Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters










Publication year range
1.
Cell Calcium ; 92: 102305, 2020 12.
Article in English | MEDLINE | ID: mdl-33069962

ABSTRACT

CaV1.2 and transient receptor potential canonical channel 3 (TRPC3) are two proteins known to have important roles in pathological cardiac hypertrophy; however, such roles still remain unclear. A better understanding of these roles is important for furthering the clinical understanding of heart failure. We previously reported that Trpc3-knockout (KO) mice are resistant to pathologic hypertrophy and that their CaV1.2 protein expression is reduced. In this study, we aimed to examine the relationship between these two proteins and characterize their role in neonatal cardiomyocytes. We measured CaV1.2 expression in the hearts of wild-type (WT) and Trpc3-/- mice, and examined the effects of Trpc3 knockdown and overexpression in the rat cell line H9c2. We also compared the hypertrophic responses of neonatal cardiomyocytes cultured from Trpc3-/- mice to a representative hypertrophy-causing drug, isoproterenol (ISO), and measured the activity of nuclear factor of activated T cells 3 (NFAT3) in neonatal cardiomyocytes (NCMCs). We inhibited the L-type current with nifedipine, and measured the intracellular calcium concentration using Fura-2 with 1-oleoyl-2-acetyl-sn-glycerol (OAG)-induced Ba2+ influx. When using the Trpc3-mediated Ca2+ influx, both intracellular calcium concentration and calcium influx were reduced in Trpc3-KO myocytes. Not only was the expression of CaV1.2 greatly reduced in Trpc3-KO cardiac lysate, but the size of the CaV1.2 currents in NCMCs was also greatly reduced. When NCMCs were treated with Trpc3 siRNA, it was confirmed that the expression of CaV1.2 and the intracellular nuclear transfer activity of NFAT decreased. In H9c2 cells, the ISO activated- and verapamil inhibited- Ca2+ influxes were dramatically attenuated by Trpc3 siRNA treatment. In addition, it was confirmed that both the expression of CaV1.2 and the size of H9c2 cells were regulated according to the expression and activation level of TRPC3. We found that after stimulation with ISO, cell hypertrophy occurred in WT myocytes, while the increase in size of Trpc3-KO myocytes was greatly reduced. These results suggest that not only the cell hypertrophy process in neonatal cardiac myocytes and H9c2 cells were regulated according to the expression level of CaV1.2, but also that the expression level of CaV1.2 was regulated by TRPC3 through the activation of NFAT.


Subject(s)
Calcium Channels, L-Type/metabolism , Cardiomegaly/metabolism , Myocytes, Cardiac/metabolism , TRPC Cation Channels/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Isoproterenol , Mice, Knockout , Myocytes, Cardiac/pathology , NFATC Transcription Factors/metabolism , RNA, Small Interfering/metabolism , Rats , TRPC Cation Channels/deficiency
2.
Front Immunol ; 11: 564, 2020.
Article in English | MEDLINE | ID: mdl-32322252

ABSTRACT

Mast cells are a heterogeneous group of immune cells. The simplest and commonly accepted classification divides them in two groups according to their protease content. We have compared the action of diverse secretagogues on bone marrow derived (BMMC) and peritoneal (PMC) mast cells which represent classical models of mucosal and connective tissue type mast cells in mice. Whereas, antigen stimulation of the FcεRI receptors was similarly effective in triggering elevations of free intracellular Ca2+ concentration ([Ca2+]i) in both BMMC and PMC, robust [Ca2+]i rise following Endothelin-1 stimulation was observed only in a fraction of BMMC. Leukotriene C4 activating cysteinyl leukotriene type I receptors failed to evoke [Ca2+]i rise in either mast cell model. Stimulation of the recently identified target of many small-molecule drugs associated with systemic pseudo-allergic reactions, Mrgprb2, with compound 48/80, a mast cell activator with unknown receptor studied for many years, triggered Ca2+ oscillations in BMMC and robust [Ca2+]i rise in PMCs similarly to that evoked by FcεRI stimulation. [Ca2+]i rise in PMC could also be evoked by other Mrgprb2 agonists such as Tubocurarine, LL-37, and Substance P. The extent of [Ca2+]i rise correlated with mast cell degranulation. Expression analysis of TRPC channels as potential candidates mediating agonist evoked Ca2+ entry revealed the presence of transcripts of all members of the TRPC subfamily of TRP channels in PMCs. The amplitude and AUC of compound 48/80-evoked [Ca2+]i rise was reduced by ~20% in PMC from Trpc1/4/6-/- mice compared to Trpc1/4-/- littermatched control mice, whereas FcεRI-evoked [Ca2+]i rise was unaltered. Whole-cell patch clamp recordings showed that the reduction in compound 48/80-evoked [Ca2+]i rise in Trpc1/4/6-/- PMC was accompanied by a reduced amplitude of Compound 48/80-induced cation currents which exhibited typical features of TRPC currents. Together, this study demonstrates that PMC are an appropriate mast cell model to study mechanisms of Mrgprb2 receptor-mediated mast cell activation, and it reveals that TRPC channels contribute at least partially to Mrgprb2-mediated mast cellactivation but not following FcεRI stimulation. However, the channels conducting most of the Ca2+ entry in mast cells triggered by Mrgprb2 receptor stimulation remains to be identified.


Subject(s)
Calcium Signaling/immunology , Cell Degranulation/immunology , Mast Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , TRPC Cation Channels/deficiency , Animals , Bone Marrow Cells/immunology , Male , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneum/cytology , Peritoneum/immunology , TRPC Cation Channels/immunology
3.
Biochem Biophys Res Commun ; 519(4): 674-681, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31543348

ABSTRACT

Long-term high salt intake leads to cardiac hypertrophy, but the mechanism remains elusive. Transient receptor potential channel, canonical 3(TRPC3), located in mitochondria, regulates mitochondrial calcium and reactive oxygen species(ROS) production. Herein, we investigated whether TRPC3 participates in high salt-induced cardiac hypertrophy by impairing cardiac mitochondrial function. High salt treatment increased the expression of mitochondrial TRPC3 in cardiomyocytes, accompanied by enhanced mitochondrial calcium uptake and elevated ROS production. Inhibition of TRPC3 significantly reduced high salt-induced ROS generation, promoted ATP production by stimulating oxidative phosphorylation, and increased enzyme activity in mitochondria in cardiomyocytes. Additionally, TRPC3 deficiency inhibited high salt-induced cardiac hypertrophy in vivo. A long-term high salt diet increased cardiac mitochondrial TRPC3 expression, elevated expression of cardiac hypertrophic markers atrial natriuretic peptide (ANP),brain natriuretic peptide (BNP) and ß-myosin heavy chain (ß-MHC) and decreased ATP production and mitochondrial complex I and II enzyme activity in a TRPC3-dependent manner. TRPC3 deficiency antagonises high salt diet-mediated cardiac hypertrophy by ameliorating TRPC3-mediated cardiac mitochondrial dysfunction. TRPC3 may therefore represent a novel target for preventing high salt-induced cardiac damage.


Subject(s)
Calcium/metabolism , Cardiomegaly/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , TRPC Cation Channels/deficiency , Adenosine Triphosphate/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Cardiomegaly/etiology , Cardiomegaly/genetics , Cell Line , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Mice, Knockout , Myocytes, Cardiac/drug effects , Natriuretic Peptide, Brain/metabolism , Rats , Sodium Chloride, Dietary/adverse effects , TRPC Cation Channels/genetics
4.
Proc Natl Acad Sci U S A ; 116(30): 15236-15243, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285329

ABSTRACT

Dopamine neurons of the hypothalamic arcuate nucleus (ARC) tonically inhibit the release of the protein hormone prolactin from lactotropic cells in the anterior pituitary gland and thus play a central role in prolactin homeostasis of the body. Prolactin, in turn, orchestrates numerous important biological functions such as maternal behavior, reproduction, and sexual arousal. Here, we identify the canonical transient receptor potential channel Trpc5 as an essential requirement for normal function of dopamine ARC neurons and prolactin homeostasis. By analyzing female mice carrying targeted mutations in the Trpc5 gene including a conditional Trpc5 deletion, we show that Trpc5 is required for maintaining highly stereotyped infraslow membrane potential oscillations of dopamine ARC neurons. Trpc5 is also required for eliciting prolactin-evoked tonic plateau potentials in these neurons that are part of a regulatory feedback circuit. Trpc5 mutant females show severe prolactin deficiency or hypoprolactinemia that is associated with irregular reproductive cyclicity, gonadotropin imbalance, and impaired reproductive capabilities. These results reveal a previously unknown role for the cation channel Trpc5 in prolactin homeostasis of female mice and provide strategies to explore the genetic basis of reproductive disorders and other malfunctions associated with defective prolactin regulation in humans.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Dopaminergic Neurons/metabolism , Genetic Diseases, Inborn/genetics , Lactation Disorders/genetics , Prolactin/deficiency , Prolactin/genetics , TRPC Cation Channels/genetics , Animals , Arcuate Nucleus of Hypothalamus/pathology , Arousal/physiology , Dopaminergic Neurons/pathology , Feedback, Physiological , Female , Gene Expression Regulation , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Gonadotropins/blood , Gonadotropins/genetics , Homeostasis/genetics , Humans , Lactation Disorders/metabolism , Lactation Disorders/pathology , Membrane Potentials/physiology , Mice , Mutation , Prolactin/blood , Prolactin/metabolism , Reproduction/physiology , Signal Transduction , TRPC Cation Channels/deficiency
5.
J Cell Sci ; 132(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-31182642

ABSTRACT

Properties of adipocytes, including differentiation and adipokine secretion, are crucial factors in obesity-associated metabolic syndrome. Here, we provide evidence that Ca2+ influx in primary adipocytes, especially upon Ca2+ store depletion, plays an important role in adipocyte differentiation, functionality and subsequently metabolic regulation. The endogenous Ca2+ entry channel in both subcutaneous and visceral adipocytes was found to be dependent on TRPC1-STIM1, and blocking Ca2+ entry with SKF96365 or using TRPC1-/- knockdown adipocytes inhibited adipocyte differentiation. Additionally, TRPC1-/- mice have decreased organ weight, but increased adipose deposition and reduced serum adiponectin and leptin concentrations, without affecting total adipokine expression. Mechanistically, TRPC1-mediated Ca2+ entry regulated SNARE complex formation, and agonist-mediated secretion of adipokine-loaded vesicles was inhibited in TRPC1-/- adipose. These results suggest an unequivocal role of TRPC1 in adipocyte differentiation and adiponectin secretion, and that loss of TRPC1 disturbs metabolic homeostasis.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Calcium/metabolism , Cell Differentiation , SNARE Proteins/metabolism , TRPC Cation Channels/metabolism , Adipocytes/metabolism , Adipogenesis , Adiponectin/blood , Adiponectin/metabolism , Adiposity , Aging/metabolism , Animals , Male , Mice , Protein Isoforms/metabolism , Subcutaneous Fat/cytology , TRPC Cation Channels/deficiency
6.
Cell Physiol Biochem ; 52(3): 455-467, 2019.
Article in English | MEDLINE | ID: mdl-30873821

ABSTRACT

BACKGROUND/AIMS: Transient receptor potential canonical 6 (TRPC6) protein is a nonselective cation channel permitting the uptake of essential elements such as iron (Fe) and zinc (Zn). TRPC6 is found throughout the body with high expression levels in the placenta. However, its role in this organ is still to be determined. To further advance our understanding of the physiological relevance of TRPC6, we have studied the placental histology, pregnancy outcome and the Fe and Zn status of organs (placenta, brain, kidney, liver and lung) collected from TRPC6 deficient (TRPC6-/-) mice and sex and age-matched C57Bl6/J and B6129SF2/J mice. METHODS: Metal content was quantified by inductively coupled plasma-atomic emission spectrometry (ICP-AES). Quantitative reverse transcriptase PCR (qRT-PCR) and Western Blottings (WB) were performed to analyze the expression of placental markers and TRPC6. RESULTS: Our data show that TRPC6-/- mice displayed reduced litter sizes, structural changes of the placenta, along with altered mRNA levels of CD31 and Gcm1, two markers of placental development. Furthermore, immunoblots revealed elevated amounts of TRPC6 proteins in placentas from women diagnosed with preeclampsia, a common gestational disease. When compared to C57Bl6/J and B6129SF2/J, TRPC6-/- mice had elevated Zn levels in placenta, liver and kidney during embryonic development and postnatally, but not at adulthood. High amounts of Fe were found in the adult brain and liver of TRPC6-/- mice. The lung was however not affected by the deletion of TRPC6, indicating that this mouse strain developed organ and age-dependent perturbations in their Zn and Fe status. CONCLUSION: This work indicates that TRPC6 exerts critical pathophysiological functions in placenta, and provides further evidence for a role of this channel in the homeostasis of cations like Zn and Fe.


Subject(s)
Brain/metabolism , Iron/metabolism , Liver/metabolism , Placenta/metabolism , Pre-Eclampsia/genetics , TRPC Cation Channels/genetics , Zinc/metabolism , Adult , Animals , Cations, Divalent , DNA-Binding Proteins , Female , Gene Expression , Homeostasis/genetics , Humans , Ion Transport , Kidney/metabolism , Litter Size , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , TRPC Cation Channels/deficiency , TRPC6 Cation Channel , Transcription Factors
7.
Biochim Biophys Acta Mol Cell Res ; 1866(7): 1124-1136, 2019 07.
Article in English | MEDLINE | ID: mdl-30452936

ABSTRACT

Store operated Ca2+ entry (SOCE) is the most important Ca2+ entry pathway in non-excitable cells. However, SOCE can also play a pivotal role in excitable cells such as anterior pituitary (AP) cells. The AP gland contains five different cell types that release six major AP hormones controlling most of the entire endocrine system. AP hormone release is modulated by Ca2+ signals induced by different hypothalamic releasing hormones (HRHs) acting on specific receptors in AP cells. TRH and LHRH both induce Ca2+ release and Ca2+ entry in responsive cells while GHRH and CRH only induce Ca2+ entry. SOCE has been shown to contribute to Ca2+ responses induced by TRH and LHRH but no molecular evidence has been provided. Accordingly, we used AP cells isolated from mice devoid of Orai1 channels (noted as Orai1-/- or Orai1 KO mice) and mice lacking expression of all seven canonical TRP channels (TRPC) from TRPC1 to TRPC7 (noted as heptaTRPC KO mice) to investigate contribution of these putative channel proteins to SOCE and intracellular Ca2+ responses induced by HRHs. We found that thapsigargin-evoked SOCE is lost in AP cells from Orai1-/- mice but unaffected in cells from heptaTRPC KO mice. Conversely, while spontaneous intracellular Ca2+-oscillations related to electrical activity were not affected in the Orai1-/- mice, these responses were significantly reduced in heptaTRPC KO mice. We also found that Ca2+ entry induced by TRH and LHRH is decreased in AP cells isolated from Orai1-/-. In addition, Ca2+ responses to several HRHs, particularly TRH and GHRH, are decreased in the heptaTRPC KO mice. These results indicate that expression of Orai1, and not TRPC channel proteins, is necessary for thapsigargin-evoked SOCE and is required to support Ca2+ entry induced by TRH and LHRH in mouse AP cells. In contrast, TRPC channel proteins appear to contribute to spontaneous Ca2+-oscillations and Ca2+ responses induced by TRH and GHRH. We conclude that expression of Orai1 and TRPC channels proteins may play differential and significant roles in AP physiology and endocrine control.


Subject(s)
Calcium Signaling , Calcium , Gonadotropin-Releasing Hormone/metabolism , ORAI1 Protein/deficiency , Pituitary Gland, Anterior/metabolism , TRPC Cation Channels/deficiency , Thyrotropin/metabolism , Animals , Gonadotropin-Releasing Hormone/genetics , Mice , Mice, Knockout , Thyrotropin/genetics
8.
Sci Rep ; 8(1): 9264, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29915209

ABSTRACT

Acetylcholine contracts the bladder by binding to muscarinic M3 receptors on the detrusor, leading to Ca2+ influx via voltage-gated Ca2+ channels. The cellular mechanisms linking these events are poorly understood, but studies have suggested that activation of TRPC4 channels could be involved. The purpose of this study was to investigate if spontaneous and cholinergic-mediated contractions of the detrusor were impaired in TRPC4 deficient (TRPC4-/-) mice. Isometric tension recordings were made from strips of wild-type (WT) and TRPC4-/- detrusor. Spontaneous phasic detrusor contractions were significantly smaller in TRPC4-/- mice compared to wild-type, however no difference in response to exogenous application of 60 mM KCl was observed. Cholinergic responses, induced by electric-field stimulation (EFS), bath application of the cholinergic agonist carbachol, or the acetylcholinesterase inhibitor neostigmine were all significantly smaller in TRPC4-/- detrusor strips than wild-type. Surprisingly, the TRPC4/5 inhibitor ML204 reduced EFS and CCh-evoked contractions in TRPC4-/- detrusor strips. However, TRPC5 expression was up-regulated in these preparations and, in contrast to wild-type, EFS responses were reduced in amplitude by the TRPC5 channel inhibitor clemizole hydrochloride. This study demonstrates that TRPC4 channels are involved in spontaneous and cholinergic-mediated contractions of the murine detrusor. TRPC5 expression is up-regulated in TRPC4-/- detrusor strips, and may partially compensate for loss of TRPC4 channels.


Subject(s)
Muscle Contraction/physiology , Receptors, Muscarinic/metabolism , TRPC Cation Channels/deficiency , Urinary Bladder/physiology , Acetylcholine/metabolism , Animals , Carbachol/pharmacology , Electric Stimulation , Indoles/pharmacology , Mice , Muscle Contraction/drug effects , Piperidines/pharmacology , Potassium Chloride/pharmacology , TRPC Cation Channels/metabolism , Urinary Bladder/drug effects
9.
J Alzheimers Dis ; 63(2): 761-772, 2018.
Article in English | MEDLINE | ID: mdl-29660945

ABSTRACT

The transient receptor potential cation (TRPC) channels are widely expressed in nervous system but their functions remain largely unclear. Here, we found that TRPC1 deletion did not affect learning and memory in physiological conditions, while it aggravated learning and memory deficits induced by amyloid-ß (Aß), the major component of the senile plaques observed in the brains of Alzheimer's disease (AD). Further studies demonstrated that TRPC1 deletion did not affect cell apoptosis in physiological condition, but it exacerbated the Aß-induced cell death in mouse hippocampus. Moreover, the level of TRPC1 was decreased in AD cell and mouse models, and upregulation of TRPC1 decreased Aß levels with attenuation of apoptosis in the cells stably overexpressing amyloid-ß protein precursor (AßPP). Finally, the transmembrane domain of TRPC1 could bind to AßPP and thus decreased Aß production. These findings indicate that loss of TRPC1 exacerbates Aß-induced memory deficit and cell apoptosis, though it does not impair cognitive function or induce cell death in physiological conditions.


Subject(s)
Amyloid beta-Peptides/metabolism , Apoptosis/physiology , Memory Disorders/metabolism , Peptide Fragments/metabolism , TRPC Cation Channels/deficiency , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Learning/physiology , Male , Memory/physiology , Memory Disorders/pathology , Mice, 129 Strain , Mice, Knockout , Protein Domains , TRPC Cation Channels/genetics
10.
J Cardiovasc Transl Res ; 11(4): 339-345, 2018 08.
Article in English | MEDLINE | ID: mdl-29532428

ABSTRACT

Endothelial progenitor cells (EPCs) promote angiogenesis and play a pivotal role in endothelial repair and re-endothelialization after vascular injury. Transient receptor potential-canonical1 (TRPC1) has been recently implied to play important roles on EPC function. Here, we studied the role of TRPC1 in regulating EPC function in vivo and in vitro. EPCs were cultured from TRPC1-knockout mice and their controls. In vitro, TRPC1 knockout reduced EPC functional activities, including migration and tube formation. Additionally, calmodulin (CaM)/endothelial nitric oxide synthase (eNOS) signaling activity were downregulated after TRPC1 knockout. Administration of CaM or eNOS inhibitor ameliorated TRPC1 knockout-reduced EPC migration and tube formation. In vivo Matrigel plug assay confirmed that TRPC1 knockout decreased formation of functional blood vessels of EPCs compared with wild-type EPCs. Taken together, these data suggest that TRPC1 is a critical regulator of angiogenesis.


Subject(s)
Calmodulin/metabolism , Endothelial Progenitor Cells/metabolism , Neovascularization, Pathologic/metabolism , Nitric Oxide Synthase Type III/metabolism , TRPC Cation Channels/deficiency , Vascular System Injuries/metabolism , Animals , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Progenitor Cells/pathology , Male , Mice , Neovascularization, Pathologic/pathology , Signal Transduction , TRPC Cation Channels/metabolism , Vascular System Injuries/pathology
11.
Channels (Austin) ; 12(1): 119-125, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29560783

ABSTRACT

Store depletion has been shown to induce Ca2+ entry by Na+/Ca+ exchange (NCX) 1 reversal in proliferative vascular smooth muscle cells (VSMCs). The study objective was to investigate the role of transient receptor potential canonical (TRPC) channels in store depletion and NCX1 reversal in proliferative VSMCs. In cultured VSMCs, expressing TRPC1, TRPC4, and TRPC6, the removal of extracellular Na+ was followed by a significant increase of cytosolic Ca2+ concentration that was inhibited by KBR, a selective NCX1 inhibitor. TRPC1 knockdown significantly suppressed store-operated, channel-mediated Ca2+ entry, but TRPC4 knockdown and TRPC6 knockdown had no effect. Separate knockdown of TRPC1, TRPC4, or TRPC6 did not have a significant effect on thapsigargin-initiated Na+ increase in the peripheral regions with KBR treatment, but knockdown of both TRPC4 and TRPC6 did. Stromal interaction molecule (STIM)1 knockdown significantly reduced TRPC4 and TRPC6 binding. The results demonstrated that TRPC4-TRPC6 heteromultimerization linked Ca2+ store depletion and STIM1 accumulation with NCX reversal in proliferative VSMCs.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Sodium-Calcium Exchanger/metabolism , TRPC Cation Channels/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Myocytes, Smooth Muscle/drug effects , Rats , Sodium-Calcium Exchanger/antagonists & inhibitors , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/deficiency
12.
Atherosclerosis ; 270: 199-204, 2018 03.
Article in English | MEDLINE | ID: mdl-29290366

ABSTRACT

BACKGROUND AND AIMS: Recent in vitro studies have showed that in macrophages, deletion of the non-selective Ca2+-permeable channel TRPC3 impairs expression of the osteogenic protein BMP-2. The pathophysiological relevance of this effect in atherosclerotic plaque calcification remains to be determined. METHODS: We used Ldlr-/- mice with macrophage-specific loss of TRPC3 (MacTrpc3-/-/Ldlr-/-) to examine the effect of macrophage Trpc3 on plaque calcification and osteogenic features in advanced atherosclerosis. RESULTS: After 25 weeks on high fat diet, aortic root plaques in MacTrpc3-/-/Ldlr-/- mice showed reduced size, lipid and macrophage content compared to controls. Plaque calcification was decreased in MacTrpc3-/-/Ldlr-/- mice, and this was accompanied by marked reduction in BMP-2, Runx-2 and phospho-SMAD1/5 contents within macrophage-rich areas. Expression of Bmp-2 and Runx-2 was also reduced in bone marrow-derived macrophages from MacTrpc3-/-/Ldlr-/- mice. CONCLUSIONS: These findings show that, in advanced atherosclerosis, selective deletion of TRPC3 in macrophages favors plaque regression and impairs the activity of a novel macrophage-associated, BMP-2-dependent mechanism of calcification.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Macrophages/metabolism , Osteogenesis , Plaque, Atherosclerotic , TRPC Cation Channels/deficiency , Vascular Calcification/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Bone Morphogenetic Protein 2/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Diet, High-Fat , Disease Models, Animal , Female , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , TRPC Cation Channels/genetics , Vascular Calcification/genetics , Vascular Calcification/pathology , Vascular Calcification/prevention & control
13.
Cell Rep ; 21(11): 3079-3088, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241537

ABSTRACT

Sexual preference for the opposite sex is a fundamental behavior underlying reproductive success, but the neural mechanisms remain unclear. Here, we examined the role of dopamine signaling in the nucleus accumbens core (NAcc) in governing chemosensory-mediated preference for females in TrpC2-/- and wild-type male mice. TrpC2-/- males, deficient in VNO-mediated signaling, do not display mating or olfactory preference toward females. We found that, during social interaction with females, TrpC2-/- males do not show increased NAcc dopamine levels, observed in wild-type males. Optogenetic stimulation of VTA-NAcc dopaminergic neurons in TrpC2-/- males during exposure to a female promoted preference response to female pheromones and elevated copulatory behavior toward females. Additionally, we found that signaling through the D1 receptor in the NAcc is necessary for the olfactory preference for female-soiled bedding. Our study establishes a critical role for the mesolimbic dopaminergic system in governing pheromone-mediated responses and mate choice in male mice.


Subject(s)
Dopamine/metabolism , Nucleus Accumbens/physiology , Receptors, Dopamine D1/genetics , Sexual Behavior, Animal/physiology , TRPC Cation Channels/genetics , Vomeronasal Organ/physiology , Animals , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Female , Gene Expression Regulation , Male , Mice , Mice, Knockout , Nucleus Accumbens/cytology , Optogenetics , Receptors, Dopamine D1/metabolism , Sex Attractants/biosynthesis , Sex Attractants/metabolism , Signal Transduction , Smell/physiology , TRPC Cation Channels/deficiency , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology , Vomeronasal Organ/cytology
14.
Sci Rep ; 7(1): 7511, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790356

ABSTRACT

Excess production of reactive oxygen species (ROS) caused by hyperglycemia is a major risk factor for heart failure. We previously reported that transient receptor potential canonical 3 (TRPC3) channel mediates pressure overload-induced maladaptive cardiac fibrosis by forming stably functional complex with NADPH oxidase 2 (Nox2). Although TRPC3 has been long suggested to form hetero-multimer channels with TRPC6 and function as diacylglycerol-activated cation channels coordinately, the role of TRPC6 in heart is still obscure. We here demonstrated that deletion of TRPC6 had no impact on pressure overload-induced heart failure despite inhibiting interstitial fibrosis in mice. TRPC6-deficient mouse hearts 1 week after transverse aortic constriction showed comparable increases in fibrotic gene expressions and ROS production but promoted inductions of inflammatory cytokines, compared to wild type hearts. Treatment of TRPC6-deficient mice with streptozotocin caused severe reduction of cardiac contractility with enhancing urinary and cardiac lipid peroxide levels, compared to wild type and TRPC3-deficient mice. Knockdown of TRPC6, but not TRPC3, enhanced basal expression levels of cytokines in rat cardiomyocytes. TRPC6 could interact with Nox2, but the abundance of TRPC6 was inversely correlated with that of Nox2. These results strongly suggest that Nox2 destabilization through disrupting TRPC3-Nox2 complex underlies attenuation of hyperglycemia-induced heart failure by TRPC6.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Heart Failure/genetics , Hyperglycemia/genetics , NADPH Oxidase 2/genetics , TRPC Cation Channels/genetics , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Gene Expression Regulation , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Hyperglycemia/chemically induced , Hyperglycemia/complications , Hyperglycemia/metabolism , Lipid Peroxides/metabolism , Mice , Mice, Knockout , Myocardial Contraction , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NADPH Oxidase 2/metabolism , Primary Cell Culture , Protein Binding , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , Streptozocin , TRPC Cation Channels/deficiency , TRPC6 Cation Channel
15.
Oncogene ; 36(45): 6306-6314, 2017 11 09.
Article in English | MEDLINE | ID: mdl-28714964

ABSTRACT

The SMOOTHENED inhibitor vismodegib is FDA approved for advanced basal cell carcinoma (BCC), and shows promise in clinical trials for SONIC HEDGEHOG (SHH)-subgroup medulloblastoma (MB) patients. Clinical experience with BCC patients shows that continuous exposure to vismodegib is necessary to prevent tumor recurrence, suggesting the existence of a vismodegib-resistant reservoir of tumor-propagating cells. We isolated such tumor-propagating cells from a mouse model of SHH-subgroup MB and grew them as sphere cultures. These cultures were enriched for the MB progenitor marker SOX2 and formed tumors in vivo. Moreover, while their ability to self-renew was resistant to SHH inhibitors, as has been previously suggested, this self-renewal was instead WNT-dependent. We show here that loss of Trp53 activates canonical WNT signaling in these SOX2-enriched cultures. Importantly, a small molecule WNT inhibitor was able to reduce the propagation and growth of SHH-subgroup MB in vivo, in an on-target manner, leading to increased survival. Our results imply that the tumor-propagating cells driving the growth of bulk SHH-dependent MB are themselves WNT dependent. Further, our data suggest combination therapy with WNT and SHH inhibitors as a therapeutic strategy in patients with SHH-subgroup MB, in order to decrease the tumor recurrence commonly observed in patients treated with vismodegib.


Subject(s)
Cerebellar Neoplasms/metabolism , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway , Anilides/pharmacology , Animals , Cell Line, Tumor , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Disease Models, Animal , HEK293 Cells , Humans , Male , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Transgenic , Pyridines/pharmacology , Random Allocation , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Small Molecule Libraries/pharmacology , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Transfection , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Veratrum Alkaloids/pharmacology , Wnt Proteins/metabolism
16.
Sci Rep ; 7(1): 2338, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28539583

ABSTRACT

Transient receptor potential canonical 5 (TRPC5), a calcium-permeable, non-selective cation channel is expressed in the periphery, but there is limited knowledge of its regulatory roles in vivo. Endogenous modulators of TRPC5 include a range of phospholipids that have an established role in liver disease, including lysophosphatidylcholine (LPC). Cholestasis is characterized by impairment of excretion of bile acids, leading to elevation of hepatic bile acids. We investigated the contribution of TRPC5 in a murine model of cholestasis. Wild-type (WT) and TRPC5 knock-out (KO) mice were fed a diet supplemented with 0.5% cholic acid (CA) for 21 days. CA-diet supplementation resulted in enlargement of the liver in WT mice, which was ameliorated in TRPC5 KO mice. Hepatic bile acid and lipid content was elevated in WT mice, with a reduction observed in TRPC5 KO mice. Consistently, liver enzymes were significantly increased in cholestatic WT mice and significantly blunted in TRPC5 KO mice. Localized dyslipidaemia, secondary to cholestasis, was investigated utilizing a selected lipid analysis. This revealed significant perturbations in the lipid profile following CA-diet feeding, with increased cholesterol, triglycerides and phospholipids, in WT, but not TRPC5 KO mice. Our results suggest that activation of TRPC5 contributes to the development of cholestasis and associated dyslipidemia. Modulation of TRPC5 activity may present as a novel therapeutic target for liver disease.


Subject(s)
Cholestasis/metabolism , Dyslipidemias/metabolism , Liver/metabolism , TRPC Cation Channels/physiology , Animals , Bile Acids and Salts/blood , Bile Acids and Salts/metabolism , Cholestasis/genetics , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Dyslipidemias/genetics , Gene Expression , Lipids/analysis , Liver/pathology , Male , Mice, Inbred ICR , Mice, Knockout , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics
17.
Proc Natl Acad Sci U S A ; 114(23): E4582-E4591, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28526717

ABSTRACT

The injury phase after myocardial infarcts occurs during reperfusion and is a consequence of calcium release from internal stores combined with calcium entry, leading to cell death by apoptopic and necrotic processes. The mechanism(s) by which calcium enters cells has(ve) not been identified. Here, we identify canonical transient receptor potential channels (TRPC) 3 and 6 as the cation channels through which most of the damaging calcium enters cells to trigger their death, and we describe mechanisms activated during the injury phase. Working in vitro with H9c2 cardiomyoblasts subjected to 9-h hypoxia followed by 6-h reoxygenation (H/R), and analyzing changes occurring in areas-at-risk (AARs) of murine hearts subjected to a 30-min ischemia followed by 24-h reperfusion (I/R) protocol, we found: (i) that blocking TRPC with SKF96365 significantly ameliorated damage induced by H/R, including development of the mitochondrial permeability transition and proapoptotic changes in Bcl2/BAX ratios; and (ii) that AAR tissues had increased TUNEL+ cells, augmented Bcl2/BAX ratios, and increased p(S240)NFATc3, p(S473)AKT, p(S9)GSK3ß, and TRPC3 and -6 proteins, consistent with activation of a positive-feedback loop in which calcium entering through TRPCs activates calcineurin-mediated NFATc3-directed transcription of TRPC genes, leading to more Ca2+ entry. All these changes were markedly reduced in mice lacking TRPC3, -6, and -7. The changes caused by I/R in AAR tissues were matched by those seen after H/R in cardiomyoblasts in all aspects except for p-AKT and p-GSK3ß, which were decreased after H/R in cardiomyoblasts instead of increased. TRPC should be promising targets for pharmacologic intervention after cardiac infarcts.


Subject(s)
Cell Hypoxia/physiology , Myocardial Reperfusion Injury/etiology , TRPC Cation Channels/metabolism , Animals , Apoptosis , Calcium Channel Blockers/pharmacology , Calcium Signaling , Cell Hypoxia/drug effects , Cell Line , Disease Models, Animal , Imidazoles/pharmacology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Signal Transduction , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics , TRPC6 Cation Channel
18.
Sci Rep ; 7: 42526, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28186192

ABSTRACT

In previous work we reported that ApoeKO mice transplanted with bone marrow cells deficient in the Transient Receptor Potential Canonical 3 (TRPC3) channel have reduced necrosis and number of apoptotic macrophages in advanced atherosclerotic plaques. Also, in vitro studies with polarized macrophages derived from mice with macrophage-specific loss of TRPC3 showed that M1, but not M2 macrophages, deficient in Trpc3 are less susceptible to ER stress-induced apoptosis than Trpc3 expressing cells. The questions remained (a) whether the plaque phenotype in transplanted mice resulted from a genuine effect of Trpc3 on macrophages, and (b) whether the reduced necrosis and macrophage apoptosis in plaques of these mice was a manifestation of the selective effect of TRPC3 on apoptosis of M1 macrophages previously observed in vitro. Here, we addressed these questions using Ldlr knockout (Ldlr-/-) mice with macrophage-specific loss of Trpc3 (MacTrpc3-/-/Ldlr-/- → Ldlr-/-). Compared to controls, we observed decreased plaque necrosis and number of apoptotic macrophages in MacTrpc3-/-/Ldlr-/- → Ldlr-/- mice. Immunohistochemical analysis revealed a reduction in apoptotic M1, but not apoptotic M2 macrophages. These findings confirm an effect of TRPC3 on plaque necrosis and support the notion that this is likely a reflection of the reduced susceptibility of Trpc3-deficient M1 macrophages to apoptosis.


Subject(s)
Apoptosis/genetics , Macrophages/metabolism , Necrosis/genetics , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/metabolism , TRPC Cation Channels/deficiency , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers , Disease Models, Animal , Female , Immunohistochemistry , Mice , Mice, Knockout , Plaque, Atherosclerotic/pathology , Receptors, LDL/genetics
19.
Eur J Pharmacol ; 802: 27-35, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28237267

ABSTRACT

As a Ca2+-permeable cationic channel, canonical transient receptor potential 5 channel (TRPC5) has been known to be involved in various functions of cells. Although TRPC5 is abundantly expressed in vascular endothelium, it remains unknown whether TRPC5 plays a role in regulating endothelial senescence. In the present study, we investigated the involvement of TRPC5 in the senescence of mouse aortic endothelial cells (MAECs). Meanwhile, the regulatory role of TRPC5 in the endothelial dysfunction in aged mice was also studied. Results showed that the endothelium-dependent relaxations were significantly promoted but the endothelium-dependent contractions were markedly attenuated in aortic rings from aged mice with TRPC5 gene knocked out compared with those from aged C57BL/6J mice. Meanwhile, the nuclear telomerase activity and nitric oxide generation were notably enhanced but the reactive oxygen species production was significantly inhibited in aged mice or in hydrogen peroxide-induced senescent MAECs lacking the TRPC5 gene. In addition, by performing the senescence-associated ß-galactosidase staining the oxidative stress-induced premature senescence was markedly depressed in MAECs with TRPC5 gene deficient. Expressions of endothelial nitric oxide synthase and silent information regulator protein 1 (SIRT1) were significantly up-regulated but those of P53 and P21 were markedly down-regulated both in aged mice and in hydrogen peroxide-treated MAECs in the absence of the TRPC5 gene. These results of our study uncover novel functions of TRPC5 in regulating vascular aging.


Subject(s)
Cellular Senescence , Endothelial Cells/cytology , TRPC Cation Channels/metabolism , Aging/metabolism , Animals , Aorta, Thoracic/physiology , Endothelial Cells/metabolism , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics
20.
Cell Rep ; 18(3): 583-592, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099839

ABSTRACT

The molecular mechanisms underlying acute leptin and serotonin 2C receptor-induced hypophagia remain unclear. Here, we show that neuronal and pro-opiomelanocortin (Pomc)-specific loss of transient receptor potential cation 5 (TrpC5) subunits is sufficient to decrease energy expenditure and increase food intake resulting in elevated body weight. Deficiency of Trpc5 subunits in Pomc neurons is also sufficient to block the anorexigenic effects of leptin and serotonin 2C receptor (Ht2Cr) agonists. The loss of acute anorexigenic effects of these receptors is concomitant with a blunted electrophysiological response to both leptin and Ht2Cr agonists in arcuate Pomc neurons. We also demonstrate that the Ht2Cr agonist lorcaserin-induced improvements in glucose and insulin tolerance are blocked by TrpC5 deficiency in Pomc neurons. Together, our results link TrpC5 subunits in the brain with leptin- and serotonin 2C receptor-dependent changes in neuronal activity, as well as energy balance, feeding behavior, and glucose metabolism.


Subject(s)
Leptin/pharmacology , Neurons/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology , TRPC Cation Channels/genetics , Animals , Appetite Depressants/pharmacology , Benzazepines/pharmacology , Body Weight/drug effects , Eating/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Glucose Tolerance Test , Insulin Resistance , Male , Membrane Potentials/drug effects , Mice , Mice, Knockout , Neurons/metabolism , Patch-Clamp Techniques , Pro-Opiomelanocortin/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , TRPC Cation Channels/deficiency , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...