Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.090
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 374, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39210039

RESUMEN

Lysophosphatidylcholine (LPC) is a bioactive lipid present at high concentrations in inflamed and injured tissues where it contributes to the initiation and maintenance of pain. One of its important molecular effectors is the transient receptor potential canonical 5 (TRPC5), but the explicit mechanism of the activation is unknown. Using electrophysiology, mutagenesis and molecular dynamics simulations, we show that LPC-induced activation of TRPC5 is modulated by xanthine ligands and depolarizing voltage, and involves conserved residues within the lateral fenestration of the pore domain. Replacement of W577 with alanine (W577A) rendered the channel insensitive to strong depolarizing voltage, but LPC still activated this mutant at highly depolarizing potentials. Substitution of G606 located directly opposite position 577 with tryptophan rescued the sensitivity of W577A to depolarization. Molecular simulations showed that depolarization widens the lower gate of the channel and this conformational change is prevented by the W577A mutation or removal of resident lipids. We propose a gating scheme in which depolarizing voltage and lipid-pore helix interactions act together to promote TRPC5 channel opening.


Asunto(s)
Lisofosfatidilcolinas , Simulación de Dinámica Molecular , Canales Catiónicos TRPC , Humanos , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/química , Lisofosfatidilcolinas/metabolismo , Lisofosfatidilcolinas/farmacología , Animales , Activación del Canal Iónico/efectos de los fármacos , Células HEK293 , Mutación , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Potenciales de la Membrana/efectos de los fármacos
2.
Cancer Med ; 13(16): e70111, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39189437

RESUMEN

INTRODUCTION: Peritumoral brain edema (PTBE) has been widely reported with many brain tumors, especially with glioma. Since the blood-brain barrier (BBB) is essential for maintaining minimal permeability, any alteration in the interaction of BBB components, specifically in astrocytes and tight junctions (TJ), can result in disrupting the homeostasis of the BBB and making it severely leaky, which subsequently generates edema. OBJECTIVE: This study aimed to evaluate the functional gliovascular unit of the BBB by examining changes in the expression of claudin (CLDN) genes and the expression of transient receptor potential (TRP) membrane channels, additionally to define the correlation between their expressions. The evaluation was conducted using in vitro spheroid swelling models and tumor samples from glioma patients with PTBE. RESULTS: The results of the spheroid model showed that the genes TRPC3, TRPC4, TRPC5, and TRPV1 were upregulated in glioma cells either wild-type isocitrate dehydrogenase 1 (IDH1) or the IDH1 R132H mutant, with or without NaCl treatment. Furthermore, TRP genes appeared to adversely correlate with the up regulation of CLDN1, CLDN3, and CLDN5 genes. Besides, the upregulation of TRPC1 and TRPC4 in IDH1mt-R132H glioma cells. On the other hand, the correlation analysis revealed different correlations between different proteins in PTBE. CLDN1 exhibits a slight positive correlation with CLDN3. Similarly, TRPV1 displays a slight positive correlation with TRPC1. In contrast, TRPC4 shows a slight negative correlation with TRPC5. On the other hand, TRPC3 demonstrates a slight positive correlation with TRPC5, while the non-PTBE analysis highlights a moderate positive correlation between CLDN1 and TRPM4 while CLDN3 exhibits a moderate negative correlation with TRPC4. Additionally, CLDN5 demonstrates a slight negative correlation with TRPC4 but a moderate positive correlation with TRPC3. Furthermore, TRPC1 have a slight negative correlation with TRPV1, TRPC3 exhibiting a slight positive correlation with TRPC4, and TRPV1 showing a slight negative correlation with TRPC5. CONCLUSION: As a conclusion, the current study provided evidence of a slight negative correlation between TRPs and CLDN gene expression in PTBE patients and confirmatory results with some of the genes in cell model of edema.


Asunto(s)
Edema Encefálico , Neoplasias Encefálicas , Claudina-5 , Glioma , Humanos , Edema Encefálico/genética , Edema Encefálico/metabolismo , Edema Encefálico/patología , Glioma/genética , Glioma/metabolismo , Glioma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Claudina-5/genética , Claudina-5/metabolismo , Regulación Neoplásica de la Expresión Génica , Claudina-3/genética , Claudina-3/metabolismo , Barrera Hematoencefálica/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Línea Celular Tumoral , Claudina-1/genética , Claudina-1/metabolismo , Claudinas/genética , Claudinas/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Masculino
3.
J Biol Chem ; 300(9): 107705, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39178948

RESUMEN

The cell signaling molecules nitric oxide (NO) and Ca2+ regulate diverse biological processes through their closely coordinated activities directed by signaling protein complexes. However, it remains unclear how dynamically the multicomponent protein assemblies behave within the signaling complexes upon the interplay between NO and Ca2+ signals. Here we demonstrate that TRPC5 channels activated by the stimulation of G-protein-coupled ATP receptors mediate Ca2+ influx, that triggers NO production from endothelial NO synthase (eNOS), inducing secondary activation of TRPC5 via cysteine S-nitrosylation and eNOS in vascular endothelial cells. Mutations in the caveolin-1-binding domains of TRPC5 disrupt its association with caveolin-1 and impair Ca2+ influx and NO production, suggesting that caveolin-1 serves primarily as the scaffold for TRPC5 and eNOS to assemble into the signal complex. Interestingly, during ATP receptor activation, eNOS is dissociated from caveolin-1 and in turn directly associates with TRPC5, which accumulates at the plasma membrane dependently on Ca2+ influx and calmodulin. This protein reassembly likely results in a relief of eNOS from the inhibitory action of caveolin-1 and an enhanced TRPC5 S-nitrosylation by eNOS localized in the proximity, thereby facilitating the secondary activation of Ca2+ influx and NO production. In isolated rat aorta, vasodilation induced by acetylcholine was significantly suppressed by the TRPC5 inhibitor AC1903. Thus, our study provides evidence that dynamic remodeling of the protein assemblies among TRPC5, eNOS, caveolin-1, and calmodulin determines the ensemble of Ca2+ mobilization and NO production in vascular endothelial cells.


Asunto(s)
Calcio , Caveolina 1 , Óxido Nítrico Sintasa de Tipo III , Óxido Nítrico , Canales Catiónicos TRPC , Óxido Nítrico Sintasa de Tipo III/metabolismo , Caveolina 1/metabolismo , Caveolina 1/genética , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Animales , Óxido Nítrico/metabolismo , Humanos , Ratas , Calcio/metabolismo , Masculino , Señalización del Calcio/fisiología , Células Endoteliales/metabolismo , Transducción de Señal , Retroalimentación Fisiológica , Células HEK293
4.
Cell ; 187(16): 4176-4192.e17, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38959890

RESUMEN

Hypothalamic neural circuits regulate instinctive behaviors such as food seeking, the fight/flight response, socialization, and maternal care. Here, we identified microdeletions on chromosome Xq23 disrupting the brain-expressed transient receptor potential (TRP) channel 5 (TRPC5). This family of channels detects sensory stimuli and converts them into electrical signals interpretable by the brain. Male TRPC5 deletion carriers exhibited food seeking, obesity, anxiety, and autism, which were recapitulated in knockin male mice harboring a human loss-of-function TRPC5 mutation. Women carrying TRPC5 deletions had severe postpartum depression. As mothers, female knockin mice exhibited anhedonia and depression-like behavior with impaired care of offspring. Deletion of Trpc5 from oxytocin neurons in the hypothalamic paraventricular nucleus caused obesity in both sexes and postpartum depressive behavior in females, while Trpc5 overexpression in oxytocin neurons in knock-in mice reversed these phenotypes. We demonstrate that TRPC5 plays a pivotal role in mediating innate human behaviors fundamental to survival, including food seeking and maternal care.


Asunto(s)
Depresión Posparto , Neuronas , Obesidad , Canales Catiónicos TRPC , Animales , Femenino , Ratones , Obesidad/metabolismo , Obesidad/genética , Masculino , Humanos , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Depresión Posparto/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratones Endogámicos C57BL , Oxitocina/metabolismo , Conducta Materna
5.
Ren Fail ; 46(2): 2376929, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39022902

RESUMEN

The transient receptor potential canonical 6 (TRPC6) channel, a nonselective cation channel that allows the passage of Ca2+, plays an important role in renal diseases. TRPC6 is activated by Ca2+ influx, oxidative stress, and mechanical stress. Studies have shown that in addition to glomerular diseases, TRPC6 can contribute to renal tubular disorders, such as acute kidney injury, renal interstitial fibrosis, and renal cell carcinoma (RCC). However, the tubule-specific physiological functions of TRPC6 have not yet been elucidated. Its pathophysiological role in ischemia/reperfusion (I/R) injury is debatable. Thus, TRPC6 may have dual roles in I/R injury. TRPC6 induces renal fibrosis and immune cell infiltration in a unilateral ureteral obstruction (UUO) mouse model. Additionally, TRPC6 overexpression may modify G2 phase transition, thus altering the DNA damage checkpoint, which can cause genomic instability and RCC tumorigenesis and can control the proliferation of RCC cells. This review highlights the importance of TRPC6 in various conditions of the renal tubular system. To better understand certain renal disorders and ultimately identify new therapeutic targets to improve patient care, the pathophysiology of TRPC6 must be clarified.


Asunto(s)
Canal Catiónico TRPC6 , Humanos , Canal Catiónico TRPC6/metabolismo , Canal Catiónico TRPC6/genética , Animales , Túbulos Renales/patología , Túbulos Renales/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/etiología , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Daño por Reperfusión/metabolismo , Fibrosis , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Neoplasias Renales/genética , Ratones , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Estrés Oxidativo , Enfermedades Renales/metabolismo , Enfermedades Renales/etiología
6.
Biol Open ; 13(7)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38885005

RESUMEN

Metabolic syndrome has become a global epidemic, affecting all developed countries and communities with growing economies. Worldwide, increasing efforts have been directed at curbing this growing problem. Mice deleted of the gene encoding Type 1 Transient Receptor Potential Canonical Channel (Trpc1) were found to weigh heavier than controls. They had fasting hyperglycemia and impaired glucose tolerance compared with wild-type controls. Beyond 1 year of age, plasma triglyceride level in Trpc1-/- mice was elevated. Plasma cholesterol levels tended to be higher than in controls. The livers of Trpc1-/- mice were heavier, richer in triglyceride, and more echogenic than those of controls on ultrasound evaluation. Hematocrit was lower in Trpc1-/- mice of both genders beginning at the second to third months of age in the absence of bleeding or hemolysis. Measured by the indirect tail-cuff method or by the direct arterial cannulation, blood pressures in null mice were lower than controls. We conclude that TRPC1 gene regulates body metabolism and that except for hypertension, phenotypes of mice after deletion of the Trpc1 gene resemble mice with metabolic syndrome, suggesting that this could be a good experimental model for future investigation of the pathogenesis and management of this disorder.


Asunto(s)
Síndrome Metabólico , Canales Catiónicos TRPC , Animales , Femenino , Masculino , Ratones , Presión Sanguínea , Modelos Animales de Enfermedad , Síndrome Metabólico/etiología , Síndrome Metabólico/metabolismo , Síndrome Metabólico/genética , Ratones Noqueados , Fenotipo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/deficiencia
7.
Eur J Pharmacol ; 977: 176722, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38851562

RESUMEN

Transient receptor potential canonical 3 (TRPC3) channels are important in regulating Ca2+ homeostasis and have been implicated in the pathophysiology of chemically induced seizures. Inherited seizure susceptibility in genetically epilepsy-prone rats (GEPR-3s) has been linked to increased voltage-gated Ca2+ channel currents in the inferior colliculus neurons, which can affect intraneuronal Ca2+ homeostasis. However, whether TRPC3 channels also contribute to inherited seizure susceptibility in GEPR-3s is unclear. This study investigated the effects of JW-65, a potent and selective inhibitor of TRPC3 channels, on acoustically evoked seizure susceptibility in adult male and female GEPR-3s. These seizures consisted of wild running seizures (WRSs) that evolved into generalized tonic-clonic seizures (GTCSs). The results showed that acute administration of low doses of JW-65 significantly decreased by 55-89% the occurrence of WRSs and GTCSs and the seizure severity in both male and female GEPR-3s. This antiseizure effect was accompanied by increased seizure latency and decreased seizure duration. Additionally, female GEPR-3s were more responsive to JW-65's antiseizure effects than males. Moreover, JW-65 treatment for five consecutive days completely suppressed acoustically evoked seizures in male and female GEPR-3s. These findings suggest that inhibiting TRPC3 channels could be a promising antiseizure strategy targeting Ca2+ signaling mechanisms in inherited generalized tonic-clonic epilepsy.


Asunto(s)
Convulsiones , Canales Catiónicos TRPC , Animales , Masculino , Femenino , Convulsiones/fisiopatología , Convulsiones/genética , Convulsiones/inducido químicamente , Ratas , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Predisposición Genética a la Enfermedad , Epilepsia/fisiopatología , Epilepsia/genética , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico
8.
Zhen Ci Yan Jiu ; 49(6): 558-565, 2024 Jun 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38897799

RESUMEN

OBJECTIVES: To observe the effect of electroacupuncture (EA) at "Neiguan"(PC6) on cardiac function, cardiac morphology and transient receptor potential channel (TRPC) protein expressions in myocardial tissue of mice with myocardial hypertrophy, so as to explore its mechanisms underlying improvement of myocardial hypertrophy. METHODS: Forty-five male C57BL/6 mice were randomly divided into control, model and EA groups (15 mice/group). The myocardial hypertrophy model was established by subcutaneous injection of isoproterenol hydrochloride (15 mg·kg-1·d-1) for 14 days. The mice of the control group received subcutaneous injection of same amount of normal saline. The mice of the EA group received EA stimulation (frequency of 2 Hz, intensity of 1 mA) of bilateral PC6 for 20 min each time, once a day for 14 consecutive days. After the intervention, the body weight, tibia length and heart weight were measured. The left ventricular ejection fraction (EF), fractional shortening index (FS), left ventricular end-systolic volume (LVEV), left ventricular end-systolic internal diameter (LVID) and left ventricular posterior wall thickness (LVPW) were measured by using echocardiography for evaluating the cardiac function. The mean number and surface area of myocardial cells was detected by wheat germ agglutinin (WGA) staining, and changes of the cardiac morphology were observed under light microscopy after HE staining. The expression levels of TRPC1, TRPC3, TRPC4 and TRPC6 (TRPC1/3/4/6) in the myocardial tissue were detected by real-time quantitative PCR (qPCR) and Western blot, separately. RESULTS: Compared with the control group, the heart-body weight ratio(P<0.05) and heart-weight-to-tibia-length ratio (P<0.01), LVEV and LVID levels, the relative surface area, left ventricular area ratio, and the expression levels of cardiac TRPC1/3/4/6 were significantly increased (P<0.01, P<0.05), while the EF, FS, LVPW, number of cardiomyocytes, and the left ventricular posterior wall ratio were obviously decreased (P<0.01, P<0.05) in the model group. In comparison with the model group, the heart/body weight ratio, heart-weight-to-tibia-length ratio, LVEV and LVID levels, relative surface area, left ventricular area ratio, and the expression levels of cardiac TRPC1/3/4/6 were significantly decreased (P<0.01, P<0.05), while the EF, FS, LVPW, number of cardiomyocytes and left ventricular posterior wall ratio were significantly increased (P<0.01, P<0.05) in the EA group. H.E. staining showed disordered arrangement of cardiomyocytes and obvious myocardial interstitial inflammatory cell infiltration in the model group, and evident reduction of degree of cardiac fibrosis and interstitial edema in the EA group. CONCLUSIONS: EA of PC6 can improve the cardiac function and cardiac morphology in mice with myocardial hypertrophy, which may be related to its functions in down-regulating the expression of transient receptor potential channels.


Asunto(s)
Electroacupuntura , Ratones Endogámicos C57BL , Miocardio , Animales , Ratones , Masculino , Humanos , Miocardio/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Cardiomegalia/metabolismo , Cardiomegalia/terapia , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Puntos de Acupuntura , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética
9.
Nat Commun ; 15(1): 5220, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890374

RESUMEN

The transient receptor potential canonical type 3 (TRPC3) channel plays a pivotal role in regulating neuronal excitability in the brain via its constitutive activity. The channel is intricately regulated by lipids and has previously been demonstrated to be positively modulated by PIP2. Using molecular dynamics simulations and patch clamp techniques, we reveal that PIP2 predominantly interacts with TRPC3 at the L3 lipid binding site, located at the intersection of pre-S1 and S1 helices. We demonstrate that PIP2 sensing involves a multistep mechanism that propagates from L3 to the pore domain via a salt bridge between the TRP helix and S4-S5 linker. Notably, we find that both stimulated and constitutive TRPC3 activity require PIP2. These structural insights into the function of TRPC3 are invaluable for understanding the role of the TRPC subfamily in health and disease, in particular for cardiovascular diseases, in which TRPC3 channels play a major role.


Asunto(s)
Simulación de Dinámica Molecular , Fosfatidilinositol 4,5-Difosfato , Canales Catiónicos TRPC , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/química , Canales Catiónicos TRPC/genética , Humanos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Células HEK293 , Sitios de Unión , Animales , Técnicas de Placa-Clamp , Unión Proteica
10.
Int J Mol Sci ; 25(11)2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38892448

RESUMEN

Canonical transient receptor potential channel 3 (TRPC3) is the most abundant TRPC channel in the brain and is highly expressed in all subfields of the hippocampus. Previous studies have suggested that TRPC3 channels may be involved in the hyperexcitability of hippocampal pyramidal neurons and seizures. Genetic ablation of TRPC3 channel expression reduced the intensity of pilocarpine-induced status epilepticus (SE). However, the underlying cellular mechanisms remain unexplored and the contribution of TRPC3 channels to SE-induced neurodegeneration is not determined. In this study, we investigated the contribution of TRPC3 channels to the electrophysiological properties of hippocampal pyramidal neurons and hippocampal synaptic plasticity, and the contribution of TRPC3 channels to seizure-induced neuronal cell death. We found that genetic ablation of TRPC3 expression did not alter basic electrophysiological properties of hippocampal pyramidal neurons and had a complex impact on epileptiform bursting in CA3. However, TRPC3 channels contribute significantly to long-term potentiation in CA1 and SE-induced neurodegeneration. Our results provided further support for therapeutic potential of TRPC3 inhibitors and raised new questions that need to be answered by future studies.


Asunto(s)
Muerte Celular , Hipocampo , Células Piramidales , Convulsiones , Canales Catiónicos TRPC , Animales , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Ratones , Células Piramidales/metabolismo , Células Piramidales/patología , Hipocampo/metabolismo , Hipocampo/patología , Convulsiones/metabolismo , Convulsiones/patología , Estado Epiléptico/metabolismo , Estado Epiléptico/patología , Estado Epiléptico/inducido químicamente , Masculino , Neuronas/metabolismo , Pilocarpina , Potenciación a Largo Plazo , Ratones Noqueados , Ratones Endogámicos C57BL , Plasticidad Neuronal
11.
Neuromolecular Med ; 26(1): 19, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38703217

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder associated with mitochondrial dysfunctions and oxidative stress. However, to date, therapeutics targeting these pathological events have not managed to translate from bench to bedside for clinical use. One of the major reasons for the lack of translational success has been the use of classical model systems that do not replicate the disease pathology and progression with the same degree of robustness. Therefore, we employed a more physiologically relevant model involving alpha-synuclein-preformed fibrils (PFF) exposure to SH-SY5Y cells and Sprague Dawley rats. We further explored the possible involvement of transient receptor potential canonical 5 (TRPC5) channels in PD-like pathology induced by these alpha-synuclein-preformed fibrils with emphasis on amelioration of oxidative stress and mitochondrial health. We observed that alpha-synuclein PFF exposure produced neurobehavioural deficits that were positively ameliorated after treatment with the TRPC5 inhibitor clemizole. Furthermore, Clemizole also reduced p-alpha-synuclein and diminished oxidative stress levels which resulted in overall improvements in mitochondrial biogenesis and functions. Finally, the results of the pharmacological modulation were further validated using siRNA-mediated knockdown of TRPC5 channels, which also decreased p-alpha-synuclein expression. Together, the results of this study could be superimposed in the future for exploring the beneficial effects of TRPC5 channel modulation for other neurodegenerative disorders and synucleopathies.


Asunto(s)
Mitocondrias , Estrés Oxidativo , Ratas Sprague-Dawley , Canales Catiónicos TRPC , alfa-Sinucleína , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Animales , Ratas , Estrés Oxidativo/efectos de los fármacos , Humanos , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Línea Celular Tumoral , Masculino , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/inducido químicamente , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/tratamiento farmacológico
12.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38732005

RESUMEN

In calcium nephrolithiasis (CaNL), most calcium kidney stones are identified as calcium oxalate (CaOx) with variable amounts of calcium phosphate (CaP), where CaP is found as the core component. The nucleation of CaP could be the first step of CaP+CaOx (mixed) stone formation. High urinary supersaturation of CaP due to hypercalciuria and an elevated urine pH have been described as the two main factors in the nucleation of CaP crystals. Our previous in vivo findings (in mice) show that transient receptor potential canonical type 3 (TRPC3)-mediated Ca2+ entry triggers a transepithelial Ca2+ flux to regulate proximal tubular (PT) luminal [Ca2+], and TRPC3-knockout (KO; -/-) mice exhibited moderate hypercalciuria and microcrystal formation at the loop of Henle (LOH). Therefore, we utilized TRPC3 KO mice and exposed them to both hypercalciuric [2% calcium gluconate (CaG) treatment] and alkalineuric conditions [0.08% acetazolamide (ACZ) treatment] to generate a CaNL phenotype. Our results revealed a significant CaP and mixed crystal formation in those treated KO mice (KOT) compared to their WT counterparts (WTT). Importantly, prolonged exposure to CaG and ACZ resulted in a further increase in crystal size for both treated groups (WTT and KOT), but the KOT mice crystal sizes were markedly larger. Moreover, kidney tissue sections of the KOT mice displayed a greater CaP and mixed microcrystal formation than the kidney sections of the WTT group, specifically in the outer and inner medullary and calyceal region; thus, a higher degree of calcifications and mixed calcium lithiasis in the kidneys of the KOT group was displayed. In our effort to find the Ca2+ signaling pathophysiology of PT cells, we found that PT cells from both treated groups (WTT and KOT) elicited a larger Ca2+ entry compared to the WT counterparts because of significant inhibition by the store-operated Ca2+ entry (SOCE) inhibitor, Pyr6. In the presence of both SOCE (Pyr6) and ROCE (receptor-operated Ca2+ entry) inhibitors (Pyr10), Ca2+ entry by WTT cells was moderately inhibited, suggesting that the Ca2+ and pH levels exerted sensitivity changes in response to ROCE and SOCE. An assessment of the gene expression profiles in the PT cells of WTT and KOT mice revealed a safeguarding effect of TRPC3 against detrimental processes (calcification, fibrosis, inflammation, and apoptosis) in the presence of higher pH and hypercalciuric conditions in mice. Together, these findings show that compromise in both the ROCE and SOCE mechanisms in the absence of TRPC3 under hypercalciuric plus higher tubular pH conditions results in higher CaP and mixed crystal formation and that TRPC3 is protective against those adverse effects.


Asunto(s)
Oxalato de Calcio , Hipercalciuria , Cálculos Renales , Ratones Noqueados , Animales , Hipercalciuria/metabolismo , Hipercalciuria/genética , Concentración de Iones de Hidrógeno , Ratones , Oxalato de Calcio/metabolismo , Cálculos Renales/metabolismo , Cálculos Renales/etiología , Cálculos Renales/patología , Fosfatos de Calcio/metabolismo , Nefrolitiasis/metabolismo , Nefrolitiasis/genética , Nefrolitiasis/patología , Calcio/metabolismo , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Acetazolamida/farmacología
13.
Med Sci Monit ; 30: e942667, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38771735

RESUMEN

BACKGROUND Obstructive sleep apnea-hypopnea syndrome (OSAHS) presents a significant health concern, particularly among individuals with essential hypertension (EH). Understanding the genetic underpinnings of this association is crucial for effective management and intervention. We investigated the relationship between TRPC3 gene polymorphisms and susceptibility to OSAHS in patients with EH. MATERIAL AND METHODS We enrolled 373 patients with EH hospitalized at the First Affiliated Hospital of Xinjiang Medical University between April 2015 and November 2017. Patients were categorized into EH (n=74) and EH+OSAHS (n=299) groups according to the apnea-hypopnea index. Sequenom detection technology was used for TRPC3 gene single-nucleotide polymorphism genotyping, including genotypes at rs953691, rs10518289, rs2292232, rs4995894, rs951974, and rs4292355. RESULTS Sex, smoking history, alcohol history, hypertension duration, fasting blood glucose, urea, creatinine, total cholesterol, HDL-C, LDL-C, glycosylated hemoglobin, 24-h mean systolic BP, and 24-h mean diastolic BP were not significantly different between the 2 groups (P>0.05); however, age, BMI, triglyceride levels differed significantly (P<0.05). No significant difference was detected in distribution frequency of polymorphisms of TRPC3 gene between the 2 groups (P>0.05), while genotype, dominant genotype, and recessive genotype at rs10518289 and alleles at rs4292355 differed significantly (P<0.05). Logistic regression analysis showed age, BMI, and CG+GG genotypes at rs10518289 were risk factors for OSAHS in patients with EH. Interaction between TRPC3 (rs10518289) and obesity was not a risk of OSAHS with EH (P>0.05). CONCLUSIONS CC genotype of rs10518289 in the TRPC3 gene could be a protective genetic marker of OSAHS, and CG+GG genotype may be a risk genetic marker of OSAHS with EH.


Asunto(s)
Predisposición Genética a la Enfermedad , Genotipo , Hipertensión , Polimorfismo de Nucleótido Simple , Apnea Obstructiva del Sueño , Canales Catiónicos TRPC , Humanos , Femenino , Masculino , Persona de Mediana Edad , Apnea Obstructiva del Sueño/genética , Polimorfismo de Nucleótido Simple/genética , Hipertensión/genética , Canales Catiónicos TRPC/genética , Anciano , China , Factores de Riesgo , Adulto , Alelos , Hipertensión Esencial/genética
14.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 284-288, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38814201

RESUMEN

Podocyte injury plays a vital role in focal segmental glomerulosclerosis (FSGS), and apoptosis is one of its mechanisms. The transient receptor potential channel 6 (TRPC6) is highly expressed in podocytes and mutations mediate podocyte injury. We found TRPC6 gene mutation (N110S) was a new mutation and pathogenic in the preliminary clinical work. The purpose of this study was to investigate the potential mechanism of mutation in TRPC6 (TRPC6-N110S) in the knock-in gene mouse model and in immortalized mouse podocytes (MPC5). Transmission electron microscopy was used to evaluate renal injury morphology. We measured 24-hour urinary albumin-to-creatinine ratios and major biochemical parameters such as serum albumin, urea nitrogen, and total cholesterol. The results of CCK-8 assay and apoptosis experiments showed that the TRPC6-N110S overexpression group had slower proliferative activity and increased apoptosis than the control group. FluO-3 assay revealed increased calcium influx in the TRPC6-N110S overexpression group. Podocin level was decreased in TRPC6-N110S group, while TRPC6 and desmin levels were increased in TRPC6-N110S group. The 24 h uACR at 6 weeks was significantly higher in the pure-zygotes group than in the WT and heterozygotes groups, and this difference was found at 8 and 10 weeks.TRPC6 levels showed no significant difference between homozygote and WT mice. Compared to homozygote group, expression of podocin and nephrin were increased in WT, but levels of desmin was decreased in WT. Our results suggest that this new mutation causes podocyte injury probably by enhancing calcium influx and podocyte apoptosis, accompanied by increased proteinuria and decreased expression of nephrin and podocin.


Asunto(s)
Apoptosis , Mutación con Ganancia de Función , Podocitos , Canal Catiónico TRPC6 , Podocitos/metabolismo , Podocitos/patología , Animales , Canal Catiónico TRPC6/genética , Canal Catiónico TRPC6/metabolismo , Apoptosis/genética , Ratones , Mutación con Ganancia de Función/genética , Calcio/metabolismo , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Desmina/genética , Desmina/metabolismo , Proliferación Celular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Masculino , Ratones Endogámicos C57BL
15.
Nucleic Acids Res ; 52(9): 4784-4798, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38621757

RESUMEN

Antisense oligonucleotide (ASO) therapy is a novel therapeutic approach in which ASO specifically binds target mRNA, resulting in mRNA degradation; however, cellular uptake of ASOs remains critically low, warranting improvement. Transient receptor potential canonical (TRPC) channels regulate Ca2+ influx and are activated upon stimulation by phospholipase C-generated diacylglycerol. Herein, we report that a novel TRPC3/C6/C7 activator, L687, can induce cellular ASO uptake. L687-induced ASO uptake was enhanced in a dose- and incubation-time-dependent manner. L687 enhanced the knockdown activity of various ASOs both in vitro and in vivo. Notably, suppression of TRPC3/C6 by specific siRNAs reduced ASO uptake in A549 cells. Application of BAPTA-AM, a Ca2+ chelator, and SKF96365, a TRPC3/C6 inhibitor, suppressed Ca2+ influx via TRPC3/C6, resulting in reduced ASO uptake, thereby suggesting that Ca2+ influx via TRPC3/C6 is critical for L687-mediated increased ASO uptake. L687 also induced dextran uptake, indicating that L687 increased endocytosis. Adding ASO to L687 resulted in endosome accumulation; however, the endosomal membrane disruptor UNC7938 facilitated endosomal escape and enhanced knockdown activity. We discovered a new function for TRPC activators regarding ASO trafficking in target cells. Our findings provide an opportunity to formulate an innovative drug delivery system for the therapeutic development of ASO.


Asunto(s)
Calcio , Oligonucleótidos Antisentido , Canales Catiónicos TRPC , Humanos , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/metabolismo , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/antagonistas & inhibidores , Calcio/metabolismo , Células A549 , Animales , Ratones , Imidazoles/farmacología , Canal Catiónico TRPC6/metabolismo , Canal Catiónico TRPC6/genética , Canal Catiónico TRPC6/antagonistas & inhibidores , Ácido Egtácico/farmacología , Ácido Egtácico/análogos & derivados , Endosomas/metabolismo , Endosomas/efectos de los fármacos , Línea Celular Tumoral
16.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673977

RESUMEN

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.


Asunto(s)
Regulación hacia Abajo , Hiperparatiroidismo Primario , Glándulas Paratiroides , Canales Catiónicos TRPC , Canal Catiónico TRPC6 , Humanos , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Hiperparatiroidismo Primario/metabolismo , Hiperparatiroidismo Primario/genética , Hiperparatiroidismo Primario/patología , Glándulas Paratiroides/metabolismo , Glándulas Paratiroides/patología , Femenino , Masculino , Canal Catiónico TRPC6/metabolismo , Canal Catiónico TRPC6/genética , Persona de Mediana Edad , Anciano , Adulto , Inmunohistoquímica , Hormona Paratiroidea/metabolismo
17.
Biomolecules ; 14(4)2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38672459

RESUMEN

TRPC5 is a non-selective cation channel that is expressed in cardiomyocytes, but there is a lack of knowledge of its (patho)physiological role in vivo. Here, we examine the role of TRPC5 on cardiac function under basal conditions and during cardiac hypertrophy. Cardiovascular parameters were assessed in wild-type (WT) and global TRPC5 knockout (KO) mice. Despite no difference in blood pressure or activity, heart rate was significantly reduced in TRPC5 KO mice. Echocardiography imaging revealed an increase in stroke volume, but cardiac contractility was unaffected. The reduced heart rate persisted in isolated TRPC5 KO hearts, suggesting changes in basal cardiac pacing. Heart rate was further investigated by evaluating the reflex change following drug-induced pressure changes. The reflex bradycardic response following phenylephrine was greater in TRPC5 KO mice but the tachycardic response to SNP was unchanged, indicating an enhancement in the parasympathetic control of the heart rate. Moreover, the reduction in heart rate to carbachol was greater in isolated TRPC5 KO hearts. To evaluate the role of TRPC5 in cardiac pathology, mice were subjected to abdominal aortic banding (AAB). An exaggerated cardiac hypertrophy response to AAB was observed in TRPC5 KO mice, with an increased expression of hypertrophy markers, fibrosis, reactive oxygen species, and angiogenesis. This study provides novel evidence for a direct effect of TRPC5 on cardiac function. We propose that (1) TRPC5 is required for maintaining heart rate by regulating basal cardiac pacing and in response to pressure lowering, and (2) TRPC5 protects against pathological cardiac hypertrophy.


Asunto(s)
Cardiomegalia , Frecuencia Cardíaca , Ratones Noqueados , Canales Catiónicos TRPC , Animales , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Cardiomegalia/metabolismo , Ratones , Masculino , Miocitos Cardíacos/metabolismo , Ratones Endogámicos C57BL , Presión Sanguínea
18.
Neurobiol Dis ; 195: 106492, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38575093

RESUMEN

We performed a comprehensive study of the morphological, functional, and genetic features of moonwalker (MWK) mice, a mouse model of spinocerebellar ataxia caused by a gain of function of the TRPC3 channel. These mice show numerous behavioral symptoms including tremor, altered gait, circling behavior, impaired motor coordination, impaired motor learning and decreased limb strength. Cerebellar pathology is characterized by early and almost complete loss of unipolar brush cells as well as slowly progressive, moderate loss of Purkinje cell (PCs). Structural damage also includes loss of synaptic contacts from parallel fibers, swollen ER structures, and degenerating axons. Interestingly, no obvious correlation was observed between PC loss and severity of the symptoms, as the phenotype stabilizes around 2 months of age, while the cerebellar pathology is progressive. This is probably due to the fact that PC function is severely impaired much earlier than the appearance of PC loss. Indeed, PC firing is already impaired in 3 weeks old mice. An interesting feature of the MWK pathology that still remains to be explained consists in a strong lobule selectivity of the PC loss, which is puzzling considering that TRPC is expressed in every PC. Intriguingly, genetic analysis of MWK cerebella shows, among other alterations, changes in the expression of both apoptosis inducing and resistance factors possibly suggesting that damaged PCs initiate specific cellular pathways that protect them from overt cell loss.


Asunto(s)
Modelos Animales de Enfermedad , Fenotipo , Animales , Ratones , Cerebelo/patología , Cerebelo/metabolismo , Células de Purkinje/patología , Células de Purkinje/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Genotipo , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Ratones Mutantes Neurológicos , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Free Radic Biol Med ; 219: 141-152, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38636714

RESUMEN

Pulmonary hypertension (PH) is a devastating disease that lacks effective treatment options and is characterized by severe pulmonary vascular remodeling. Pulmonary arterial endothelial cell (PAEC) dysfunction drives the initiation and pathogenesis of pulmonary arterial hypertension. Canonical transient receptor potential (TRPC) channels, a family of Ca2+-permeable channels, play an important role in various diseases. However, the effect and mechanism of TRPCs on PH development have not been fully elucidated. Among the TRPC family members, TRPC4 expression was markedly upregulated in PAECs from hypoxia combined with SU5416 (HySu)-induced PH mice and monocrotaline (MCT)-treated PH rats, as well as in hypoxia-exposed PAECs, suggesting that TRPC4 in PAECs may participate in the occurrence and development of PH. In this study, we aimed to investigate whether TRPC4 in PAECs has an aggravating effect on PH and elucidate the molecular mechanisms. We observed that hypoxia treatment promoted PAEC apoptosis through a caspase-12/endoplasmic reticulum stress (ERS)-dependent pathway. Knockdown of TRPC4 attenuated hypoxia-induced apoptosis and caspase-3/caspase-12 activity in PAECs. Accordingly, adeno-associated virus (AAV) serotype 6-mediated pulmonary endothelial TRPC4 silencing (AAV6-Tie-shRNA-TRPC4) or TRPC4 antagonist suppressed PH progression as evidenced by reduced right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, PAEC apoptosis and reactive oxygen species (ROS) production. Mechanistically, unbiased RNA sequencing (RNA-seq) suggested that TRPC4 deficiency suppressed the expression of the proapoptotic protein sushi domain containing 2 (Susd2) in hypoxia-exposed mouse PAECs. Moreover, TRPC4 activated hypoxia-induced PAEC apoptosis by promoting Susd2 expression. Therefore, inhibiting TRPC4 ameliorated PAEC apoptosis and hypoxic PH in animals by repressing Susd2 signaling, which may serve as a therapeutic target for the management of PH.


Asunto(s)
Apoptosis , Células Endoteliales , Hipertensión Pulmonar , Hipoxia , Canales Catiónicos TRPC , Animales , Ratones , Ratas , Células Cultivadas , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Células Endoteliales/metabolismo , Células Endoteliales/patología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/genética , Hipoxia/metabolismo , Indoles , Ratones Endogámicos C57BL , Monocrotalina/toxicidad , Arteria Pulmonar/patología , Arteria Pulmonar/metabolismo , Pirroles , Ratas Sprague-Dawley , Transducción de Señal , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética , Remodelación Vascular/genética
20.
Int J Biol Macromol ; 265(Pt 1): 130855, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38490377

RESUMEN

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


Asunto(s)
Canales de Potencial de Receptor Transitorio , beta Catenina , Ratones , Animales , Porcinos , beta Catenina/genética , beta Catenina/metabolismo , Músculo Esquelético/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Calcio/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA