dysregulated ASAP1-IT1 on biological processes of Cholangiocarcinoma. The high expression level of ASAP1-IT1 was tested in Cholangiocarcinoma tissues and cells with qRT-PCR. Upregulation of ASAP1-IT predicted the unfavorable prognosis for Cholangiocarcinoma patients. Next, ASAP1-IT1 was knocked down in cancerous cells for loss-of function assay. MTT, colony formation and transwell and western bot assays were performed to demonstrate the specific impacts of ASAP1-IT1 on proliferation, migration and EMT progression of Cholangiocarcinoma. Cells. As a results, the Cholangiocarcinoma progression was inhibited. Hedgehog signaling pathway has been discovered to be a treatment target in Cholangiocarcinoma. In this study, the interaction between ASAP1-IT1 and hedgehog pathway was specifically investigated. Smo and Gli1, two hedgehog-related proteins were examined in Cholangiocarcinoma cells. The results of qRT-PCR and western blot assay suggested that ASAP1-IT1 could positively modulate Smo and Gli1 in Cholangiocarcinoma. Finally, rescue assays were carried out to prove that ASAP1-IT1 could improve Cholangiocarcinoma progression and development via hedgehog signaling pathway.


Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Hedgehog Proteins/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Knockdown Techniques , Gene Silencing , Humans , Male , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , RNA, Long Noncoding/genetics
15.
Tumour Biol ; 37(11): 14721-14731, 2016 Nov.
Article En | MEDLINE | ID: mdl-27629139

The melastatin-like transient receptor potential 7 (TRPM7) has been implicated in proliferation or apoptosis of some cancers, indicating the potential of TRPM7 as an anti-anaplastic target. Here, we identified the characteristic TRPM7 channel currents in human malignant glioma MGR2 cells, which could be blocked by a pharmacologic inhibitor Gd3+. We mined the clinical sample data from Oncomine Database and found that human malignant glioma tissues expressed higher TRPM7 mRNA than normal brain ones. Importantly, we identified a widely used clinical anesthetic midazolam as a TRPM7 inhibitor. Midazolam treatment for seconds suppressed the TRPM7 currents and calcium influx, and treatment for 48 h inhibited the TRPM7 expression. The inhibitory effect on TRPM7 accounts for the proliferation loss and G0/G1 phase cell cycle arrest induced by midazolam. Our data demonstrates that midazolam represses proliferation of human malignant glioma cells through inhibiting TRPM7 currents, which may be further potentiated by suppressing the expression of TRPM7. Our result indicates midazolam as a pharmacologic lead compound with brain-blood barrier permeability for targeting TRPM7 in the glioma.


Anti-Anxiety Agents/pharmacology , Calcium/metabolism , Cell Proliferation/drug effects , Glioma/drug therapy , Midazolam/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Data Mining , Databases, Factual , Fluorescent Antibody Technique , Glioma/metabolism , Glioma/pathology , Humans , Image Processing, Computer-Assisted/methods , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Tumor Cells, Cultured
16.
PLoS One ; 10(3): e0119912, 2015.
Article En | MEDLINE | ID: mdl-25799367

Transient receptor potential melastatin 7 (TRPM7), a non-selective cation channel, is highly expressed expressed in the brain and plays a critical role in ischemic neuronal death. Astrocyte, the most abundant cell type in central nervous system (CNS), exerts many essential functions in the physiological and pathological conditions. Here we investigated the expression and functions of the TRPM7 channel in mouse cortical astrocytes. Using reverse transcription (RT)-PCR, immunostaining, western blot and patch clamp recording, we showed that functional TRPM7 channel is expressed in cultured mouse cortical astrocytes. Knocking down TRPM7 with specific siRNA impairs the proliferation and migration of astrocytes by 40.2% ± 3.9% and 40.1% ± 11.5%, respectively. Consistently, inhibition of TRPM7 with 2-aminoethoxydiphenyl borate (2-APB) also decreases astrocyte proliferation and migration by 46.1% ± 2.5% and 64.2% ± 2.4%. MAPKs and Akt signaling pathways have been shown to be implicated in TRPM7-mediated responses including cell proliferation and migration. Our data show that suppression of TRPM7 in astrocytes reduces the phosphorylation of extracellular signal-regulated kinases (ERK) and c-Jun N-terminal kinases (JNK), but not p38 mitogen-activated protein kinase and Akt. In addition, TRPM7, as a cation channel, has been involved in the Ca²âº and Mg²âº homeostasis in several types of cells. In our study, we found that silencing TRPM7 decreases the intracellular basal Mg²âº concentration without affecting Ca²âº concentration in astrocytes. However, an addition of Mg²âº to the growth medium could not rescue the impaired proliferation of astrocytes. Together, our data suggest that TRPM7 channel may play a critical role in the proliferation and migration of astrocytes via the ERK and JNK pathways.


Astrocytes/cytology , Cell Movement , Cell Proliferation , Cerebral Cortex/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors , Animals , Apoptosis , Astrocytes/metabolism , Blotting, Western , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Silencing , HEK293 Cells , Humans , Immunoenzyme Techniques , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
17.
Am J Physiol Cell Physiol ; 308(4): C308-18, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-25472964

Transient receptor potential melastatin 7 (TRPM7) is a nonselective cation channel with an α-kinase domain in its COOH terminal, known to play a role in diverse physiological and pathological processes such as Mg2+ homeostasis, cell proliferation, and hypoxic neuronal injury. Increasing evidence suggests that TRPM7 contributes to the physiology/pathology of vascular systems. For example, we recently demonstrated that silencing TRPM7 promotes growth and proliferation and protects against hyperglycemia-induced injury in human umbilical vein endothelial cells (HUVECs). Here we investigated the potential effects of TRPM7 on morphology, adhesion, migration, and tube formation of vascular endothelial cells and the potential underlying mechanism. We showed that inhibition of TRPM7 function in HUVECs by silencing TRPM7 decreases the density of TRPM7-like current and cell surface area and inhibits cell adhesion to Matrigel. Silencing TRPM7 also promotes cell migration, wound healing, and tube formation. Further studies showed that the extracellular signal-regulated kinase (ERK) pathway is involved in the change of cell morphology and the increase in HUVEC migration induced by TRPM7 silencing. We also demonstrated that silencing TRPM7 enhances the phosphorylation of myosin light chain (MLC) in HUVECs, which might be involved in the enhancement of cell contractility and motility. Collectively, our data suggest that the TRPM7 channel negatively regulates the function of vascular endothelial cells. Further studies on the underlying mechanism may facilitate the development of the TRPM7 channel as a target for the therapeutic intervention of vascular diseases.


Cell Adhesion , Cell Movement , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , TRPM Cation Channels/metabolism , Cell Shape , HEK293 Cells , Humans , Membrane Potentials , Myosin Light Chains/metabolism , Phosphorylation , Protein Serine-Threonine Kinases , RNA Interference , Signal Transduction , TRPM Cation Channels/genetics , Transfection , Wound Healing
18.
CNS Neurosci Ther ; 21(1): 32-9, 2015 Jan.
Article En | MEDLINE | ID: mdl-25169754

BACKGROUND: Previous study demonstrated that overstimulation of TRPM7 substantially contributes to zinc-mediated neuronal toxicity. Inhibition of TRPM7 activity and TRPM7-mediated intracellular Zn(2+) accumulation may represent a promising strategy in the treatment of stroke. AIMS: To investigate whether local anesthetics lidocaine could inhibit TRPM7 channel and TRPM7-mediated zinc toxicity. METHODS: Whole-cell patch-clamp technique was used to investigate the effect of local anesthetics on TRPM7 currents in cultured mouse cortical neurons and TRPM7-overexpressed HEK293 cells. Fluorescent Zn(2+) imaging technique was used to study the effect of lidocaine on TRPM7-mediated intracellular Zn(2+) accumulation. TRPM7-mediated zinc toxicity in neurons was used to evaluate the neuroprotective effect of lidocaine. RESULTS: (1) Lidocaine dose dependently inhibits TRPM7-like currents, with an IC50 of 11.55 and 11.06 mM in cultured mouse cortical neurons and TRPM7-overexpressed HEK293 cells, respectively; (2) Lidocaine inhibits TRPM7 currents in a use/frequency-dependent manner; (3) Lidocaine inhibits TRPM7-mediated intracellular Zn(2+) accumulation in both cortical neurons and TRPM7-overexpressed HEK293 cells; (4) TRPM7-mediated Zn(2+) toxicity is ameliorated by lidocaine in cortical neurons; (5) QX-314 has a similar inhibitory effect as lidocaine on TRPM7 currents when applied extracellularly; (6) Procaine also shows potent inhibitory effect on the TRPM7 currents in cortical neurons. CONCLUSION: Our data provide the first evidence that local anesthetic lidocaine inhibits TRPM7 channel and TRPM7-mediated zinc toxicity.


Lidocaine/pharmacology , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors , Zinc/toxicity , Anesthetics, Local/chemistry , Anesthetics, Local/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , HEK293 Cells , Humans , Intracellular Space/metabolism , Ions/metabolism , Ions/toxicity , Lidocaine/analogs & derivatives , Lidocaine/chemistry , Membrane Potentials/drug effects , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/chemistry , Patch-Clamp Techniques , Procaine/chemistry , Procaine/pharmacology , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , Zinc/metabolism
19.
CNS Neurosci Ther ; 21(3): 252-61, 2015 Mar.
Article En | MEDLINE | ID: mdl-25438992

BACKGROUND: Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor with a dismal prognosis. Despite intensive study on tumor biology, the underlying mechanisms of the unlimited proliferation and progressive local invasion are still poorly understood, and no effective treatment has been developed for GBM patients. AIMS: We determine the role of TRPM7 channels in the growth, migration, and infiltration of malignant glioma cells. METHODS: Using a combination of RT-PCR, Western blot, and patch-clamp techniques, we demonstrated the expression of functional TRPM7 channels of A172 cells, a human glioma cell line, as well as in human glioma tissues. Furthermore, we evaluated the role of TRPM7 in growth, migration, and infiltration of A172 cells with MTT and transwell migration and invasion assays. RESULTS: We showed the expression of functional TRPM7 channels in both A172 cells and human glioma tissues. Suppression of TRPM7 expression with TRPM7-siRNA dramatically reduced the proliferation, migration, and invasion of A172 cells. Pharmacological inhibition of TRPM7 channel with 2-aminoethoxydiphenyl borate (2-APB) showed a similar effect as TRPM7-siRNA. CONCLUSION: We demonstrate that human glioma cells express functional TRPM7 channel and that activation of this channel plays an important role in the proliferation, migration, and invasion of malignant glioma cells. TRPM7 channel may represent a novel and promising target for therapeutic intervention of malignant glioma.


Brain Neoplasms/physiopathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioblastoma/physiopathology , Neoplasm Invasiveness/physiopathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors , Blotting, Western , Boron Compounds/pharmacology , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cells, Cultured , Glioblastoma/drug therapy , Humans , Patch-Clamp Techniques , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
20.
PLoS One ; 8(11): e79540, 2013.
Article En | MEDLINE | ID: mdl-24223965

This study investigated the change of transient receptor potential melastatin 7 (TRPM7) expression by high glucose and its role in hyperglycemia induced injury of vascular endothelial cells. Human umbilical vein endothelial cells (HUVECs) were incubated in the presence or absence of high concentrations of D-glucose (HG) for 72 h. RT-PCR, Real-time PCR, Western blotting, Immunofluorescence staining and whole-cell patch-clamp recordings showed that TRPM7 mRNA, TRPM7 protein expression and TRPM7-like currents were increased in HUVECs following exposure to HG. In contrast to D-glucose, exposure of HUVECs to high concentrations of L-glucose had no effect. HG increased reactive oxygen species (ROS) generation, cytotoxicity and decreased endothelial nitric oxide synthase protein expression, which could be attenuated by knockdown of TRPM7 with TRPM7 siRNA. The protective effect of silencing TRPM7 against HG induced endothelial injury was abolished by U0126, an inhibitor of the extracellular signal-regulated kinase signaling pathway. These observations suggest that TRPM7 channels play an important role in hyperglycemia-induced injury of vascular endothelial cells.


Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Hyperglycemia/metabolism , Hyperglycemia/pathology , TRPM Cation Channels/metabolism , Butadienes/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Silencing , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Hyperglycemia/genetics , MAP Kinase Signaling System/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitriles/pharmacology , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics
...