Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Pharmaceuticals (Basel) ; 17(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38675401

ABSTRACT

Exposure to hypoxia results in the development of pulmonary arterial hypertension (PAH). An increase in the intracellular Ca2+ concentration ([Ca2+]i) in pulmonary artery smooth muscle cells (PASMCs) is a major trigger for pulmonary vasoconstriction and proliferation. This study investigated the mechanism by which KMUP-1, a xanthine derivative with phosphodiesterase inhibitory activity, inhibits hypoxia-induced canonical transient receptor potential channel 1 (TRPC1) protein overexpression and regulates [Ca2+]i through store-operated calcium channels (SOCs). Ex vivo PASMCs were cultured from Sprague-Dawley rats in a modular incubator chamber under 1% O2/5% CO2 for 24 h to elucidate TRPC1 overexpression and observe the Ca2+ release and entry. KMUP-1 (1 µM) inhibited hypoxia-induced TRPC family protein encoded for SOC overexpression, particularly TRPC1. KMUP-1 inhibition of TRPC1 protein was restored by the protein kinase G (PKG) inhibitor KT5823 (1 µM) and the protein kinase A (PKA) inhibitor KT5720 (1 µM). KMUP-1 attenuated protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA, 1 µM)-upregulated TRPC1. We suggest that the effects of KMUP-1 on TRPC1 might involve activating the cyclic guanosine monophosphate (cGMP)/PKG and cyclic adenosine monophosphate (cAMP)/PKA pathways and inhibiting the PKC pathway. We also used Fura 2-acetoxymethyl ester (Fura 2-AM, 5 µM) to measure the stored calcium release from the sarcoplasmic reticulum (SR) and calcium entry through SOCs in hypoxic PASMCs under treatment with thapsigargin (1 µM) and nifedipine (5 µM). In hypoxic conditions, store-operated calcium entry (SOCE) activity was enhanced in PASMCs, and KMUP-1 diminished this activity. In conclusion, KMUP-1 inhibited the expression of TRPC1 protein and the activity of SOC-mediated Ca2+ entry upon SR Ca2+ depletion in hypoxic PASMCs.

2.
Int J Biol Macromol ; 265(Pt 1): 130855, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490377

ABSTRACT

Transient receptor potential canonical (TRPC) channels allow the intracellular entry of Ca2+ and play important roles in several physio-pathological processes. In this study, we constructed transgenic mice expressing porcine TRPC1 (Tg-pTRPC1) to verify the effects of TRPC1 on skeletal muscle growth and elucidate the underlying mechanism. Porcine TRPC1 increased the muscle mass, fiber cross-sectional area, and exercise endurance of mice and accelerated muscle repair and regeneration. TRPC1 overexpression enhanced ß-catenin expression and promoted myogenesis, which was partly reversed by inhibitors of ß-catenin. TRPC1 facilitated the accumulation of intracellular Ca2+ and nuclear translocation of the NFATC2/NFATC2IP complex involved in the Wnt/Ca2+ pathway, promoting muscle growth. Paired related homeobox 1 (Prrx1) promoted the expression of TRPC1, NFATC2, and NFATC2IP that participate in the regulation of muscle growth. Taken together, our findings indicate that porcine TRPC1 promoted by Prrx1 could regulate muscle development through activating the canonical Wnt/ß-catenin and non-canonical Wnt/Ca2+ pathways.


Subject(s)
Transient Receptor Potential Channels , beta Catenin , Mice , Animals , Swine , beta Catenin/genetics , beta Catenin/metabolism , Muscle, Skeletal/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Transient Receptor Potential Channels/metabolism , Calcium/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
3.
Brain Res Bull ; 208: 110894, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38325758

ABSTRACT

Neutrophil infiltration has been linked to worse clinical outcomes after ischemic stroke. Microglia, a key type of immune-competent cell, engage in cross-talk with the infiltrating immune cells in the inflamed brain area, yet the molecular mechanisms involved remain largely unexplored. In this study, we investigated the mechanisms of how canonical transient receptor potential 1 (TRPC1) modulated neutrophil infiltration in male mouse cerebral ischemia and reperfusion injury (CIRI) models. Our findings revealed a notable upregulation of TRPC1 in microglia within both middle cerebral artery occlusion reperfusion (MCAO/R) and in vitro oxygen-glucose deprivation/regeneration (OGD/R) model. Conditional Trpc1 knockdown in microglia markedly reduced infarct volumes and alleviated neurological deficits. Microglia conditional Trpc1 knockdown mice displayed less neutrophil infiltration in peri-infarct area. Trpc1 knockdown microglia exhibited a reduced primed proinflammatory phenotype with less secretion of CC-Chemokines ligand (CCL) 5 and CCL2 after MCAO/R. Blocking CCL5/2 significantly mitigated neutrophil infiltration in microglia/neutrophil transwell co-culture system upon OGD/R condition. Trpc1 knockdown markedly reduced store-operated calcium entry and nuclear factor of activated T-cells c1 (NFATc1) level in OGD/R treated microglia. Overexpression of Nfatc1 reversed the CCL5/2 reducing effect of Trpc1 knockdown, which is mediated by small interfering RNA in BV2 cells upon OGD/R. Our data indicate that upregulation of TRPC1 in microglia stimulates the production of CCL5/2 through the Ca2+/NFATc1 pathway. Upregulated CCL5/2 leads to an increase in neutrophil infiltration into the brain, thereby aggravating reperfusion injury. Our results demonstrate the importance of TRPC1 in microglia-mediated neuroinflammation and suggest a potential means for reducing CIRI induced neurological injury.


Subject(s)
Brain Ischemia , Ischemic Stroke , Reperfusion Injury , Stroke , Male , Mice , Animals , Up-Regulation , Ischemic Stroke/metabolism , Microglia/metabolism , Neutrophil Infiltration , Brain Ischemia/metabolism , Infarction, Middle Cerebral Artery/metabolism , Reperfusion Injury/metabolism , Stroke/metabolism
4.
J Clin Lab Anal ; 36(8): e24560, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35754147

ABSTRACT

BACKGROUND: Transient receptor potential channel 1 (TRPC1) promotes tumor growth and metastasis in endometrial carcinoma (EC) cell lines, whereas its clinical role in EC management remains unclear. Therefore, this study aimed to investigate the association of TRPC1 protein expression with the clinical features and survival of EC patients, then was further validated by TRPC1 mRNA measurement and data from The Human Protein Atlas. METHODS: TRPC1 protein expression in tumor tissues and normal endometria of 176 resectable EC patients was determined using immunohistochemistry. Besides, TRPC1 mRNA expression of partial patients (n = 80) was detected using RT-qPCR. Additionally, survival data from The Human Protein Atlas (derived from The Cancer Genome Atlas [TCGA]) was analyzed. RESULTS: TRPC1 protein expression was up-regulated in tumor tissue compared with normal endometrium (p < 0.001). Up-regulated TRPC1 protein expression was associated with stromal cervical invasion (p = 0.044), lymphovascular invasion (p = 0.032), and increased federation of gynecology and obstetrics (FIGO) stage (p = 0.005). Tumor TRPC1 protein high was linked with shortened accumulating disease-free survival (DFS) (p = 0.009) and overall survival (OS) (p = 0.026), which were also confirmed by multivariate Cox's regression analysis (both p < 0.050). Further, TRPC1 mRNA validation disclosed that TRPC1 mRNA high was related to shortened accumulating DFS (p = 0.038) and exhibited a correlating trend with declined OS (lacked statistical significance) (p = 0.162). Meanwhile, survival analysis on the data from The Human Protein Atlas (derived from TCGA) also exhibited that TRPC1 mRNA high was correlated with reduced accumulating OS (p < 0.001). CONCLUSION: Our findings support TRPC1 as a prognostic biomarker in resectable EC patients.


Subject(s)
Carcinoma , Endometrial Neoplasms , Carcinoma/pathology , Disease-Free Survival , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/surgery , Endometrium/metabolism , Female , Humans , Lymphatic Metastasis , Neoplasm Staging , Prognosis , RNA, Messenger/genetics
5.
Front Surg ; 9: 853310, 2022.
Article in English | MEDLINE | ID: mdl-35548183

ABSTRACT

Background: Transient receptor potential channel 1 (TRPC1) regulates the progression of several cancers, but its clinical implication in renal cell carcinoma (RCC) has not been explored yet. This study aimed to investigate the correlation of TRPC1 with clinical characteristics and prognosis in patients with RCC. Methods: Totally, 177 patients with primary RCC who received surgical resection were retrospectively screened. Their tumor and paired adjacent tissue specimens were retrieved to assess TRPC1 mRNA expression using RT-qPCR and TRPC1 protein expression using immunohistochemistry (IHC). Results: Both TRPC1 IHC score and TRPC1 mRNA expression were elevated in RCC tissue than in adjacent tissue (both P < 0.001). Meanwhile, both TRPC1 IHC score and TRPC1 mRNA expression in tumor were associated with higher T stage (both P = 0.02) and TNM stage (P = 0.009, P = 0.003, respectively). However, no correlation was found in tumor TRPC1 IHC score or TRPC1 mRNA expression with other tumor properties (all P > 0.05). Besides, the 3-, 5-, and 7-year overall survival (OS) were 81.4, 68.6, and 60.2%, respectively in patients with high tumor TRPC1 protein, while they were 89.3, 82.7, and 76.7%, respectively in patients with low tumor TRPC1 protein. High (vs. low) TRPC1 protein in the tumor was associated with shorter OS (P = 0.017), while high (vs. low) TRPC1 mRNA in the tumor was not correlated with OS (P = 0.144). By the forward stepwise method, TRPC1 protein expression independently predicted poor OS (P = 0.01, hazard ratio = 2.052). Conclusion: TRPC1 serves as a potential biomarker reflecting tumor features and long-term survival profile in patients with RCC.

6.
J Clin Lab Anal ; 36(3): e24229, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35106847

ABSTRACT

BACKGROUND: Transient receptor potential channel 1 (TRPC1) facilitates the tumor growth, metastasis, and chemoresistance in a series of neoplasms, while its correlation with clinical features and survival profile in NSCLC patients remains elusive. Hence, this study aimed to explore this topic. METHODS: Totally, 192 NSCLC patients were enrolled. Protein and mRNA expression of TRPC1 in carcinoma tissue and para-carcinoma tissue were evaluated by immunohistochemistry (IHC) assay and reverse transcription quantitative polymerase chain reaction (RT-qPCR) assay, respectively. RESULTS: Immunohistochemistry score and mRNA expression of TRPC1 were higher in carcinoma tissue compared with para-carcinoma tissue (both p < 0.001). Besides, increased TRPC1 IHC score (p = 0.004) and elevated TRPC1 mRNA overexpression (p = 0.016) were linked with occurrence of LYN metastasis; meanwhile, increased TRPC1 IHC score (p = 0.015) and raised TRPC1 mRNA expression (p = 0.009) were also linked with advanced TNM stage, whereas TRPC1 IHC score and TRPC1 mRNA expression were not correlated with other clinical features (all p > 0.05). Additionally, TRPC1 protein high (p = 0.007) and TRPC1 mRNA high (p = 0.015) were correlated with poor disease-free survival (DFS) but not correlated with overall survival (OS). Moreover, multivariate Cox's proportional hazards regression analysis showed that high TRPC1 protein expression (p = 0.046) and advanced TNM stage (p < 0.001) were independently correlated with poor DFS. However, TRPC1 protein and mRNA expression were not linked with OS (both p > 0.05), while poor differentiation (p = 0.003) and advanced TNM stage (p < 0.001) were independently associated with worse OS. CONCLUSIONS: TRPC1 is unregulated in NSCLC tissue with its overexpression relating to the occurrence of LYN metastasis and worse DFS in NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Transient Receptor Potential Channels , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/surgery , Disease-Free Survival , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/surgery , Lymphatic Metastasis , Neoplasm Staging , Prognosis
7.
Eur J Neurosci ; 54(3): 4827-4837, 2021 08.
Article in English | MEDLINE | ID: mdl-34132424

ABSTRACT

Disturbance in calcium (Ca2+ ) homeostasis has been involved in a variety of neuropathological conditions including Parkinson's disease (PD). The Ca2+ channel, transient receptor potential channel 1 (TRPC1), plays a protective role in regulating entry of Ca2+ activated by store depletion of Ca2+ in endoplasmic reticulum (ER). We have showed that thioredoxin-1 (Trx-1) plays a role in suppressing ER stress in PD. However, whether Trx-1 regulates TRPC1 expression in PD is still unknown. In the present study, we demonstrated that treatment of 1-methyl-4-phenylpyridinum ion (MPP+ ) significantly reduced the expression of TRPC1 in PC12 cells, which was restored by Trx-1 overexpression, and further decreased significantly by Trx-1 siRNA. Moreover, we found that Ca2+ entered into the cells was decreased by MPP+ in PC 12 cells, which was restored by Trx-1 overexpression, and further decreased by Trx-1 siRNA. MPP+ significantly increased calcium-dependent cysteine protease calpain1 expression in PC12 cells, which was suppressed by Trx-1 overexpression. Calpain1 expression was increased by Trx-1 siRNA or SKF96365, an inhibitor of TRPC1. Moreover, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) decreased TRPC1 expression in the substantia nigra pars compacta region (SNpc), which was restored in mice overexpressing Trx-1, and further decreased in mice of knockdown Trx-1. Inversely, the expression of calpain1 was increased by MPTP, which was suppressed in mice overexpressing Trx-1, and further increased in mice of knockdown Trx-1. In conclusion, Trx-1 regulates the Ca2+ entry through regulating TRPC1 expression after treatment of MPP+ /MPTP.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Calcium , Disease Models, Animal , Homeostasis , Mice , Mice, Inbred C57BL , PC12 Cells , Rats , Thioredoxins/genetics
8.
World J Stem Cells ; 13(4): 260-280, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33959218

ABSTRACT

Stem cells hold indefinite self-renewable capability that can be differentiated into all desired cell types. Based on their plasticity potential, they are divided into totipotent (morula stage cells), pluripotent (embryonic stem cells), multipotent (hematopoietic stem cells, multipotent adult progenitor stem cells, and mesenchymal stem cells [MSCs]), and unipotent (progenitor cells that differentiate into a single lineage) cells. Though bone marrow is the primary source of multipotent stem cells in adults, other tissues such as adipose tissues, placenta, amniotic fluid, umbilical cord blood, periodontal ligament, and dental pulp also harbor stem cells that can be used for regenerative therapy. In addition, induced pluripotent stem cells also exhibit fundamental properties of self-renewal and differentiation into specialized cells, and thus could be another source for regenerative medicine. Several diseases including neurodegenerative diseases, cardiovascular diseases, autoimmune diseases, virus infection (also coronavirus disease 2019) have limited success with conventional medicine, and stem cell transplantation is assumed to be the best therapy to treat these disorders. Importantly, MSCs, are by far the best for regenerative medicine due to their limited immune modulation and adequate tissue repair. Moreover, MSCs have the potential to migrate towards the damaged area, which is regulated by various factors and signaling processes. Recent studies have shown that extracellular calcium (Ca2+) promotes the proliferation of MSCs, and thus can assist in transplantation therapy. Ca2+ signaling is a highly adaptable intracellular signal that contains several components such as cell-surface receptors, Ca2+ channels/pumps/exchangers, Ca2+ buffers, and Ca2+ sensors, which together are essential for the appropriate functioning of stem cells and thus modulate their proliferative and regenerative capacity, which will be discussed in this review.

9.
Curr Med Sci ; 40(6): 1085-1091, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33428136

ABSTRACT

Cigarette smoking contributes to the development of pulmonary artery hypertension (PAH). As the basic pathological change of PAH, pulmonary vascular remodeling is considered to be related to the abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs). However, the molecular mechanism underlying this process remains not exactly clear. The aim of this research was to study the molecular mechanism of PASMCs proliferation induced by smoking. Human PASMCs (HPASMCs) were divided into 6 groups: 0% (control group), cigarette smoking extract (CSE)-treated groups at concentrations of 0.5%, 1%, 2%, 5%, 10% CSE respectively. HPASMCs proliferation was observed after 24 h. HPASMCs were divided into two groups: 0 (control group), 0.5% CSE group. The mRNA and protein expression levels of transient receptor potential channel 1 (TRPC1) and cyclin D1 in HPASMCs after CSE treatment were respectively detected by RT-PCR and Western blotting. The intracellular calcium ion concentration was measured by the calcium probe in each group. In the negative control group and TRPC1-siRNA transfection group, the proliferation of HPASMCs and the expression of cyclin D1 mRNA and protein were detected. Data were compared with one-way ANOVA (for multiple-group comparison) and independent t-test (for two-group comparison) followed by the least significant difference (LSD) test with the computer software SPSS 17.0. It was found that 0.5% and 1% CSE could promote the proliferation of HPASMCs (P<0.05), and the former was more effective than the latter (P<0.05), while 3% and above CSE had inhibitory effect on HPASMCs (P<0.05). The mRNA and protein expression levels of TRPC1 and cyclin D1 in 0.5% and 1% CSE groups were significantly higher than those in the control group (P<0.05), while those in 3% CSE group were significantly decreased (P<0.05). Moreover, the proliferation of HPASMCs and the expression of cyclin D1 mRNA and protein in TRPC1-siRNA transfection group were significantly reduced as compared with those in the negative control group (P<0.05). It was concluded that low concentration of CSE can promote the proliferation of HPASMCs, while high concentrations of CSE inhibit HPASMCs proliferation. These findings suggested that CSE induced proliferation of HPASMCs at least in part via TRPC1-mediated cyclin D1 expression.


Subject(s)
Cyclin D1/metabolism , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/cytology , Smoke/adverse effects , TRPC Cation Channels/metabolism , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , TRPC Cation Channels/genetics , Nicotiana , Up-Regulation
10.
Theranostics ; 9(19): 5517-5531, 2019.
Article in English | MEDLINE | ID: mdl-31534500

ABSTRACT

Pulsed focused ultrasound (pFUS) technology is being developed for clinical neuro/immune modulation and regenerative medicine. Biological signal transduction of pFUS forces can require mechanosensitive or voltage-gated plasma membrane ion channels. Previous studies suggested pFUS is capable of activating either channel type, but their mechanistic relationship remains ambiguous. We demonstrated pFUS bioeffects increased mesenchymal stem cell tropism (MSC) by altering molecular microenvironments through cyclooxygenase-2 (COX2)-dependent pathways. This study explored specific relationships between mechanosensitive and voltage-gated Ca2+ channels (VGCC) to initiate pFUS bioeffects that increase stem cell tropism. Methods: Murine kidneys and hamstring were given pFUS (1.15 or 1.125 MHz; 4MPa peak rarefactional pressure) under ultrasound or magnetic resonance imaging guidance. Cavitation and tissue displacement were measure by hydrophone and ultrasound radiofrequency data, respectively. Elastic modeling was performed from displacement measurements. COX2 expression and MSC tropism were evaluated in the presence of pharmacological ion channel inhibitors or in transient-receptor-potential-channel-1 (TRPC1)-deficient mice. Immunohistochemistry and co-immunoprecipitation examined physical channel relationships. Fluorescent ionophore imaging of cultured C2C12 muscle cells or TCMK1 kidney cells probed physiological interactions. Results: pFUS induced tissue deformations resulting in kPa-scale forces suggesting mechanical activation of pFUS-induced bioeffects. Inhibiting VGCC or TRPC1 in vivo blocked pFUS-induced COX2 upregulation and MSC tropism to kidneys and muscle. A TRPC1/VGCC complex was observed in plasma membranes. VGCC or TRPC1 suppression blocked pFUS-induced Ca2+ transients in TCMK1 and C2C12 cells. Additionally, Ca2+ transients were blocked by reducing transmembrane Na+ potentials and observed Na+ transients were diminished by genetic TRPC1 suppression. Conclusion: This study suggests that pFUS acoustic radiation forces mechanically activate a Na+-containing TRPC1 current upstream of VGCC rather than directly opening VGCC. The electrogenic function of TRPC1 provides potential mechanistic insight into other pFUS techniques for physiological modulation and optimization strategies for clinical implementation.


Subject(s)
Calcium Channels/metabolism , Kidney/metabolism , Muscle, Skeletal/metabolism , TRPC Cation Channels/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Cyclooxygenase 2/metabolism , Female , Kidney/diagnostic imaging , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Muscle, Skeletal/diagnostic imaging , Sodium/metabolism , TRPC Cation Channels/genetics , Ultrasonic Waves , Ultrasonography
11.
Acta Pharmacol Sin ; 40(4): 477-485, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30038339

ABSTRACT

Pulmonary arterial hypertension (PAH) is the most common form of pulmonary hypertension. Pulmonary arterial remodeling is closely related to the abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs), which leads to the thickening of the medial layer of muscular arteries and then results in the narrowing or occlusion of the precapillary arterioles and PAH. However, the mechanisms underlying the abnormal proliferation of PASMCs remain unclear. In this study, we established rat primary PAH models using monocrotaline (MCT) injection or hypoxic exposure, then investigated the expression patterns of seven miRNAs associated with multiple pathogenic pathways central to pulmonary hypertension, and further explored the roles and the possible mechanisms of miR-135a during the development of PAH. In the rat primary PAH models, we observed that the expression of miR-135a-5p in lungs was drastically decreased at the initial stage of PAH development after MCT administration or hypoxic exposure, but it increased by 12-fold or 10-fold at the later stage. In vitro study in PASMCs showed a similar pattern of miR-135a-5p expression, with downregulation at 6 h but upregulation at 18, 24, and 48 h after hypoxic exposure. Early, but not late, administration of a miR-135a-5p mimic inhibited hypoxia-induced proliferation of PASMCs. The protective role of early miR-135a-5p agomir in the PAH rat model further supported the hypothesis that the early decrease in the expression of miR-135a-5p contributes to the proliferation of PASMCs and development of PAH, as early administration of miR-135a-5p agomir (10 nM, i.v.) reversed the elevated mean pulmonary arterial pressure and pulmonary vascular remodeling in MCT-treated rats. We revealed that miR-135a-5p directly bound to the 3'-UTR sequence of rat transient receptor potential channel 1 (TRPC1) mRNA and decreased TRPC1 protein expression, thus inhibiting PASMC proliferation. Collectively, our data suggest that dysregulation of miR-135a-5p in PASMCs contributes to the abnormal proliferation of PASMCs and the pathogenesis of PAH. Increasing miR-135a-5p expression at the early stage of PAH is a potential new avenue to prevent PAH development.


Subject(s)
Hypertension, Pulmonary/metabolism , MicroRNAs/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Hypertension, Pulmonary/chemically induced , Hypoxia , Male , MicroRNAs/genetics , Monocrotaline , Rats , Rats, Sprague-Dawley
12.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-806965

ABSTRACT

Objective@#To understand the changes of serum transient receptor potential channel 1(TRPC1) in patients with vascular depression.@*Methods@#58 patients with vascular depression, 49 patients with major depressive disorder and 38 healthy controls were recruited.The TRPC1 of all subjects were detected by ELISA.Patients with vascular depression and patients with primary depression were scaled by HAMD-17.The level of TRPC1 was contrasted in different ages groups and in different nosogenesis (cerebral infarction, ischemic cerebrovascular disease, old cerebral hemorrhage, cerebral microbleeds, vascular risk factors, etc.) of vascular depression.The relationship between TRPC1 level and severity of depressive symptoms was further analyzed.@*Results@#(1)The level of TRPC1 of serum((643.76±118.43)pg/ml) was decreased in patients with vascular depression compared with that in healthy controls ((712.48± 98.75) pg/ml). The level of TRPC1 in patients with vascular depression over 60 years ((601.43±113.55)pg/ml)was lower than that in patients with major depressive disorder over 60 years ((626.32±125.46)pg/ml) and healthy controls over 60 years((721.84± 99.62)pg/ml) .(2) Among the various causes of vascular depression, the level of TRPC1 in patients with cerebral infarction, ischemic cerebrovascular disease and cerebral microbleeds was significantly lower (P<0.05). (3) The levels of TRPC1 in the patients with vascular depression (r=-0.962, P<0.05) and patients with major depressive disorder (r=-0.674, P<0.05) were negatively correlated with HAMD-17 score.@*Conclusion@#The level of TRPC1 is lower in patients with vascular depression, which is more obvious in patients with cerebral infarction, ischemic cerebrovascular disease, and cerebral microhaemorrhage.The lower the level of TRPC1, the more severe the depression.The neuroprotective effect of TRPC1 is reduced in patients with vascular depression.The TRPC1 can be used as a biomarker for vascular depression.

13.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-614675

ABSTRACT

Objective To investigate the effect of thymopentin combined with routine medication on serum interleukin 17A(IL-17A),chemokine 12 (CXCL12) levels and transient receptor potential channel 1 (TRPC1)expression in elderly patients with chronic obstructive pulmonary disease (COPD).Methods 156 COPD patients were selected as study objects.They were randomly divided into two groups(n =78) according to the digital table.The control group received routine treatment,and the observation group received subcutaneous injection of thymopentin on the basis of routine treatment.Before and after treatment,the CD3+,CD4+,CD8+,CD4+/CD8+ levels were tested to compare the immunologic function of the two groups.Before and after treatment,the FEV1,FEV1/FVC,PEF were tested to compare the pulmonary ventilation function of the two groups.The IL-17A,CXCL12 levels in peripheral blood and TRPC1 level in bronchoalveolar liquid before and after treatment were tested and compared between the two groups.The adverse effect was compared.Results Before treatment,the CD3+,CD4+,CD8+ and CD4+/CD8+ between the two groups had not statistically significant differences (all P > 0.05),which of the control group after treatment had no significant change (all P > 0.05),the CD3+,CD4+,CD4+/CD8+ of the observation group [(65.17 ± 2.39) %,(41.06 ±2.15) %,(1.50 ± 0.74) %] were significantly higher than control group [(52.66 ± 2.38) %,(32.30 ± 2.05) %,(0.80 ± 0.81) %] (all P < 0.05),and the CDs+ of the observation group [(24.02 ± 2.23) %] was significantly lower than control group[(32.66 ± 1.97) %,P <0.05].Before treatment,the FEV1,FEV1/FVC,PEF,the IL-17A,CXCL12 levels and TRPC1 level had no statistically significant differences between the two groups(P > 0.05).After treatment,the peripheral blood IL-17A [(24.18 ± 3.69) pg/mL],CXCL12 levels [(193.50 ± 2.90) pg/mL] and TRPC1 level in BALF [(7.69 ± 1.14)ng/L] in the observation group decreased significantly(P < 0.05),which in the control group also decreased significantly [IL-17A (34.25 ± 3.74) pg/mL,CXCL12 (205.37 ± 3.21) pg/mL,TRPC1 (14.25 ± 1.20)ng/L] (P < 0.05),which of the observation group were significantly lower than those of the control group(all P <0.05).The incidence rates of adverse effects of the two groups were t6.67%,12.82%,there was no statistically significant difference(P > 0.05).Conclusion Thymopentin combined with conventional therapy can effectively improve the elderly COPD patients'immunologic function and pulmonary ventilation function,decrease the inflammatory factor,relieve inflammatory reaction and it is safe and effective.

14.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-486795

ABSTRACT

AIM:To study the effect of transient receptor potential channel 1 ( TRPC1) on the survival of hip-pocampal neurons in mice.METHODS:TRPC1 knockout mice and the control mice (6 months old) were used in this study.Immunofluorescence staining of neuron-specific marker NeuN, Nissl staining and TUNEL staining were performed to measure the changes of the neurons in hippocampal CA1, CA3 and dentate gyrus (DG).Western blot analysis was used to detect the levels of pro-apoptotic protein C/EBP homologous protein ( CHOP) and cleaved caspase-3.RESULTS:Immuno-fluorescence staining and Nissl staining showed that the number of neuronal cells was significantly decreased in hippocampal CA1, CA3 and DG of TRPC1 knockout mice compared with the control mice.TUNEL staining showed that the apoptosis neuronal cell number of the above areas in TRPC1 knockout mice was significantly increased compared with the control mice.The results of Western blot revealed that the levels of CHOP and cleaved caspase-3 were significantly increased in the hippocampus of TRPC1 knockout mice relative to the control mice.CONCLUSION:The depletion of TRPC1 induces neu-ronal loss through a mechanism of TRPC1-mediated apoptosis.

15.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-486661

ABSTRACT

AIM:To investigate the effects of vesicular transport inhibition on the proliferation and regulation of store-operated calcium entry ( SOCE) in rat endothelial progenitor cells ( EPCs) .METHODS:EPCs were isolated from the rats with density-gradient centrifugation and confirmed via double fluorescence staining with acLDL-DiI and FITC-UEA-I.After inhibition of vesicular transport with brefeldin A ( BFA) , the proliferation of EPCs was measured by CCK-8 assay and real-time cell analyzer instrument, apoptosis was analyzed by flow cytometry, and the expression of ADP-ribosylation factor GTPase-activating protein 1 (ARFGAP1), a key protein to vesicular transport, was also detected.SOCE was ob-served under laser scanning confocal microscope after the vesicular transport was inhibited, and the protein expression of SOCC complex was determined by Western blot.Furthermore, the influences of vesicular transport inhibition on the expres-sion of transient receptor potential channel 1 ( TRPC1 ) and SOCE were examined with a RNA interference method.RE-SULTS:The acLDL-DiI and FITC-UEA-I double positive rate of the cells was 82.53%±6.12%.BFA insult significantly inhibited the proliferation of EPCs and down-regulated the expression of ARFGAP1, and no influence on the apoptosis of the EPCs was observed, suggesting that vesicular transport of EPCs was inhibited.Vesicular transport inhibition remarkably down-regulated the expression of TRPC1 and decreased SOCE level.No evident difference in the level of SOCE between siTRPC1 group and siTRPC1+BFA group, in which the cells were pretreated with siTRPC1 before BFA addition, was ob-served.CONCLUSION:Vesicular transport inhibition in EPCs reduces the proliferation of EPCs and decreases SOCE lev-el through down-regulation of TRPC1.

16.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-474075

ABSTRACT

[ ABSTRACT] AIM:To investigate the role of canonical transient receptor potential channel 1 ( TRPC1 ) in the epithelial-mesenchymal transition ( EMT) of human bronchial epithelial ( HBE) cells induced by transforming growth fac-tor-β1 (TGF-β1).METHODS:EMT of 16HBE cells induced by TGF-β1 were identified by microscopy, immunofluores-cence and Western blotting.Immunofluorescence, real-time PCR and Western blotting were applied to detect the mRNA and the protein expression of TRPC1 in the 16HBE cells.The influence of SKF96365 (a TRPC1 blocker) and siRNA-me-diated silencing of TRPC1 on the EMT of the 16HBE cells were detected by microscopy and Western blotting.RESULTS:Treatment with TGF-β1 induced significant morphological changes of the 16HBE cells.Exposure to TGF-β1 decreased the expression of E-cadherin protein (P<0.01) and increased the expression of α-SMA protein (P<0.05) in the 16HBE cells.Immunofluorescence observation indicated that TRPC1 expression in the 16HBE cells was positive.The expression of TRPC1 at mRNA and protein levels was significantly increased in the 16HBE cells after stimulation with TGF-β1 ( P<0.05).The morphological changes of the 16HBE cells induced by TGF-β1 were inhibited by SKF96365 and TRPC1 silen-cing compared with TGF-β1 group.The protein expression of E-cadherin andα-SMA induced by TGF-β1 were inhibited by SKF96365 and TRPC1 silencing compared with TGF-β1 group (P<0.05).CONCLUSION:TGF-β1 induces EMT with the mechanism of up-regulating TRPC1 in human bronchial epithelial cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...