Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
1.
Behav Brain Res ; 467: 115005, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38641178

ABSTRACT

BACKGROUND: Post-traumatic stress disorder (PTSD) refers to a chronic impairing psychiatric disorder occurring after exposure to the severe traumatic event. Studies have demonstrated that medicinal cannabis oil plays an important role in neuroprotection, but the mechanism by which it exerts anti-PTSD effects remains unclear. METHODS: The chronic complex stress (CCS) simulating the conditions of long voyage stress for 4 weeks was used to establish the PTSD mice model. After that, behavioral tests were used to evaluate PTSD-like behaviors in mice. Mouse brain tissue index was detected and hematoxylin-eosin staining was used to assess pathological changes in the hippocampus. The indicators of cell apoptosis and the BDNF/TRPC6 signaling activation in the mice hippocampus were detected by western blotting or real-time quantitative reverse transcription PCR experiments. RESULTS: We established the PTSD mice model induced by CCS, which exhibited significant PTSD-like phenotypes, including increased anxiety-like and depression-like behaviors. Medicinal cannabis oil treatment significantly ameliorated PTSD-like behaviors and improved brain histomorphological abnormalities in CCS mice. Mechanistically, medicinal cannabis oil reduced CCS-induced cell apoptosis and enhanced the activation of BDNF/TRPC6 signaling pathway. CONCLUSIONS: We constructed a PTSD model with CCS and medicinal cannabis oil that significantly improved anxiety-like and depressive-like behaviors in CCS mice, which may play an anti-PTSD role by stimulating the BDNF/TRPC6 signaling pathway.


Subject(s)
Anxiety , Brain-Derived Neurotrophic Factor , Depression , Disease Models, Animal , Hippocampus , Signal Transduction , Stress Disorders, Post-Traumatic , TRPC6 Cation Channel , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/drug effects , Mice , Signal Transduction/drug effects , Anxiety/drug therapy , Anxiety/metabolism , Male , Depression/drug therapy , Depression/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , TRPC6 Cation Channel/metabolism , Behavior, Animal/drug effects , Medical Marijuana/pharmacology , Mice, Inbred C57BL , Apoptosis/drug effects , Plant Oils/pharmacology , Plant Oils/administration & dosage , Stress, Psychological/drug therapy , Stress, Psychological/metabolism
2.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673977

ABSTRACT

Transient receptor potential canonical sub-family channel 3 (TRPC3) is considered to play a critical role in calcium homeostasis. However, there are no established findings in this respect with regard to TRPC6. Although the parathyroid gland is a crucial organ in calcium household regulation, little is known about the protein distribution of TRPC channels-especially TRPC3 and TRPC6-in this organ. Our aim was therefore to investigate the protein expression profile of TRPC3 and TRPC6 in healthy and diseased human parathyroid glands. Surgery samples from patients with healthy parathyroid glands and from patients suffering from primary hyperparathyroidism (pHPT) were investigated by immunohistochemistry using knockout-validated antibodies against TRPC3 and TRPC6. A software-based analysis similar to an H-score was performed. For the first time, to our knowledge, TRPC3 and TRPC6 protein expression is described here in the parathyroid glands. It is found in both chief and oxyphilic cells. Furthermore, the TRPC3 staining score in diseased tissue (pHPT) was statistically significantly lower than that in healthy tissue. In conclusion, TRPC3 and TRPC6 proteins are expressed in the human parathyroid gland. Furthermore, there is strong evidence indicating that TRPC3 plays a role in pHPT and subsequently in parathyroid hormone secretion regulation. These findings ultimately require further research in order to not only confirm our results but also to further investigate the relevance of these channels and, in particular, that of TRPC3 in the aforementioned physiological functions and pathophysiological conditions.


Subject(s)
Down-Regulation , Hyperparathyroidism, Primary , Parathyroid Glands , TRPC Cation Channels , TRPC6 Cation Channel , Humans , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Hyperparathyroidism, Primary/metabolism , Hyperparathyroidism, Primary/genetics , Hyperparathyroidism, Primary/pathology , Parathyroid Glands/metabolism , Parathyroid Glands/pathology , Female , Male , TRPC6 Cation Channel/metabolism , TRPC6 Cation Channel/genetics , Middle Aged , Aged , Adult , Immunohistochemistry , Parathyroid Hormone/metabolism
3.
Physiol Res ; 73(1): 69-80, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38466006

ABSTRACT

beta3-adrenergic activation causes Ca2+ release from the mitochondria and subsequent Ca2+ release from the endoplasmic reticulum (ER), evoking store-operated Ca2+ entry (SOCE) due to Ca2+ depletion from the ER in mouse brown adipocytes. In this study, we investigated how Ca2+ depletion from the ER elicits SOCE in mouse brown adipocytes using fluorometry of intracellular Ca2+ concentration ([Ca2+]i). The administration of cyclopiazonic acid (CPA), a reversible sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA) pump blocker in the ER, caused an increase in [Ca2+]i. Moreover, CPA induced SOCE was suppressed by the administration of a Ca2+ free Krebs solution and the transient receptor potential canonical 6 (TRPC6) selective blockers 2-APB, ML-9 and GsMTx-4 but not Pico145, which blocks TRPC1/4/5. Administration of TRPC6 channel agonist 1-oleoyl-2-acetyl-sn-glycerol (OAG) and flufenamic acid elicited Ca2+ entry. Moreover, our RT-PCR analyses detected mRNAs for TRPC6 in brown adipose tissues. In addition, western blot analyses showed the expression of the TRPC6 protein. Thus, TRPC6 is one of the Ca2+ pathways involved in SOCE. These modes of Ca2+ entry provide the basis for heat production via activation of Ca2+-dependent dehydrogenase and the expression of uncoupling protein 1 (UCP1). Enhancing thermogenic metabolism in brown adipocytes may serve as broad therapeutic utility to reduce obesity and metabolic syndrome.


Subject(s)
Transient Receptor Potential Channels , Mice , Animals , TRPC6 Cation Channel/metabolism , Transient Receptor Potential Channels/metabolism , TRPC Cation Channels/metabolism , Calcium/metabolism , Adipocytes, Brown/metabolism , Endoplasmic Reticulum/metabolism , Calcium Signaling
4.
Int J Mol Sci ; 25(3)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38338818

ABSTRACT

TRPV4 channels, which respond to mechanical activation by permeating Ca2+ into the cell, may play a pivotal role in cardiac remodeling during cardiac overload. Our study aimed to investigate TRPV4 involvement in pathological and physiological remodeling through Ca2+-dependent signaling. TRPV4 expression was assessed in heart failure (HF) models, induced by isoproterenol infusion or transverse aortic constriction, and in exercise-induced adaptive remodeling models. The impact of genetic TRPV4 inhibition on HF was studied by echocardiography, histology, gene and protein analysis, arrhythmia inducibility, Ca2+ dynamics, calcineurin (CN) activity, and NFAT nuclear translocation. TRPV4 expression exclusively increased in HF models, strongly correlating with fibrosis. Isoproterenol-administered transgenic TRPV4-/- mice did not exhibit HF features. Cardiac fibroblasts (CFb) from TRPV4+/+ animals, compared to TRPV4-/-, displayed significant TRPV4 overexpression, elevated Ca2+ influx, and enhanced CN/NFATc3 pathway activation. TRPC6 expression paralleled that of TRPV4 in all models, with no increase in TRPV4-/- mice. In cultured CFb, the activation of TRPV4 by GSK1016790A increased TRPC6 expression, which led to enhanced CN/NFATc3 activation through synergistic action of both channels. In conclusion, TRPV4 channels contribute to pathological remodeling by promoting fibrosis and inducing TRPC6 upregulation through the activation of Ca2+-dependent CN/NFATc3 signaling. These results pose TRPV4 as a primary mediator of the pathological response.


Subject(s)
Calcineurin , Heart Failure , TRPV Cation Channels , Ventricular Remodeling , Animals , Mice , Calcineurin/metabolism , Cells, Cultured , Fibrosis , Heart Failure/metabolism , Isoproterenol , Mice, Transgenic , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Ventricular Remodeling/genetics
5.
Int J Mol Sci ; 25(4)2024 Feb 18.
Article in English | MEDLINE | ID: mdl-38397074

ABSTRACT

We recently reported that transient receptor potential canonical (TRPC) 6 channel activity contributes to intracellular Zn2+ homeostasis in the heart. Zn2+ has also been implicated in the regulation of intestinal redox and microbial homeostasis. This study aims to investigate the role of TRPC6-mediated Zn2+ influx in the stress resistance of the intestine. The expression profile of TRPC1-C7 mRNAs in the actively inflamed mucosa from inflammatory bowel disease (IBD) patients was analyzed using the GEO database. Systemic TRPC3 knockout (KO) and TRPC6 KO mice were treated with dextran sulfate sodium (DSS) to induce colitis. The Zn2+ concentration and the mRNA expression levels of oxidative/inflammatory markers in colon tissues were quantitatively analyzed, and gut microbiota profiles were compared. TRPC6 mRNA expression level was increased in IBD patients and DSS-treated mouse colon tissues. DSS-treated TRPC6 KO mice, but not TRPC3 KO mice, showed severe weight loss and increased disease activity index compared with DSS-treated WT mice. The mRNA abundances of antioxidant proteins were basically increased in the TRPC6 KO colon, with changes in gut microbiota profiles. Treatment with TRPC6 activator prevented the DSS-induced colitis progression accompanied by increasing Zn2+ concentration. We suggest that TRPC6-mediated Zn2+ influx activity plays a key role in stress resistance against IBD, providing a new strategy for treating colitis.


Subject(s)
Inflammatory Bowel Diseases , TRPC6 Cation Channel , Animals , Humans , Mice , Colon/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Intestines , Mice, Inbred C57BL , RNA, Messenger/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(1): 60-69, 2024 Jan 20.
Article in Chinese | MEDLINE | ID: mdl-38293977

ABSTRACT

OBJECTIVE: To assess the effect of platycodin D (PD) for alleviating pulmonary fibrosis in mice and explore the underlying mechanism. METHODS: C57BL/6J mouse models of pulmonary fibrosis induced by bleomycin injection into the airway were treated with daily intragastric administration of 10 mg/kg PD for 28 days. The changes of pulmonary fibrosis and the expression and distribution of transient receptor potential cation channel subfamily C member 6 (TRPC6) were evaluated with immunohistochemistry, HE staining and Sirius Red staining. Western blotting was used to detect α-SMA expression in the lung tissues of the mice. Primary cultures of mouse lung fibroblasts were pretreated with PD (2.5, 5.0, and 10 µmol/L) or larixyl acetate (LA; 10 µmol/L) before exposure to 10 ng/mL transforming growth factor-ß1 (TGF-ß1), and the changes in cell survival rate, expressions of collagen Ⅰ, α-SMA and TRPC6, reactive oxygen species (ROS) production, mitochondrial membrane potential, and cell proliferation capacity were assessed. Network pharmacology analysis was performed to explore the mechanism by which PD alleviated pulmonary fibrosis. RESULTS: PD treatment significantly alleviated pulmonary fibrosis and reduced α-SMA expression in BLM-induced mouse models (P<0.05). In TGF-ß1-induced primary mouse lung fibroblasts, PD effectively inhibited the cell proliferation, reduced ROS production (P<0.0001), rescued the reduction of mitochondrial membrane potential (P<0.001), and inhibited the expressions of α-SMA and collagen Ⅰ (P<0.05). Network pharmacology analysis suggested that TRPC6 mediated the effect of PD for alleviating pulmonary fibrosis. Immunohistochemistry showed that PD significantly reduced TRPC6 expression in the lung tissues of BLM-induced mice. In primary mouse lung fibroblasts, PD significantly inhibited TGF-ß1-induced TRPC6 expression (P<0.05), and LA treatment obviously lowered the expression levels of TRPC6, α-SMA and collagenⅠ (P<0.05). CONCLUSION: PD alleviated pulmonary fibrosis in mice possibly by down-regulating TRPC6 and reducing ROS production.


Subject(s)
Pulmonary Fibrosis , Saponins , Triterpenes , Mice , Animals , Pulmonary Fibrosis/chemically induced , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , TRPC6 Cation Channel/metabolism , TRPC6 Cation Channel/therapeutic use , Mice, Inbred C57BL , Lung/pathology , Fibroblasts , Bleomycin/adverse effects , Collagen Type I
7.
ACS Nano ; 18(6): 4871-4885, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38290527

ABSTRACT

Diabetic wounds exhibit delayed and incomplete healing, usually due to vascular and nerve damage. Dysregulation of cellular Ca2+ homeostasis has recently been shown to be closely related to insulin resistance and type 2 diabetes mellitus. However, the involvement of this dysregulation in diabetic wound complications remains unknown. In this study, we found calcium dysregulation in patients with diabetic ulcers via tissue protein profiling. High glucose and glucometabolic toxicant stimulation considerably impaired the function of TRPC6, a pore subunit of transient receptor potential channels mediating Ca2+ influx, and mitochondria, which regulate calcium cycling and metabolism. Furthermore, we found that mesenchymal stem cell (MSC)-derived small extracellular vesicles (MSC-sEVs) could play a dual role in restoring the function of TRPC6 and mitochondria by delivering transcription factor SP2 and deubiquitinating enzyme USP9, respectively. MSC-sEVs could transfer SP2 that activated TRPC6 expression by binding to its specific promoter regions (-1519 to -1725 bp), thus recovering Ca2+ influx and downstream pathways. MSC-sEVs also promoted mitophagy to restore mitochondrial function by transporting USP9 that stabilized the expression of Parkin, a major player in mitophagy, thereby guaranteeing Ca2+ efflux and avoidance of Ca2+ overload. Targeting the regulation of calcium homeostasis provides a perspective for understanding diabetic wound healing, and the corresponding design of MSC-sEVs could be a potential therapeutic strategy.


Subject(s)
Diabetes Mellitus, Type 2 , Extracellular Vesicles , Mesenchymal Stem Cells , Humans , Diabetes Mellitus, Type 2/metabolism , TRPC6 Cation Channel/metabolism , Calcium/metabolism , Wound Healing/physiology , Mesenchymal Stem Cells/metabolism , Extracellular Vesicles/metabolism , Mitochondria/metabolism
8.
Clin Hemorheol Microcirc ; 86(3): 369-382, 2024.
Article in English | MEDLINE | ID: mdl-37980653

ABSTRACT

AIM: To investigate the mechanism by which NF-κB p65 activates miR-150 to suppress TRPC6 expression and promote renal ischemia-reperfusion injury. METHODS: To assess the transcription of miR-150, NF-B p65, and TRPC6 in HK-2 cells treated with hypoxia reperfusion and rat kidney tissue damaged by ischemia-reperfusion (I/R), qPCR was implemented. The protein production of NF-κB p65 and TRPC6 was assessed by Western blot (WB) analysis. The histological score of rat kidney tissue was assessed using H&E (hematoxylin and eosin) staining. To assess the rate of apoptosis of renal tissue cells following I/R injury, we used the TACS TdT In Situ Apoptosis Detection Kit. To find out the impairment of renal function, blood levels of creatinine (Cr) and blood urea nitrogen (BUN) were tested in rats. Concentrations of inflammatory cytokines, including IL-1ß, IL-10, and TNF-α, were detected in HK-2 cells and rat renal tissue cells utilizing ELISA kits. FITC and CCK-8 were employed to analyze the death rate and cellular proliferation of HK-2 cells. To analyse the mechanism of engagement between NF-κB p65 and the miR-150 promoter, coupled with the detrimental impact of miR-150 on TRPC6, we adopted the dual-luciferase reporter assay. To confirm the activating effect of NF-κB p65 on miR-150,we implemented the ChIP assay. RESULTS: NF-κB p65 expression was significantly upregulated in rat renal tissue following IRI. Applying the dual-luciferase reporter assay, we demonstrated that the specific attachment of NF-B p65 with the miR-150 promoter location is viable, resulting in the promotion of the activity of the promoter. When miR-150 was overexpressed, we observed a notable reduction in cell proliferation. And it notably increased the rate of cellular apoptosis rate and amounts of the proinflammatory cytokines IL-1ß, IL-10, and TNF-α. Employing the dual-luciferase reporter assay, we demonstrated that miR-150 transfection diminished the function of luciferase in the TRPC6-WT group, whereas luciferase activity in the TRPC6-MUT group remained unchanged, indicating that miR-150 is a targeted inhibitor of TRPC6. In the rat renal I/R model, when miR-150 was inhibited or TRPC6 was overexpressed in the rat kidney I/R model, the histological score of rat kidney tissue significantly decreased, so did the quantities of proinflammatory cytokines IL-1ß, IL-10, TNF-α, creatinine (Cr) and blood urea nitrogen (BUN) contents and the rate of cell apoptosis in kidney tissue. CONCLUSION: Activation of miR-150 by NF-κB p65 results in downregulation of TRPC6 expression and promotion of IRI in the kidney.


Subject(s)
MicroRNAs , Reperfusion Injury , Rats , Animals , NF-kappa B/metabolism , Interleukin-10/metabolism , Tumor Necrosis Factor-alpha/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism , Creatinine/pharmacology , Signal Transduction , Rats, Sprague-Dawley , Kidney/pathology , Cytokines/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Luciferases/metabolism , Luciferases/pharmacology
9.
Curr Probl Cardiol ; 49(1 Pt B): 102112, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37774899

ABSTRACT

Cardiovascular disease is the leading cause of death, medical complications, and healthcare costs. Although recent advances have been in treating cardiovascular disorders linked with a reduced ejection fraction, acutely decompensate cardiac failure remains a significant medical problem. The transient receptor potential cation channel (TRPC6) family responds to neurohormonal and mechanical stress, playing critical roles in cardiovascular diseases. Therefore, TRP C6 channels have great promise as therapeutic targets. Numerous studies have investigated the roles of TRP C6 channels in pain neurons, highlighting their significance in cardiovascular research. The TRPC6 protein exhibits a broad distribution in various organs and tissues, including the brain, nerves, heart, blood vessels, lungs, kidneys, gastrointestinal tract, and other bodily structures. Its activation can be triggered by alterations in osmotic pressure, mechanical stimulation, and diacylglycerol. Consequently, TRPC6 plays a significant role in the pathophysiological mechanisms underlying diverse diseases within living organisms. A recent study has indicated a strong correlation between the disorder known as TRPC6 and the development of cardiovascular diseases. Consequently, investigations into the association between TRPC6 and cardiovascular diseases have gained significant attention in the scientific community. This review explores the most recent developments in the recognition and characterization of TRPC6. Additionally, it considers the field's prospects while examining how TRPC6 might be altered and its clinical applications.


Subject(s)
Cardiovascular Diseases , TRPC6 Cation Channel , Humans , Lung/metabolism , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/metabolism
10.
Int J Mol Sci ; 24(23)2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38069081

ABSTRACT

Acute lung injury (ALI) is characterized by endothelial barrier disruption and associated inflammatory responses, and transient receptor potential cation channel 6 (TRPC6)-mediated Ca2+ influx is critical for endothelial hyperpermeability. In this study, we investigated the role of TRPC6 in LPS-induced ALI, analyzed gene expression in WT and TRPC6-/- lungs using RNA sequencing, and explored the effects of TRPC6 in the LPS-induced hyperpermeability in human umbilical vein endothelial cells (HUVECs) to elucidate the underlying mechanisms. Intratracheal instillation of LPS caused edema in the mouse lungs. Deletion of TRPC6 reduced LPS-induced lung edema and decreased cell infiltration. RNA sequencing analysis suggested that downregulated cell adhesion molecules in TRPC6-/- lungs may be responsible for their resistance to LPS-induced injury. In addition, downregulation of TRPC6 significantly alleviated the LPS-induced decrease in eNOS expression in lung tissue as well as in HUVECs. Moreover, inhibition of TRPC6 with the channel antagonist larixyl led to a decrease in LPS-induced hyperpermeability and ROS production in HUVECs, which could be reversed by blocking eNOS. Our findings suggest that inhibition of TRPC6 ameliorates LPS-induced ALI, which may be achieved by acting on the cell adhesion molecule signaling pathway and participating in the regulation of eNOS levels in endothelial cells.


Subject(s)
Acute Lung Injury , Transient Receptor Potential Channels , Animals , Humans , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Edema/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Lipopolysaccharides/adverse effects , Lung/metabolism , Transient Receptor Potential Channels/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism
11.
Clin Sci (Lond) ; 137(24): 1789-1804, 2023 12 22.
Article in English | MEDLINE | ID: mdl-38051199

ABSTRACT

Angiotensin receptor blockers (ARBs) are the first-line treatment for hypertension; they act by inhibiting signaling through the angiotensin 1 receptor (AT1R). Recently, a novel biased AT1R agonist, TRV120027 (TRV), which selectively activates the ß-arrestin cascade and blocks the G-protein-coupled receptor pathway has been proposed as a potential blood pressure medication. Here, we explored the effects of TRV and associated ß-arrestin signaling in podocytes, essential cells of the kidney filter. We used human podocyte cell lines to determine ß-arrestin's involvement in calcium signaling and cytoskeletal reorganization and Dahl SS rats to investigate the chronic effects of TRV administration on glomerular health. Our experiments indicate that the TRV-activated ß-arrestin pathway promotes the rapid elevation of intracellular Ca2+ in a dose-dependent manner. Interestingly, the amplitude of ß-arrestin-mediated Ca2+ influx was significantly higher than the response to similar Ang II concentrations. Single-channel analyses show rapid activation of transient receptor potential canonical (TRPC) channels following acute TRV application. Furthermore, the pharmacological blockade of TRPC6 significantly attenuated the ß-arrestin-mediated Ca2+ influx. Additionally, prolonged activation of the ß-arrestin pathway in podocytes resulted in pathological actin cytoskeleton rearrangements, higher apoptotic cell markers, and augmented glomerular damage. TRV-activated ß-arrestin signaling in podocytes may promote TRPC6 channel-mediated Ca2+ influx, foot process effacement, and apoptosis, possibly leading to severe defects in glomerular filtration barrier integrity and kidney health. Under these circumstances, the potential therapeutic application of TRV for hypertension treatment requires further investigation to assess the balance of the benefits versus possible deleterious effects and off-target damage.


Subject(s)
Hypertension , Kidney Diseases , Podocytes , Rats , Animals , Humans , Podocytes/metabolism , TRPC6 Cation Channel/metabolism , Calcium/metabolism , beta-Arrestins/metabolism , Angiotensin Receptor Antagonists/pharmacology , Rats, Inbred Dahl , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney Diseases/metabolism , Hypertension/metabolism , TRPC Cation Channels/metabolism , TRPC Cation Channels/pharmacology
12.
Front Biosci (Landmark Ed) ; 28(10): 240, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37919067

ABSTRACT

Calcium (Ca2+) plays a critical role in podocyte function. The Ca2+-sensitive receptors on the cell surface can sense changes in Ca2+ concentration, and Ca2+ flow into podocytes, after activation of Ca2+ channels (such as transient receptor potential canonical (TRPC) channels and N-type calcium channels) by different stimuli. In addition, the type 2 ryanodine receptor (RyR2) and the voltage-dependent anion channel 1 (VDAC1) on mitochondrial store-operated calcium channels (SOCs) on the endoplasmic reticulum maintain the Ca2+ homeostasis of the organelle. Ca2+ signaling is transmitted through multiple downstream signaling pathways and participates in the morphogenesis, structural maintenance, and survival of podocytes. When Ca2+ is dysregulated, it leads to the occurrence and progression of various diseases, such as focal segmental glomerulosclerosis, diabetic kidney disease, lupus nephritis, transplant glomerulopathy, and hypertensive renal injury. Ca2+ signaling is a promising therapeutic target for podocyte-related diseases. This review first summarizes the role of Ca2+ sensing, Ca2+ channels, and different Ca2+-signaling pathways in the biological functions of podocytes, then, explores the status of Ca2+ signaling in different podocyte-related diseases and its advances as a therapeutic target.


Subject(s)
Diabetic Nephropathies , Podocytes , Humans , Podocytes/metabolism , Podocytes/pathology , Calcium Signaling , TRPC6 Cation Channel/metabolism , Calcium/metabolism , Diabetic Nephropathies/metabolism
13.
Front Biosci (Landmark Ed) ; 28(9): 231, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37796683

ABSTRACT

BACKGROUND: Diabetes mellitus type 2 is a risk factor for developing heart failure and myocardial fibrosis, but there is no specific therapy for diabetic heart disease. 1-[2-(4-methoxyphenyl)]-2-[3-(4-methoxyphenyl) propoxy]ethyl-1H-imidazole (SKF96365) is regarded as an inhibitor of receptor-mediated calcium ion (Ca2+) entry. This study aimed to explore the effects of SKF96365 on diabetic myocardial fibrosis. METHODS: A type 2 diabetic rat model induced by a high-sugar and high-fat diet combined with streptozotocin was established. Thirty specific pathogen-free male Wistar rats were divided randomly into three groups: group A (the blank control group), group B (the diabetes group) and group C (the diabetes + transient receptor potential canonical channel [TRPC] blocker intervention group). Group C was given 0.74-µmol/kg SKF96365 by intraperitoneal injection, and groups A and B were given the same amount of normal saline by intraperitoneal injection. The weight and blood sugar of the rats were monitored. After 12 weeks, the weight of the whole heart and the left ventricle was measured, and the heart and the left ventricular weight ratios were calculated. Haematoxylin-eosin (HE) staining was used to observe pathological changes in the myocardial tissue and the distribution of nuclei. Masson staining was used to identify collagen and muscle fibres, and the myocardial collagen volume fraction (CVF) was calculated. Semi-quantitative reverse transcription-polymerase chain reaction was used to detect the messenger ribonucleic acid (mRNA) expression of SKF96365 target genes. A value of p < 0.05 indicated that the difference between the groups was statistically significant. RESULTS: Compared with the weight of the rats in group A, the weight of those in groups B and C decreased, while blood sugar, whole heart weight and left ventricular weight increased (p < 0.05). There was no significant difference in body weight between the rats in groups B and C (p > 0.05). The HE staining results showed that the arrangement of cardiomyocytes in groups B and C was irregular, and focal necrosis was seen in severe cases. The degree of diabetic cardiomyopathy (DCM) in group C was less severe than that in group B. Masson staining showed that the CVF increased in groups B and C, with group B > group C (p < 0.05); the mRNA expressions of TRPC3 and TRPC6 were upregulated in groups A, B and C, and the mRNA expressions of TRPC3 and TRPC6 in group C were downregulated compared with those in group B (p < 0.05). Compared with the expression levels of SKF96365 target genes (STIM1, Orai1 and Homer1) in group A, those in group B were lower, while the administration of SKF96365 in group C did not affect the expression levels of those genes. CONCLUSIONS: SKF96365 can effectively improve myocardial fibrosis in type-II diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Rats , Male , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Blood Glucose/metabolism , TRPC6 Cation Channel/metabolism , Rats, Wistar , Myocardium/metabolism , Imidazoles/pharmacology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Fibrosis , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Collagen/metabolism
14.
Diabetes ; 72(12): 1795-1808, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37722138

ABSTRACT

There is clinical evidence that increased urinary serine proteases are associated with the disease severity in the setting of diabetic nephropathy (DN). Elevation of serine proteases may mediate [Ca2+]i dynamics in podocytes through the protease-activated receptors (PARs) pathway, including associated activation of nonspecific cation channels. Cultured human podocytes and freshly isolated glomeruli were used for fluorescence and immunohistochemistry stainings, calcium imaging, Western blot analysis, scanning ion conductance microscopy, and patch clamp analysis. Goto-Kakizaki, Wistar, type 2 DN (T2DN), and a novel PAR1 knockout on T2DN rat background rats were used to test the importance of PAR1-mediated signaling in DN settings. We found that PAR1 activation increases [Ca2+]i via TRPC6 channels. Both human cultured podocytes exposed to high glucose and podocytes from freshly isolated glomeruli of T2DN rats had increased PAR1-mediated [Ca2+]i compared with controls. Imaging experiments revealed that PAR1 activation plays a role in podocyte morphological changes. T2DN rats exhibited a significantly higher response to thrombin and urokinase. Moreover, the plasma concentration of thrombin in T2DN rats was significantly elevated compared with Wistar rats. T2DNPar1-/- rats were embryonically lethal. T2DNPar1+/- rats had a significant decrease in glomerular damage associated with DN lesions. Overall, these data provide evidence that, during the development of DN, elevated levels of serine proteases promote an excessive [Ca2+]i influx in podocytes through PAR1-TRPC6 signaling, ultimately leading to podocyte apoptosis, the development of albuminuria, and glomeruli damage. ARTICLE HIGHLIGHTS: Increased urinary serine proteases are associated with diabetic nephropathy. During the development of diabetic nephropathy in type 2 diabetes, the elevation of serine proteases could overstimulate protease-activated receptor 1 (PAR1). PAR1 signaling is involved in the development of DN via TRPC6-mediated intracellular calcium signaling. This study provides fundamental knowledge that can be used to develop efficient therapeutic approaches targeting serine proteases or corresponding PAR pathways to prevent or slow the progression of diabetes-associated kidney diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Podocytes , Rats , Humans , Animals , Diabetic Nephropathies/metabolism , Podocytes/metabolism , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Receptor, PAR-1/therapeutic use , TRPC6 Cation Channel/metabolism , TRPC6 Cation Channel/therapeutic use , Calcium/metabolism , Diabetes Mellitus, Type 2/metabolism , Thrombin/metabolism , Thrombin/therapeutic use , Rats, Wistar
15.
Arterioscler Thromb Vasc Biol ; 43(8): 1494-1509, 2023 08.
Article in English | MEDLINE | ID: mdl-37381987

ABSTRACT

BACKGROUND: MAGT1 (magnesium transporter 1) is a subunit of the oligosaccharide protein complex with thiol-disulfide oxidoreductase activity, supporting the process of N-glycosylation. MAGT1 deficiency was detected in human patients with X-linked immunodeficiency with magnesium defect syndrome and congenital disorders of glycosylation, resulting in decreased cation responses in lymphocytes, thereby inhibiting the immune response against viral infections. Curative hematopoietic stem cell transplantation of patients with X-linked immunodeficiency with magnesium defect causes fatal bleeding and thrombotic complications. METHODS: We studied the role of MAGT1 deficiency in platelet function in relation to arterial thrombosis and hemostasis using several in vitro experimental settings and in vivo models of arterial thrombosis and transient middle cerebral artery occlusion model of ischemic stroke. RESULTS: MAGT1-deficient mice (Magt1-/y) displayed accelerated occlusive arterial thrombus formation in vivo, a shortened bleeding time, and profound brain damage upon focal cerebral ischemia. These defects resulted in increased calcium influx and enhanced second wave mediator release, which further reinforced platelet reactivity and aggregation responses. Supplementation of MgCl2 or pharmacological blockade of TRPC6 (transient receptor potential cation channel, subfamily C, member 6) channel, but not inhibition of store-operated calcium entry, normalized the aggregation responses of Magt1-/y platelets to the control level. GP (glycoprotein) VI activation of Magt1-/y platelets resulted in hyperphosphorylation of Syk (spleen tyrosine kinase), LAT (linker for activation of T cells), and PLC (phospholipase C) γ2, whereas the inhibitory loop regulated by PKC (protein kinase C) was impaired. A hyperaggregation response to the GPVI agonist was confirmed in human platelets isolated from a MAGT1-deficient (X-linked immunodeficiency with magnesium defect) patient. Haploinsufficiency of TRPC6 in Magt1-/y mice could normalize GPVI signaling, platelet aggregation, and thrombus formation in vivo. CONCLUSIONS: These results suggest that MAGT1 and TRPC6 are functionally linked. Therefore, deficiency or impaired functionality of MAGT1 could be a potential risk factor for arterial thrombosis and stroke.


Subject(s)
Cation Transport Proteins , Homeostasis , Infarction, Middle Cerebral Artery , Ischemic Stroke , Thrombosis , Animals , Humans , Mice , Blood Platelets/metabolism , Calcium/metabolism , Cations/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/complications , Ischemic Stroke/metabolism , Magnesium/metabolism , Platelet Activation , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Thrombosis/genetics , Thrombosis/metabolism , TRPC6 Cation Channel/metabolism , Cation Transport Proteins/deficiency
16.
Toxicol Appl Pharmacol ; 470: 116547, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37178933

ABSTRACT

Daunorubicin (DNR-) induced cardiotoxicity seriously restricts its clinical application. Transient receptor potential cation channel subfamily C member 6 (TRPC6) is involved in multiple cardiovascular physiological and pathophysiological processes. However, the role of TRPC6 anthracycline-induced cardiotoxicity (AIC) remains unclear. Mitochondrial fragmentation greatly promotes AIC. TRPC6-mediated ERK1/2 activation has been shown to favor mitochondrial fission in dentate granule cells. The aim of the present study was to elucidate the effects of TRPC6 on daunorubicin- induced cardiotoxicity and identify the mechanisms associated with mitochondrial dynamics. The sparkling results showed that TRPC6 was upregulated in models in vitro and in vivo. TRPC6 knockdown protected cardiomyocytes from DNR-induced cell apoptosis and death. DNR largely facilitated mitochondrial fission, boosted mitochondrial membrane potential collapse and damaged debilitated mitochondrial respiratory function in H9c2 cells,these effects were accompanied by TRPC6 upregulation. siTRPC6 effectively inhibited these mitochondrial adverse aspects showing a positive unexposed effect on mitochondrial morphology and function. Concomitantly, ERK1/2-DRP1 which is related to mitochondrial fission was significantly activated with amplified phosphorylated forms in DNR-treated H9c2 cells. siTRPC6 effectively suppressed ERK1/2-DPR1 over activation, hinting at a potential correlation between TRPC6 and ERK1/2-DRP1 by which mitochondrial dynamics are possibly modulated in AIC. TRPC6 knockdown also raised the Bcl-2/Bax ratio, which may help to block mitochondrial fragmentation-related functional impairment and apoptotic signaling. These findings suggested an essential role of TRPC6 in AIC by intensifying mitochondrial fission and cell death via ERK1/2-DPR1, which could be a potential therapeutic target for AIC.


Subject(s)
Daunorubicin , Myocytes, Cardiac , TRPC6 Cation Channel , Animals , Rats , Apoptosis , Cardiotoxicity/metabolism , Cell Death , Daunorubicin/toxicity , Dynamins/metabolism , MAP Kinase Signaling System , Mitochondrial Dynamics , Myocytes, Cardiac/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/metabolism
17.
Biomed Pharmacother ; 161: 114480, 2023 May.
Article in English | MEDLINE | ID: mdl-37002575

ABSTRACT

Intracranial aneurysm (IA) is a frequent cerebrovascular disorder with unclear pathogenesis. The vascular smooth muscle cells (VSMCs) phenotypic switch is essential for IA formation. It has been reported that Ca2+ overload and excessive reactive oxygen species (ROS) are involved in VSMCs phenotypic switch. The transient receptor potential canonical 6 (TRPC6) and NADPH oxidase 4 (NOX4) are the main pathway to participate in Ca2+ overload and ROS production in VSMCs. Ca2+ overload can activate calcineurin (CN), leading to nuclear factor of activated T cell (NFAT) dephosphorylation to regulate the target gene's transcription. We hypothesized that activation of TRPC6-NFATC1 signaling may upregulate NOX4 and involve in VSMCs phenotypic switch contributing to the progression of IA. Our results showed that the expressions of NOX4, p22phox, p47phox, TRPC6, CN and NFATC1 were significantly increased, and VSMCs underwent a significant phenotypic switch in IA tissue and cellular specimens. The VIVIT (NFATC1 inhibitor) and BI-749327 (TRPC6 inhibitor) treatment reduced the expressions of NOX4, p22phox and p47phox and the production of ROS, and significantly improved VSMCs phenotypic switch in IA rats and cells. Consistent results were obtained from IA Trpc6 knockout (Trpc6-/-) mice. Furthermore, the results also revealed that NFATC1 could regulate NOX4 transcription by binding to its promoter. Our findings reveal that interrupting the TRPC6-NFATC1 signaling inhibits NOX4 and improves VSMCs phenotypic switch in IA, and regulating Ca2+ homeostasis may be an important therapeutic strategy for IA.


Subject(s)
Intracranial Aneurysm , Animals , Mice , Rats , Intracranial Aneurysm/metabolism , Muscle, Smooth, Vascular/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NADPH Oxidases/metabolism , NFATC Transcription Factors/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/metabolism
18.
Inflammation ; 46(4): 1144-1160, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37072606

ABSTRACT

The mammalian Transient Receptor Potential Canonical (TRPC) subfamily comprises seven transmembrane proteins (TRPC1-7) forming cation channels in the plasma membrane of mammalian cells. TRPC channels mediate Ca2+ and Na+ influx into the cells. Amongst TRPCs, TRPC6 deficiency or increased activity due to gain-of-function mutations has been associated with a multitude of diseases, such as kidney disease, pulmonary disease, and neurological disease. Indeed, the TRPC6 protein is expressed in various organs and is involved in diverse signalling pathways. The last decade saw a surge in the investigative studies concerning the physiological roles of TRPC6 and describing the development of new pharmacological tools modulating TRPC6 activity. The current review summarizes the progress achieved in those investigations.


Subject(s)
TRPC Cation Channels , Transient Receptor Potential Channels , Animals , TRPC6 Cation Channel/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Signal Transduction , Membrane Proteins/metabolism , Calcium/metabolism , Mammals/metabolism
19.
Int J Mol Sci ; 24(8)2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37108424

ABSTRACT

Type 2 diabetes mellitus (DM2) is a widespread metabolic disorder that results in podocyte damage and diabetic nephropathy. Previous studies demonstrated that TRPC6 channels play a pivotal role in podocyte function and their dysregulation is associated with development of different kidney diseases including nephropathy. Here, using single channel patch clamp technique, we demonstrated that non-selective cationic TRPC6 channels are sensitive to the Ca2+ store depletion in human podocyte cell line Ab8/13 and in freshly isolated rat glomerular podocytes. Ca2+ imaging indicated the involvement of ORAI and sodium-calcium exchanger in Ca2+ entry induced upon store depletion. In male rats fed a high-fat diet combined with a low-dose streptozotocin injection, which leads to DM2 development, we observed the reduction of a store-operated Ca2+ entry (SOCE) in rat glomerular podocytes. This was accompanied by a reorganization of store-operated Ca2+ influx such that TRPC6 channels lost their sensitivity to Ca2+ store depletion and ORAI-mediated Ca2+ entry was suppressed in TRPC6-independent manner. Altogether our data provide new insights into the mechanism of SOCE organization in podocytes in the norm and in pathology, which should be taken into account when developing pharmacological treatment of the early stages of diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Podocytes , Humans , Rats , Male , Animals , TRPC6 Cation Channel/metabolism , Podocytes/metabolism , Calcium Channels/metabolism , Diabetic Nephropathies/metabolism , Calcium/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Experimental/metabolism , TRPC Cation Channels/metabolism
20.
J Ethnopharmacol ; 313: 116561, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37121453

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Liuwei Dihuang (LWDH) is a classic prescription that has been used to the treatment of "Kidney-Yin" deficiency syndrome for more than 1000 years in China. Recent studies have confirmed that LWDH can prevent the progression of renal fibrosis. Numerous studies have demonstrated the critical role that TRPC6 plays in the development of renal fibrosis. Due to the complex composition of LWDH and its remarkable therapeutic effect on renal fibrosis, it is possible to discover new active ingredients targeting TRPC6 for the treatment of renal fibrosis. AIM OF STUDY: This study aimed to identify selective TRPC6 inhibitors from LWDH and evaluate their therapeutical effects on renal fibrosis. MATERIALS AND METHODS: Computer-aided drug design was used to screen the biologically active ingredients of LWDH, and their affinities to human TRPC6 protein were detected by microcalorimetry. TRPC6, TRPC3, and TRPC7 over-expressed HEK293 cells were constructed, and the selective activities of the compounds on TRPC6 were determined by measuring [Ca2+]i in these cells. To establish an in vitro model of renal fibrosis, human renal proximal tubular epithelial HK-2 cells were stimulated with TGF-ß1. The therapeutic effects of LWDH compounds on renal fibrosis were then tested by detecting the related proteins. TRPC6 was knocked-down in HK-2 cells to investigate the effects of LWDH active ingredients on TRPC6. Finally, a unilateral ureteral obstruction model of renal fibrosis was established to test the therapeutic effect. RESULTS: From hundreds of LWDH ingredients, 64 active components with oral bioavailability ≥30% and drug-likeness index ≥0.18 were acquired. A total of 10 active components were obtained by molecular docking with TRPC6 protein. Among them, 4 components had an affinity with TRPC6. Piperlonguminine (PLG) had the most potent affinity with TRPC6 and blocking effect on TRPC6-mediated Ca2+ entry. A 100 µM of PLG showed no detectable inhibition on TRPC1, TRPC3, TRPC4, TRPC5, or TRPC7-mediated Ca2+ influx into cells. In vitro results indicated that PLG concentration-dependently inhibited the abnormally high expression of α-smooth muscle actin (α-SMA), collagen I, vimentin, and TRPC6 in TGF-ß1-induced HK-2 cells. Consistently, PLG also could not further inhibit TGF-ß1-induced expressions of these protein biomarkers in TRPC6 knocked-down HK-2 cells. In vivo, PLG dose-dependently reduced urinary protein, serum creatinine, and blood urea nitrogen levels in renal fibrosis mice and markedly alleviated fibrosis and the expressions of α-SMA, collagen I, vimentin, and TRPC6 in kidney tissues. CONCLUSION: Our results showed that PLG had anti-renal fibrosis effects by selectively inhibiting TRPC6. PLG might be a promising therapeutic agent for the treatment of renal fibrosis.


Subject(s)
Kidney Diseases , Ureteral Obstruction , Humans , Mice , Animals , TRPC6 Cation Channel/metabolism , Transforming Growth Factor beta1/metabolism , Vimentin , HEK293 Cells , Molecular Docking Simulation , Kidney Diseases/metabolism , Ureteral Obstruction/metabolism , Fibrosis , Collagen/metabolism , Kidney
SELECTION OF CITATIONS
SEARCH DETAIL
...