Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 689
Filter
1.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(4): 473-479, 2024 Apr 10.
Article in Chinese | MEDLINE | ID: mdl-38565515

ABSTRACT

OBJECTIVE: To summarize the clinical and genetic characteristics, treatment and prognosis of four children with Steroid-resistant nephrotic syndrome (SRNS) due to variants of TRPC6 gene. METHODS: Clinical data of four children with SRNS admitted to Children's Hospital Affiliated to Zhengzhou University between May 2020 and August 2022 were collected. Peripheral blood samples were collected from the children and their parents, and whole exome sequencing was carried out. Sanger sequencing was used to verify the pathogenicity of the candidate variants among the children and their parents. RESULTS: All of the four children were found to harbor heterozygous variants of the TRPC6 gene, including c.523C>T (p.R175W), c.1327T>A (p.F443I), c.430G>C (p.E144Q) (unreported previously), and c.523C>T (p.R175W), which were all missense variants. Two of the children have shown a simple type, whilst two have shown a nephritis type, none had extrarenal phenotype. Comprehensive renal pathology of three children revealed focal segmental glomerulosclerosis (FSGS). Two children were treated with steroids combined with calcineurin inhibitors (CNIs), among whom one showed significant improvement in symptoms. CONCLUSION: Discoveries of the novel c.430G>C variant and the new SRNS phenotype of the c.1327T>A variant have expanded the mutational and phenotypic spectrum of the TRPC6 gene, which has provided a reference for clinical diagnosis and genetic counseling for the families.


Subject(s)
Glomerulosclerosis, Focal Segmental , Nephrotic Syndrome , Child , Humans , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/genetics , Nephrotic Syndrome/diagnosis , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/therapeutic use , Phenotype , Kidney , Genotype , Mutation , Glomerulosclerosis, Focal Segmental/genetics
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.
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
4.
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
5.
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
6.
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
7.
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
8.
Mol Med Rep ; 29(3)2024 03.
Article in English | MEDLINE | ID: mdl-38275127

ABSTRACT

Heart failure is a primary cause of global mortality. In the present study, whether larixyl acetate, an inhibitor of transient receptor potential cation channel subfamily C member 6, attenuates pressure overload­induced heart failure in mice was investigated. To test this hypothesis, a transverse aortic constriction (TAC) animal model and an angiotensin II (Ang II)­treated H9c2 cell model were used. Cardiac and cellular structure, function and the expression levels of hypertrophy, endoplasmic reticulum (ER) stress, apoptosis, autophagy and pmTOR/mTOR related mRNAs or proteins were assessed to explore the underlying molecular mechanisms. The results indicated that treatment with TAC or Ang II leads to significant hypertrophy and dysfunction of the heart or H9c2 cells, accompanied by an increase in ER stress, apoptosis and activation of the mTOR signaling pathway, and a decrease in autophagy. The administration of larixyl acetate attenuated these impairments, which can be reversed by inhibiting autophagy through the activation of the mTOR signaling pathway. These findings suggested that larixyl acetate can effectively protect against pressure overload­induced heart failure by enhancing autophagy and limiting ER stress and apoptosis through inhibition of the mTOR pathway.


Subject(s)
Acetates , Aortic Valve Stenosis , Heart Failure , Naphthalenes , Mice , Animals , TRPC6 Cation Channel , Myocytes, Cardiac/metabolism , Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/metabolism , TOR Serine-Threonine Kinases/metabolism , Autophagy , Aortic Valve Stenosis/metabolism , Hypertrophy/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Apoptosis
9.
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
10.
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
11.
Mol Med Rep ; 29(1)2024 01.
Article in English | MEDLINE | ID: mdl-38038121

ABSTRACT

Diabetic kidney disease (DKD), one of the common complications of type­2 diabetes mellitus (T2DM), has become the principal cause of end­stage kidney disease. Transient receptor potential channel 6 (TRPC6), one of non­selective cation channels with significant calcium­permeability, is associated with renal fibrosis. However, the mechanism of TRPC6 in T2DM­induced renal fibrosis is still not entirely understood. The present study explored the potential mechanism of Trpc6 knockout in T2DM­induced renal fibrosis in Trpc6­/­ mice. The results showed that Trpc6 knockout inhibited the loss of body weight and the increase of fasting blood glucose (FBG) and significantly improved renal dysfunction and glomerular fibrosis in T2DM mice. The present study also indicated that Trpc6 knockout significantly lowered the expression of phosphorylated (p­)SMAD2/3, TGF­ß, calcineurin (CN), nuclear factor of activated T­cell (NFAT)2 and Nod­like receptor (NLR) 3 inflammasome­associated proteins. Calcium imaging results revealed that Trpc6 knockdown could decrease the levels of [Ca2+]i and inhibited calcium homeostasis imbalance. Moreover, it was found that knockout of Trpc6 had no significant influence on lipid disposition and reactive oxygen species generation in the kidney cortex. The present study suggested that knockout of Trpc6 may alleviate glomerular fibrosis and delay DKD progression by reducing [Ca2+]i overload and inhibiting the CN­NFAT2 pathway in T2DM mice.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Mice , Animals , TRPC6 Cation Channel/genetics , Calcineurin/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Calcium/metabolism , Diabetic Nephropathies/metabolism , Signal Transduction , Diabetes Mellitus, Type 2/complications , Fibrosis , Mice, Knockout
12.
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
13.
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
14.
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
15.
Cell Rep ; 42(11): 113347, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37910503

ABSTRACT

Understanding the cell biological mechanisms that enable tumor cells to persist after therapy is necessary to improve the treatment of recurrent disease. Here, we demonstrate that transient receptor potential channel 6 (TRPC6), a channel that mediates calcium entry, contributes to the properties of breast cancer stem cells, including resistance to chemotherapy, and that tumor cells that persist after therapy are dependent on TRPC6. The mechanism involves the ability of TRPC6 to regulate integrin α6 mRNA splicing. Specifically, TRPC6-mediated calcium entry represses the epithelial splicing factor ESRP1 (epithelial splicing regulatory protein 1), which enables expression of the integrin α6B splice variant. TRPC6 and α6B function in tandem to facilitate stemness and persistence by activating TAZ and, consequently, repressing Myc. Therapeutic inhibition of TRPC6 sensitizes triple-negative breast cancer (TNBC) cells and tumors to chemotherapy by targeting the splicing of α6 integrin mRNA and inducing Myc. These data reveal a Ca2+-dependent mechanism of chemotherapy-induced persistence, which is amenable to therapy, that involves integrin mRNA splicing.


Subject(s)
Antineoplastic Agents , Transient Receptor Potential Channels , Calcium Channels/metabolism , Integrin alpha6 , TRPC6 Cation Channel , Calcium/metabolism , TRPC Cation Channels/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Int J Mol Sci ; 24(22)2023 Nov 16.
Article in English | MEDLINE | ID: mdl-38003608

ABSTRACT

Excessive activation of the immune system is the cause of a wide variety of renal diseases. However, the pathogenic mechanisms underlying the aberrant activation of the immune system in the kidneys often remain unknown. TRPC6, a member of the Ca2+-permeant family of TRPC channels, is important in glomerular epithelial cells or podocytes for the process of glomerular filtration. In addition, TRPC6 plays a crucial role in the development of kidney injuries by inducing podocyte injury. However, an increasing number of studies suggest that TRPC6 is also responsible for tightly regulating the immune cell functions. It remains elusive whether the role of TRPC6 in the immune system and the pathogenesis of renal inflammation are intertwined. In this review, we present an overview of the current knowledge of how TRPC6 coordinates the immune cell functions and propose the hypothesis that TRPC6 might play a pivotal role in the development of kidney injury via its role in the immune system.


Subject(s)
Kidney Diseases , Podocytes , Humans , TRPC6 Cation Channel/genetics , TRPC Cation Channels/genetics , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Podocytes/pathology
17.
J Med Chem ; 66(22): 15061-15072, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37922400

ABSTRACT

Selective modulation of TRPC6 ion channels is a promising therapeutic approach for neurodegenerative diseases and depression. A significant advancement showcases the selective activation of TRPC6 through metalated type-B PPAP, termed PPAP53. This success stems from PPAP53's 1,3-diketone motif facilitating metal coordination. PPAP53 is water-soluble and as potent as hyperforin, the gold standard in this field. In contrast to type-A, type-B PPAPs offer advantages such as gram-scale synthesis, easy derivatization, and long-term stability. Our investigations reveal PPAP53 selectively binding to the C-terminus of TRPC6. Although cryoelectron microscopy has resolved the majority of the TRPC6 structure, the binding site in the C-terminus remained unresolved. To address this issue, we employed state-of-the-art artificial-intelligence-based protein structure prediction algorithms to predict the missing region. Our computational results, validated against experimental data, indicate that PPAP53 binds to the 777LLKL780-region of the C-terminus, thus providing critical insights into the binding mechanism of PPAP53.


Subject(s)
TRPC Cation Channels , Binding Sites , Cryoelectron Microscopy , TRPC Cation Channels/drug effects , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/drug effects , Phloroglucinol/pharmacology , Polycyclic Compounds/pharmacology
18.
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
19.
Int J Mol Sci ; 24(20)2023 Oct 21.
Article in English | MEDLINE | ID: mdl-37895105

ABSTRACT

The transient receptor potential cation channel, subfamily C, member 6 (TRPC6), has been believed to adjust the formation of an excitatory synapse. The positive regulation of TRPC6 engenders synapse enlargement and improved learning and memory in animal models. TRPC6 is involved in different synaptoprotective signaling pathways, including antagonism of N-methyl-D-aspartate receptor (NMDAR), activation of brain-derived neurotrophic factor (BDNF) and postsynaptic store-operated calcium entry. Positive regulation of TRPC6 channels has been repeatedly shown to be good for memory formation and storage. TRPC6 is mainly expressed in the hippocampus, particularly in the dentate granule cells, cornu Ammonis 3 (CA3) pyramidal cells and gamma-aminobutyric acid (GABA)ergic interneurons. It has been observed that TRPC6 agonists have a great influence on animal behavior including memory formation and storage The purpose of this review is to collect the available information on the role of TRPC6 in memory formation in various parts of the brain to understand how TRPC6-specific pharmaceutical agents will affect memory in distinct parts of the central nervous system (CNS).


Subject(s)
Neurons , TRPC Cation Channels , Animals , TRPC6 Cation Channel , TRPC Cation Channels/metabolism , Neurons/metabolism , Synapses/metabolism , Behavior, Animal , Calcium/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...