Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 11: 564, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32322252

RESUMO

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.


Assuntos
Sinalização do Cálcio/imunologia , Degranulação Celular/imunologia , Mastócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Canais de Cátion TRPC/deficiência , Animais , Células da Medula Óssea/imunologia , Masculino , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritônio/citologia , Peritônio/imunologia , Canais de Cátion TRPC/imunologia
2.
Biochem Biophys Res Commun ; 519(4): 674-681, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31543348

RESUMO

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.


Assuntos
Cálcio/metabolismo , Cardiomegalia/metabolismo , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Canais de Cátion TRPC/deficiência , Trifosfato de Adenosina/metabolismo , Animais , Fator Natriurético Atrial/metabolismo , Cardiomegalia/etiologia , Cardiomegalia/genética , Linhagem Celular , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Peptídeo Natriurético Encefálico/metabolismo , Ratos , Cloreto de Sódio na Dieta/efeitos adversos , Canais de Cátion TRPC/genética
3.
Cell Physiol Biochem ; 52(3): 455-467, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30873821

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/genética , Canais de Cátion TRPC/genética , Zinco/metabolismo , Adulto , Animais , Cátions Bivalentes , Proteínas de Ligação a DNA , Feminino , Expressão Gênica , Homeostase/genética , Humanos , Transporte de Íons , Rim/metabolismo , Tamanho da Ninhada de Vivíparos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/patologia , Gravidez , Canais de Cátion TRPC/deficiência , Canal de Cátion TRPC6 , Fatores de Transcrição
4.
Biochim Biophys Acta Mol Cell Res ; 1866(7): 1124-1136, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30452936

RESUMO

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.


Assuntos
Sinalização do Cálcio , Cálcio , Hormônio Liberador de Gonadotropina/metabolismo , Proteína ORAI1/deficiência , Adeno-Hipófise/metabolismo , Canais de Cátion TRPC/deficiência , Tireotropina/metabolismo , Animais , Hormônio Liberador de Gonadotropina/genética , Camundongos , Camundongos Knockout , Tireotropina/genética
5.
J Alzheimers Dis ; 63(2): 761-772, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29660945

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apoptose/fisiologia , Transtornos da Memória/metabolismo , Fragmentos de Peptídeos/metabolismo , Canais de Cátion TRPC/deficiência , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células HEK293 , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Transtornos da Memória/patologia , Camundongos da Linhagem 129 , Camundongos Knockout , Domínios Proteicos , Canais de Cátion TRPC/genética
6.
Atherosclerosis ; 270: 199-204, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29290366

RESUMO

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.


Assuntos
Aorta/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Macrófagos/metabolismo , Osteogênese , Placa Aterosclerótica , Canais de Cátion TRPC/deficiência , Calcificação Vascular/metabolismo , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Proteína Morfogenética Óssea 2/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Canais de Cátion TRPC/genética , Calcificação Vascular/genética , Calcificação Vascular/patologia , Calcificação Vascular/prevenção & controle
7.
Sci Rep ; 7(1): 7511, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28790356

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental/genética , Insuficiência Cardíaca/genética , Hiperglicemia/genética , NADPH Oxidase 2/genética , Canais de Cátion TRPC/genética , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Regulação da Expressão Gênica , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Hiperglicemia/induzido quimicamente , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Peróxidos Lipídicos/metabolismo , Camundongos , Camundongos Knockout , Contração Miocárdica , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , NADPH Oxidase 2/metabolismo , Cultura Primária de Células , Ligação Proteica , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Estreptozocina , Canais de Cátion TRPC/deficiência , Canal de Cátion TRPC6
8.
Oncogene ; 36(45): 6306-6314, 2017 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-28714964

RESUMO

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.


Assuntos
Neoplasias Cerebelares/metabolismo , Proteínas Hedgehog/metabolismo , Meduloblastoma/metabolismo , Proteínas Wnt/antagonistas & inibidores , Via de Sinalização Wnt , Anilidas/farmacologia , Animais , Linhagem Celular Tumoral , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Masculino , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Meduloblastoma/patologia , Camundongos , Camundongos Transgênicos , Piridinas/farmacologia , Distribuição Aleatória , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Canais de Cátion TRPC/deficiência , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Transfecção , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Alcaloides de Veratrum/farmacologia , Proteínas Wnt/metabolismo
9.
Sci Rep ; 7: 42526, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28186192

RESUMO

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.


Assuntos
Apoptose/genética , Macrófagos/metabolismo , Necrose/genética , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/metabolismo , Canais de Cátion TRPC/deficiência , Animais , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Biomarcadores , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Placa Aterosclerótica/patologia , Receptores de LDL/genética
10.
Sci Rep ; 7: 39867, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-28051144

RESUMO

In previous studies using mice with macrophage-specific loss of TRPC3 we found a significant, selective effect of TRPC3 on the biology of M1, or inflammatory macrophages. Whereas activation of some components of the unfolded protein response and the pro-apoptotic mediators CamkII and Stat1 was impaired in Trpc3-deficient M1 cells, gathering insight about other molecular signatures within macrophages that might be affected by Trpc3 expression requires an alternative approach. In the present study we conducted RNA-seq analysis to interrogate the transcriptome of M1 macrophages derived from mice with macrophage-specific loss of TRPC3 and their littermate controls. We identified 160 significantly differentially expressed genes between the two groups, of which 62 were upregulated and 98 downregulated in control vs. Trpc3-deficient M1 macrophages. Gene ontology analysis revealed enrichment in processes associated to cellular movement and lipid signaling, whereas the enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways included networks for calcium signaling and cell adhesion molecules, among others. This is the first deep transcriptomic analysis of macrophages in the context of Trpc3 deficiency and the data presented constitutes a unique resource to further explore functions of TRPC3 in macrophage biology.


Assuntos
Perfilação da Expressão Gênica , Macrófagos/metabolismo , Canais de Cátion TRPC/genética , Animais , Células da Medula Óssea/citologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/farmacologia , Regulação para Baixo , Macrófagos/citologia , Camundongos , Camundongos Knockout , Netrina-1/farmacologia , Canais de Cátion TRPC/deficiência , Transcriptoma , Regulação para Cima
11.
Mol Neurobiol ; 54(3): 1992-2002, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26910815

RESUMO

Transient receptor potential-canonical 1 (TRPC1) plays a crucial role in neuronal survival, nerve regeneration, and protects neurons from neurotoxic injury, but it is not reported whether or how TRPC1 may affect learning and memory. Here, we found that TRPC1 knockout did not significantly affect the spatial learning and memory ability when the mice were housed in standard cages (SC). Interestingly, after the mice were exposed to environmental enrichment (EE) for 4 weeks, TRPC1 knockout abolished the EE-induced spatial memory enhancement, LTP induction, and neurogenesis in hippocampal DG subset. By stereotaxic infusion of the recombinant adeno-associated viruses (rAAV)-TRPC1 into the hippocampal DG subsets bilaterally, we observed that the EE-associated neurogenesis, LTP induction and the cognitive enhancement were efficiently rescued in TRPC1 knockout mice. EE increased the phosphorylation levels of ERK, p38, and cyclic AMP response element-binding protein (CREB) in wild-type mice, whereas the activation of ERK and CREB was not seen in TRPC1 knockout mice, and the phosphorylation of p38 was same in EE-TRPC1-/- and WT-EE. Finally, EE increased TRPC1 expression and overexpression of TRPC1 increased neurogenesis and activated ERK/CREB pathway in the wild-type mice. These findings suggest that TRPC1 is indispensable for the EE-induced hippocampal neurogenesis and cognitive enhancement.


Assuntos
Cognição/fisiologia , Meio Ambiente , Hipocampo/metabolismo , Neurogênese/fisiologia , Canais de Cátion TRPC/biossíntese , Canais de Cátion TRPC/deficiência , Animais , Reação de Fuga/fisiologia , Hipocampo/citologia , Hipocampo/patologia , Abrigo para Animais , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Distribuição Aleatória
12.
Sci Rep ; 6: 37001, 2016 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-27833156

RESUMO

Reactive oxygen species (ROS) produced by NADPH oxidase 2 (Nox2) function as key mediators of mechanotransduction during both physiological adaptation to mechanical load and maladaptive remodeling of the heart. This is despite low levels of cardiac Nox2 expression. The mechanism underlying the transition from adaptation to maladaptation remains obscure, however. We demonstrate that transient receptor potential canonical 3 (TRPC3), a Ca2+-permeable channel, acts as a positive regulator of ROS (PRROS) in cardiomyocytes, and specifically regulates pressure overload-induced maladaptive cardiac remodeling in mice. TRPC3 physically interacts with Nox2 at specific C-terminal sites, thereby protecting Nox2 from proteasome-dependent degradation and amplifying Ca2+-dependent Nox2 activation through TRPC3-mediated background Ca2+ entry. Nox2 also stabilizes TRPC3 proteins to enhance TRPC3 channel activity. Expression of TRPC3 C-terminal polypeptide abolished TRPC3-regulated ROS production by disrupting TRPC3-Nox2 interaction, without affecting TRPC3-mediated Ca2+ influx. The novel TRPC3 function as a PRROS provides a mechanistic explanation for how diastolic Ca2+ influx specifically encodes signals to induce ROS-mediated maladaptive remodeling and offers new therapeutic possibilities.


Assuntos
Sinalização do Cálcio/fisiologia , Miócitos Cardíacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Canais de Cátion TRPC/fisiologia , Disfunção Ventricular Esquerda/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetulus , Grupo dos Citocromos b/metabolismo , Diástole , Ativação Enzimática , Células HEK293 , Humanos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2/metabolismo , NADPH Oxidases/biossíntese , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Mapeamento de Interação de Proteínas , Estabilidade Proteica , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Estresse Mecânico , Canais de Cátion TRPC/deficiência , Canal de Cátion TRPC6 , Remodelação Ventricular/fisiologia
13.
Eur Biophys J ; 45(7): 657-670, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27670661

RESUMO

The tumor environment contributes importantly to tumor cell behavior and cancer progression. Aside from biochemical constituents, physical factors of the environment also influence the tumor. Growing evidence suggests that mechanics [e.g., tumor (stroma) elasticity, tissue pressure] are critical players of cancer progression. Underlying mechanobiological mechanisms involve among others the regulation of focal adhesion molecules, cytoskeletal modifications, and mechanosensitive (MS) ion channels of cancer- and tumor-associated cells. After reviewing the current concepts of cancer mechanobiology, we will focus on the canonical transient receptor potential 1 (TRPC1) channel and its role in mechano-signaling in tumor-associated pancreatic stellate cells (PSCs). PSCs are key players of pancreatic fibrosis, especially in cases of pancreatic ductal adenocarcinoma (PDAC). PDAC is characterized by the formation of a dense fibrotic stroma (desmoplasia), primarily formed by activated PSCs. Desmoplasia contributes to high pancreatic tissue pressure, which in turn activates PSCs, thereby perpetuating matrix deposition. Here, we investigated the role of the putatively mechanosensitive TRPC1 channels in murine PSCs exposed to elevated ambient pressure. Pressurization leads to inhibition of mRNA expression of MS ion channels. Migration of PSCs representing a readout of their activation is enhanced in pressurized PSCs. Knockout of TRPC1 leads to an attenuated phenotype. While TRPC1-mediated calcium influx is increased in wild-type PSCs after pressure incubation, loss of TRPC1 abolishes this effect. Our findings provide mechanistic insight how pressure, an important factor of the PDAC environment, contributes to PSC activation. TRPC1-mediated activation could be a potential target to disrupt the positive feedback of PSC activation and PDAC progression.


Assuntos
Células Estreladas do Pâncreas/metabolismo , Pressão , Canais de Cátion TRPC/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Cálcio/metabolismo , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Masculino , Camundongos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Células Estreladas do Pâncreas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Canais de Cátion TRPC/deficiência , Canais de Cátion TRPC/genética
14.
PLoS One ; 11(3): e0150923, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26950212

RESUMO

Exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) can enhance hippocampal neurogenesis in adult mice. However, little is focused on the effects of ELF-EMFs on embryonic neurogenesis. Here, we studied the potential effects of ELF-EMFs on embryonic neural stem cells (eNSCs). We exposed eNSCs to ELF-EMF (50 Hz, 1 mT) for 1, 2, and 3 days with 4 hours per day. We found that eNSC proliferation and maintenance were significantly enhanced after ELF-EMF exposure in proliferation medium. ELF-EMF exposure increased the ratio of differentiated neurons and promoted the neurite outgrowth of eNSC-derived neurons without influencing astrocyes differentiation and the cell apoptosis. In addition, the expression of the proneural genes, NeuroD and Ngn1, which are crucial for neuronal differentiation and neurite outgrowth, was increased after ELF-EMF exposure. Moreover, the expression of transient receptor potential canonical 1 (TRPC1) was significantly up-regulated accompanied by increased the peak amplitude of intracellular calcium level induced by ELF-EMF. Furthermore, silencing TRPC1 expression eliminated the up-regulation of the proneural genes and the promotion of neuronal differentiation and neurite outgrowth induced by ELF-EMF. These results suggest that ELF-EMF exposure promotes the neuronal differentiation and neurite outgrowth of eNSCs via up-regulation the expression of TRPC1 and proneural genes (NeuroD and Ngn1). These findings also provide new insights in understanding the effects of ELF-EMF exposure on embryonic brain development.


Assuntos
Diferenciação Celular/efeitos da radiação , Campos Eletromagnéticos , Células-Tronco Embrionárias/citologia , Células-Tronco Neurais/citologia , Neuritos/efeitos da radiação , Canais de Cátion TRPC/genética , Regulação para Cima/efeitos da radiação , Animais , Encéfalo/embriologia , Encéfalo/efeitos da radiação , Proliferação de Células/efeitos da radiação , Células-Tronco Embrionárias/efeitos da radiação , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neurais/efeitos da radiação , Neuritos/metabolismo , RNA Interferente Pequeno/genética , Canais de Cátion TRPC/deficiência
15.
J Am Soc Nephrol ; 27(3): 848-62, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26160898

RESUMO

Podocytes are specialized, highly differentiated epithelial cells in the kidney glomerulus that are exposed to glomerular capillary pressure and possible increases in mechanical load. The proteins sensing mechanical forces in podocytes are unconfirmed, but the classic transient receptor potential channel 6 (TRPC6) interacting with the MEC-2 homolog podocin may form a mechanosensitive ion channel complex in podocytes. Here, we observed that podocytes respond to mechanical stimulation with increased intracellular calcium concentrations and increased inward cation currents. However, TRPC6-deficient podocytes responded in a manner similar to that of control podocytes, and mechanically induced currents were unaffected by genetic inactivation of TRPC1/3/6 or administration of the broad-range TRPC blocker SKF-96365. Instead, mechanically induced currents were significantly decreased by the specific P2X purinoceptor 4 (P2X4) blocker 5-BDBD. Moreover, mechanical P2X4 channel activation depended on cholesterol and podocin and was inhibited by stabilization of the actin cytoskeleton. Because P2X4 channels are not intrinsically mechanosensitive, we investigated whether podocytes release ATP upon mechanical stimulation using a fluorometric approach. Indeed, mechanically induced ATP release from podocytes was observed. Furthermore, 5-BDBD attenuated mechanically induced reorganization of the actin cytoskeleton. Altogether, our findings reveal a TRPC channel-independent role of P2X4 channels as mechanotransducers in podocytes.


Assuntos
Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Mecanotransdução Celular , Podócitos/metabolismo , Receptores Purinérgicos P2X4/fisiologia , Trifosfato de Adenosina/farmacologia , Animais , Benzodiazepinonas/farmacologia , Células Cultivadas , Colesterol/metabolismo , Citoesqueleto/ultraestrutura , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X4/genética , Receptores Purinérgicos P2X4/metabolismo , Estresse Mecânico , Canais de Cátion TRPC/deficiência , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6
16.
Am J Physiol Cell Physiol ; 307(6): C521-31, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25031020

RESUMO

Endoplasmic reticulum (ER) stress is a prominent mechanism of macrophage apoptosis in advanced atherosclerotic lesions. Recent studies from our laboratory showed that advanced atherosclerotic plaques in Apoe(-/-) mice with bone marrow deficiency of the calcium-permeable channel Transient Receptor Potential Canonical 3 (TRPC3) are characterized by reduced areas of necrosis and fewer apoptotic macrophages than animals transplanted with Trpc3(+/+) bone marrow. In vitro, proinflammatory M1 but not anti-inflammatory M2 macrophages derived from Trpc3(-/-)Apoe(-/-) animals exhibited reduced ER stress-induced apoptosis. However, whether this was due to a specific effect of TRPC3 deficiency on macrophage ER stress signaling remained to be determined. In the present work we used polarized macrophages derived from mice with macrophage-specific deficiency of TRPC3 to examine the expression level of ER stress markers and the activation status of some typical mediators of macrophage apoptosis. We found that the reduced susceptibility of TRPC3-deficient M1 macrophages to ER stress-induced apoptosis correlates with an impaired unfolded protein response (UPR), reduced mitochondrion-dependent apoptosis, and reduced activation of the proapoptotic molecules calmodulin-dependent protein kinase II and signal transducer and activator of transcription 1. Notably, none of these pathways was altered in TRPC3-deficient M2 macrophages. These findings show for the first time an obligatory requirement for a member of the TRPC family of cation channels in ER stress-induced apoptosis in macrophages, underscoring a rather selective role of the TRPC3 channel on mechanisms related to the UPR signaling in M1 macrophages.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Macrófagos/metabolismo , Canais de Cátion TRPC/deficiência , Resposta a Proteínas não Dobradas , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Diferenciação Celular , Células Cultivadas , Retículo Endoplasmático/patologia , Macrófagos/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Canais de Cátion TRPC/genética
17.
Handb Exp Pharmacol ; 222: 15-51, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24756701

RESUMO

The TRPC1 ion channel was the first mammalian TRP channel to be cloned. In humans, it is encoded by the TRPC1 gene located in chromosome 3. The protein is predicted to consist of six transmembrane segments with the N- and C-termini located in the cytoplasm. The extracellular loop connecting transmembrane segments 5 and 6 participates in the formation of the ionic pore region. Inside the cell, TRPC1 is present in the endoplasmic reticulum, plasma membrane, intracellular vesicles, and primary cilium, an antenna-like sensory organelle functioning as a signaling platform. In human and rodent tissues, it shows an almost ubiquitous expression. TRPC1 interacts with a diverse group of proteins including ion channel subunits, receptors, and cytosolic proteins to mediate its effect on Ca(2+) signaling. It primarily functions as a cation nonselective channel within pathways controlling Ca(2+) entry in response to cell surface receptor activation. Through these pathways, it affects basic cell functions, such as proliferation and survival, differentiation, secretion, and cell migration, as well as cell type-specific functions such as chemotropic turning of neuronal growth cones and myoblast fusion. The biological role of TRPC1 has been studied in genetically engineered mice where the Trpc1 gene has been experimentally ablated. Although these mice live to adulthood, they show defects in several organs and tissues, such as the cardiovascular, central nervous, skeletal and muscular, and immune systems. Genetic and functional studies have implicated TRPC1 in diabetic nephropathy, Parkinson's disease, Huntington's disease, Duchenne muscular dystrophy, cancer, seizures, and Darier-White skin disease.


Assuntos
Canais de Cátion TRPC/metabolismo , Animais , Permeabilidade da Membrana Celular , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Ativação do Canal Iônico , Potenciais da Membrana , Camundongos , Camundongos Knockout , Fenótipo , Conformação Proteica , Relação Estrutura-Atividade , Canais de Cátion TRPC/química , Canais de Cátion TRPC/deficiência , Canais de Cátion TRPC/genética
18.
PLoS One ; 7(5): e36923, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22666335

RESUMO

In addition to their well-defined roles in replenishing depleted endoplasmic reticulum (ER) Ca(2+) reserves, molecular components of the store-operated Ca(2+) entry pathway regulate breast cancer metastasis. A process implicated in cancer metastasis that describes the conversion to a more invasive phenotype is epithelial-mesenchymal transition (EMT). In this study we show that EGF-induced EMT in MDA-MB-468 breast cancer cells is associated with a reduction in agonist-stimulated and store-operated Ca(2+) influx, and that MDA-MB-468 cells prior to EMT induction have a high level of non-stimulated Ca(2+) influx. The potential roles for specific Ca(2+) channels in these pathways were assessed by siRNA-mediated silencing of ORAI1 and transient receptor potential canonical type 1 (TRPC1) channels in MDA-MB-468 breast cancer cells. Non-stimulated, agonist-stimulated and store-operated Ca(2+) influx were significantly inhibited with ORAI1 silencing. TRPC1 knockdown attenuated non-stimulated Ca(2+) influx in a manner dependent on Ca(2+) influx via ORAI1. TRPC1 silencing was also associated with reduced ERK1/2 phosphorylation and changes in the rate of Ca(2+) release from the ER associated with the inhibition of the sarco/endoplasmic reticulum Ca(2+)-ATPase (time to peak [Ca(2+)](CYT) = 188.7 ± 34.6 s (TRPC1 siRNA) versus 124.0 ± 9.5 s (non-targeting siRNA); P<0.05). These studies indicate that EMT in MDA-MB-468 breast cancer cells is associated with a pronounced remodeling of Ca(2+) influx, which may be due to altered ORAI1 and/or TRPC1 channel function. Our findings also suggest that TRPC1 channels in MDA-MB-468 cells contribute to ORAI1-mediated Ca(2+) influx in non-stimulated cells.


Assuntos
Neoplasias da Mama/patologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Agonistas Purinérgicos/farmacologia , Transporte Biológico/efeitos dos fármacos , Canais de Cálcio/deficiência , Canais de Cálcio/genética , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Linhagem Celular Tumoral , Inativação Gênica , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína ORAI1 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fase S/efeitos dos fármacos , Fase S/genética , Canais de Cátion TRPC/deficiência , Canais de Cátion TRPC/genética
19.
J Thromb Haemost ; 10(3): 419-29, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22176814

RESUMO

BACKGROUND: Platelet adhesion, activation and aggregation at sites of vascular injury are essential processes for primary hemostasis. Elevation of the intracellular Ca(2+) concentration is a central event in platelet activation but the underlying mechanisms are not fully understood. Store-operated calcium entry (SOCE) through Orai1 was shown to be the main Ca(2+) influx pathway in murine platelets, but there are additional non-store-operated Ca(2+) (non-SOC) and receptor operated Ca(2+) (ROC) channels expressed in the platelet plasma membrane. OBJECTIVE: Canonical transient receptor potential (TRPC) channel 6 is found both in human and murine platelets and has been proposed to mediate diacylglycerol (DAG) activated ROCE but also a role in the regulation of SOCE has been suggested. METHODS: To investigate the function of TRPC6 in platelet Ca(2+) signaling and activation, we analyzed platelets from mice deficient in TRPC6 using a wide range of in vitro and in vivo assays. RESULTS: In the mutant platelets, DAG activated Ca(2+) influx was found to be abolished. However, this did not significantly affect SOCE or agonist induced Ca(2+) responses. Platelet function in vitro and in vivo was also unaltered in the absence of TRPC6. CONCLUSION: Our results indicate that DAG activated ROCE is mediated exclusively by TRPC6 in murine platelets, but this Ca(2+) influx has no major functional relevance for hemostasis and thrombosis. Further, in contrast to previous suggestions, based on studies with human platelets, TRPC6 appears to play an insignificant role in the regulation of SOCE in murine platelets.


Assuntos
Plaquetas/metabolismo , Sinalização do Cálcio , Diglicerídeos/metabolismo , Ativação Plaquetária , Canais de Cátion TRPC/deficiência , Difosfato de Adenosina/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Proteína C-Reativa/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Cloretos , Modelos Animais de Doenças , Compostos Férricos , Regulação da Expressão Gênica , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína ORAI1 , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , RNA Mensageiro/metabolismo , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6 , Trombina/metabolismo , Trombose/sangue , Trombose/induzido quimicamente , Trombose/genética , Fatores de Tempo
20.
Gastroenterology ; 140(7): 2107-15, 2115.e1-4, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21354153

RESUMO

BACKGROUND & AIMS: Excessive Ca2+ influx mediates many cytotoxic processes, including those associated with autoimmune inflammatory diseases such as acute pancreatitis and Sjögren syndrome. Transient receptor potential (canonical) channel (TRPC) 3 is a major Ca2+ influx channel in pancreatic and salivary gland cells. We investigated whether genetic or pharmacologic inhibition of TRPC3 protects pancreas and salivary glands from Ca2+-dependent damage. METHODS: We developed a Ca2+-dependent model of cell damage for salivary gland acini. Acute pancreatitis was induced by injection of cerulein into wild-type and Trpc3-/- mice. Mice were also given the Trpc3-selective inhibitor pyrazole 3 (Pyr3). RESULTS: Salivary glands and pancreas of Trpc3-/- mice were protected from Ca2+-mediated cell toxicity. Analysis of Ca2+ signaling in wild-type and Trpc3-/- acini showed that Pyr3 is a highly specific inhibitor of Tprc3; it protected salivary glands and pancreas cells from Ca2+-mediated toxicity by inhibiting the Trpc3-mediated component of Ca2+ influx. CONCLUSIONS: TRPC3-mediated Ca2+ influx mediates damage to pancreas and salivary glands. Pharmacologic inhibition of TRPC3 with the highly selective TRPC3 inhibitor Pyr3 might be developed for treatment of patients with acute pancreatitis and Sjögren syndrome.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Pancreatite/tratamento farmacológico , Pirazóis/farmacologia , Doenças das Glândulas Salivares/tratamento farmacológico , Glândulas Salivares/efeitos dos fármacos , Canais de Cátion TRPC/antagonistas & inibidores , Doença Aguda , Animais , Ceruletídeo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Camundongos , Camundongos Knockout , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/metabolismo , Pancreatite/patologia , Doenças das Glândulas Salivares/genética , Doenças das Glândulas Salivares/metabolismo , Doenças das Glândulas Salivares/patologia , Glândulas Salivares/metabolismo , Glândulas Salivares/patologia , Índice de Gravidade de Doença , Canais de Cátion TRPC/deficiência , Canais de Cátion TRPC/genética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA