Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
Cell Calcium ; 120: 102883, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38643716

RESUMO

The basal and glucose-induced insulin secretion from pancreatic beta cells is a tightly regulated process that is triggered in a Ca2+-dependent fashion and further positively modulated by substances that raise intracellular levels of adenosine 3',5'-cyclic monophosphate (cAMP) or by certain antidiabetic drugs. In a previous study, we have temporally resolved the subplasmalemmal [Ca2+]i dynamics in beta cells that are characterized by trains of sharply delimited spikes, reaching peak values up to 5 µM. Applying total internal reflection fluorescence (TIRF) microscopy and synaptopHluorin to visualize fusion events of individual granules, we found that several fusion events can coincide within 50 to 150 ms. To test whether subplasmalemmal [Ca2+]i microdomains around single or clustered Ca2+ channels may cause a synchronized release of insulin-containing vesicles, we applied simultaneous dual-color TIRF microscopy and monitored Ca2+ fluctuations and exocytotic events in INS-1 cells at high frame rates. The results indicate that fusions can be triggered by subplasmalemmal Ca2+ spiking. This, however, does account for a minority of fusion events. About 90 %-95 % of fusion events either happen between Ca2+ spikes or incidentally overlap with subplasmalemmal Ca2+ spikes. We conclude that only a fraction of exocytotic events in glucose-induced and tolbutamide- or forskolin-enhanced insulin release from INS-1 cells is tightly coupled to Ca2+ microdomains around voltage-gated Ca2+ channels.


Assuntos
Cálcio , Exocitose , Células Secretoras de Insulina , Insulina , Microscopia de Fluorescência , Células Secretoras de Insulina/metabolismo , Cálcio/metabolismo , Animais , Ratos , Insulina/metabolismo , Exocitose/efeitos dos fármacos , Sinalização do Cálcio , Secreção de Insulina/efeitos dos fármacos , Glucose/metabolismo , Vesículas Secretórias/metabolismo
3.
Sci Rep ; 14(1): 6684, 2024 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-38509101

RESUMO

Passing the driving school test can be very challenging, especially in big cities, where up to 52% of all students fail this test. Consequently, many learner drivers experience stress and anxiety. For some learner drivers these feelings can be extreme and negatively affect the performance in the driving test. Different strategies to face anxiety and stress are known, including, for example, psychological or pharmacological approaches and even placebo pills. Recent intriguing findings have also demonstrated that placebos without deception, so-called open-label placebos, successfully reduce anxiety. Here we aimed to test effects of this novel treatment for learner drivers. We investigated whether open-label placebos affect test performance and feelings of anxiety in learner drivers. Sixty-eight healthy participants (mean age 21.94 years, 26 females) were randomized into two groups. The open-label placebo group received placebo pills two weeks before the driving test (two pills each day). The control group received no treatment. Results revealed that the open-label placebo group experienced significantly less anxiety than the control group before the test (measured with the State-Trait-Anxiety-Inventory, STAI-S, and the German Test Anxiety Inventory, PAF). Moreover, in the open-label placebo group less learner drivers failed the driving test (29.41% vs. 52.95%). The results suggest that open-label placebos may provide an ethical unproblematic way to experience less anxiety and might also enhance the probability to pass the driving test. We discuss possible mechanisms of open-label placebos and limitations of our findings.


Assuntos
Ansiedade , Efeito Placebo , Feminino , Humanos , Adulto Jovem , Adulto , Resultado do Tratamento , Ansiedade/tratamento farmacológico , Transtornos de Ansiedade , Emoções
6.
PeerJ ; 11: e16072, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37744227

RESUMO

Background: COVID-19 is a worldwide pandemic caused by the highly infective SARS-CoV-2. There is a need for biomarkers not only for overall prognosis but also for predicting the response to treatments and thus for improvements in the clinical management of patients with COVID-19. Circulating cell-free DNA (cfDNA) has emerged as a promising biomarker in the assessment of various pathological conditions. The aim of this retrospective and observational pilot study was to investigate the range of cfDNA plasma concentrations in hospitalized COVID-19 patients during the first wave of SARS-CoV-2 infection, to relate them to established inflammatory parameters as a correlative biomarker for disease severity, and to compare them with plasma levels in a healthy control group. Methods: Lithium-Heparin plasma samples were obtained from COVID-19 patients (n = 21) during hospitalization in the University Medical Centre of Mainz, Germany between March and June 2020, and the cfDNA concentrations were determined by quantitative PCR yielding amplicons of long interspersed nuclear elements (LINE-1). The cfDNA levels were compared with those of an uninfected control group (n = 19). Results: Plasma cfDNA levels in COVID-19 patients ranged from 247.5 to 6,346.25 ng/ml and the mean concentration was 1,831 ± 1,388 ng/ml (± standard deviation), which was significantly different from the levels of the uninfected control group (p < 0.001). Regarding clinical complications, the highest correlation was found between cfDNA levels and the myositis (p = 0.049). In addition, cfDNA levels correlated with the "WHO clinical progression scale". D-Dimer and C-reactive protein (CRP) were the clinical laboratory parameters with the highest correlations with cfDNA levels. Conclusion: The results of this observational pilot study show a wide range in cfDNA plasma concentrations in patients with COVID-19 during the first wave of infection and confirm that cfDNA plasma concentrations serve as a predictive biomarker of disease severity in COVID-19.


Assuntos
COVID-19 , Ácidos Nucleicos Livres , Humanos , SARS-CoV-2/genética , Projetos Piloto , Estudos Retrospectivos , Gravidade do Paciente , Lítio
7.
Br J Pharmacol ; 180(21): 2736-2749, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37254803

RESUMO

BACKGROUND AND PURPOSE: In macrophages, transient receptor potential vanilloid 2 (TRPV2) channel contributes to various cellular processes such as cytokine production, differentiation, phagocytosis and migration. Due to a lack of selective pharmacological tools, its function in immunological processes is not well understood and the identification of novel and selective TRPV2 modulators is highly desirable. EXPERIMENTAL APPROACH: Novel and selective TRPV2 modulators were identified by screening a compound library using Ca2+ influx assays with human embryonic kidney 293 (HEK293) cells heterologously expressing rat TRPV2. Hits were further characterized and validated with Ca2+ influx and electrophysiological assays. Phagocytosis and migration of macrophages were analysed and the contribution of TRPV2 to the generation of Ca2+ microdomains was studied by total internal reflection fluorescence microscopy (TIRFM). KEY RESULTS: The compound IV2-1, a dithiolane derivative (1,3-dithiolan-2-ylidene)-4-methyl-5-phenylpentan-2-one), is a potent inhibitor of heterologously expressed TRPV2 channels (IC50 = 6.3 ± 0.7 µM) but does not modify TRPV1, TRPV3 or TRPV4 channels. IV2-1 also inhibits TRPV2-mediated Ca2+ influx in macrophages. IV2-1 inhibits macrophage phagocytosis along with valdecoxib and after siRNA-mediated knockdown. Moreover, TRPV2 inhibition inhibits lipopolysaccharide-induced migration of macrophages whereas TRPV2 activation promotes migration. After activation, TRPV2 shapes Ca2+ microdomains predominantly at the margin of macrophages, which are important cellular regions to promote phagocytosis and migration. CONCLUSIONS AND IMPLICATIONS: IV2-1 is a novel TRPV2-selective blocker and underline the role of TRPV2 in macrophage-mediated phagocytosis and migration. Furthermore, we provide evidence that TRPV2 activation generates Ca2+ microdomains, which may be involved in phagocytosis and migration of macrophages.


Assuntos
Lipopolissacarídeos , Macrófagos , Humanos , Ratos , Animais , Lipopolissacarídeos/farmacologia , Células HEK293 , Fagocitose , Expressão Gênica , Canais de Cátion TRPV/genética
8.
Sci Rep ; 13(1): 8372, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37225724

RESUMO

Placebo effects are known for numerous clinical symptoms. Until recently, deception of placebos was thought to be essential for placebo effects, but intriguing new studies suggest that even placebos without concealment (open-label placebos) may help patients with various clinical disorders. Most of those studies compared open-label placebo treatments with no treatment conditions (or treatment "as usual"). Given that open-label placebo studies obviously cannot be blinded, additional control studies are important to assess the efficacy of open-label placebos. The current study aimed to fil this gap by comparing open-label with conventional double-blind placebos and treatment as usual. Patients with seasonal allergic rhinitis were randomly divided in different groups. The first group received open-label placebos, the second double-blind placebos, and the third was treated as usual. After 4 weeks, results demonstrated that open-label placebos improved allergic symptoms more than treatment-as-usual and even more as double-blind placebos. In addition, we observed that allergic symptoms in general (and also the open-label placebo effects) were reduced by the Covid-19 pandemic. The results suggest that seasonal allergic symptoms may be relieved by open-label placebos. We discuss these results by addressing possible different mechanisms of open-label and conventionally concealed placebo treatments.


Assuntos
COVID-19 , Rinite Alérgica Sazonal , Rinite Alérgica , Humanos , Pandemias , Rinite Alérgica/tratamento farmacológico , Método Duplo-Cego
10.
Sci Rep ; 13(1): 1954, 2023 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-36732349

RESUMO

Taste may be the first sense that emerged in evolution. Taste is also a very important sense since it signals potential beneficial or dangerous effects of foods. Given this fundamental role of taste in our lives, it is not surprising that taste also affects our psychological perception and thinking. For example, previous research demonstrated remarkable psychological effects of sweet taste experiences, suggesting that sweetness may be a source domain for prosocial functioning. Recent research reports that briefly experiencing sweet taste made participants more helpful in their intentions and behavior. The current study aims to test this hypothesis and to examine the neural underpinnings of this effect by using an fMRI approach. Participants were asked to taste sweet, salty, and neutral taste while lying in the fMRI scanner. Subsequently their prosocial behavior was tested by playing the dictator game, a measure of prosocial behavior. Results showed that sweet taste was associated with an increase in prosocial behavior compared with previously experiencing salty taste but did not affect control stimuli ratings. FMRI results revealed a modulation of the dorsal anterior cingulate cortex associated with this sweetness effect. This brain area is known to play a central role for monitoring conflicts and decisions and has been directly linked to selfish and prosocial economic decisions. The results demonstrate that sweet taste has complex psychological effects including positive and socially desirable outcomes. We discuss the results with other studies on psychological sweetness effects and suggest possible implications of these findings.


Assuntos
Altruísmo , Paladar , Humanos , Disgeusia , Percepção Gustatória , Encéfalo/diagnóstico por imagem , Preferências Alimentares/psicologia
11.
Mol Pharmacol ; 103(2): 48-62, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36400433

RESUMO

G protein-coupled receptors (GPCRs) constitute the most versatile family of pharmacological target proteins. For some "orphan" GPCRs, no ligand or drug-like modulator is known. In this study, we have established and applied a parallelized assay to coscreen 29 different human GPCRs. Three compounds, chlorhexidine, Lys-05, and 9-aminoacridine, triggered transient Ca2+ signals linked to the expression of GPR30. GPR30, also named G protein-coupled estrogen receptor 1 (GPER1), was reported to elicit increases in cAMP in response to 17ß-estradiol, 4-hydroxytamoxifen, or G-1. These findings could, however, not be reproduced by other groups, and the deorphanization of GPR30 is, therefore, intensely disputed. The unbiased screen and following experiments in transiently or stably GPR30-overexpressing HEK293 cells did not show responses to 17ß-estradiol, 4-hydroxytamoxifen, or G-1. A thorough analysis of the activated signaling cascade revealed a canonical Gq-coupled pathway, including phospholipase C, protein kinase C and ERK activation, receptor internalization, and sensitivity to the Gq inhibitor YM-254890. When expressed in different cell lines, the localization of a fluorescent GPR30 fusion protein appeared variable. An efficient integration into the plasma membrane and stronger functional responses were found in HEK293 and in MCF-7 cells, whereas GPR30 appeared mostly retained in endomembrane compartments in Cos-7 or HeLa cells. Thus, conflicting findings may result from the use of different cell lines. The newly identified agonists and the finding that GPR30 couples to Gq are expected to serve as a starting point for identifying physiologic responses that are controlled by this GPCR. SIGNIFICANCE STATEMENT: This study has identified and thoroughly characterized novel and reliably acting agonists of the G protein-coupled receptor GPER1/GPR30. Applying these agonists, this study demonstrates that GPR30 couples to the canonical Gq-phospholipase C pathway and is rapidly internalized upon continuous exposure to the agonists.


Assuntos
Estradiol , Receptores Acoplados a Proteínas G , Humanos , Células HEK293 , Células HeLa , Receptores Acoplados a Proteínas G/metabolismo , Estradiol/farmacologia
12.
Neuropsychopharmacology ; 48(3): 560-566, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36456814

RESUMO

While placebo effects are well-known, research in the last decade revealed intriguing effects that placebos may have beneficial effects even when given without deception. At first glance, this seems paradoxical, but several studies have reported improvements in pain, depression, or anxiety. However, it still remains unclear whether these results represent objective biological effects or simply a bias in response and what neural underpinnings are associated with the open-label placebo effects. In two studies, we address this gap by demonstrating that open-label placebos reduce self-reported emotional distress when viewing highly arousing negative pictures. This reduced emotional distress was associated with an activation of brain areas known to modulate affective states such as the periaqueductal gray, the bilateral anterior hippocampi, and the anterior cingulate cortex. We did not find any prefrontal brain activation. Furthermore, brain activation was not associated with expectation of effects. In contrast, we found that brain responses were linked to general belief in placebos. The results demonstrate that the neural mechanisms of open-label placebo effects are partly identical to the neurobiological underpinnings of conventional placebos, but our study also highlights important differences with respect to a missing engagement of prefrontal brain regions, suggesting that expectation of effects may play a less prominent role in open-label placebos.


Assuntos
Efeito Placebo , Angústia Psicológica , Humanos , Dor , Ansiedade/tratamento farmacológico , Transtornos de Ansiedade
13.
Cell Death Dis ; 13(12): 1052, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-36535942

RESUMO

Glucocorticoids (GCs) are widely used in tumor therapy to reduce tumor growth, inflammation, edema, and other side effects. Controversially, GCs may also cause the progression of highly aggressive pancreatic ductal adenocarcinoma (PDAC). Because microRNA (miR) and autophagy signaling support the invasive growth of PDAC, we asked whether these mechanisms may be targeted by GCs. Six established human PDAC cell lines, tissue from patients who received GC medication (n = 35) prior to surgery, or not (n = 35), and tumor xenografts were examined by RT‒qPCR, transmission electron microscopy (TEM), monodansylcadaverine (MDC) staining, immunohistochemistry, in situ hybridization, gene array and Kaplan‒Meier analysis with bioinformatics, and MTT, western blot, colony, spheroid, migration, and invasion assays. We found that various GCs, including dexamethasone (DEX), induced typical features of macroautophagy with the appearance of autolysosomes, enhanced LC3-II, decreased SQSTM1/p62 expression and induced epithelial-mesenchymal transition (EMT) and gemcitabine resistance. The GC receptor (GR) antagonist mifepristone (RU486) counteracted DEX-induced autophagy features, suggesting that the GC-GR complex is involved in the induction of autophagy. The autophagy-related miR-378i and miR-378a-3p were selected as the top upregulated candidates, and their high expression in PDAC patient tissue correlated with low survival. siRNA-mediated downregulation of miR-378 inhibited DEX-induced autophagy, and tumor progression. Bioinformatics confirmed the contribution of miR-378 to the regulation of signaling networks involved in GC-induced autophagy and tumor progression. The construction of a molecular docking model revealed stable binding of miR-378 to the DEX-GR complex, suggesting direct regulation. These substantial, novel, in-depth data reveal that GCs favor autophagy-mediated cancer progression by inducing miR-378 and GR binding and implicate GR and miR-378 as new therapeutic targets.


Assuntos
Carcinoma Ductal Pancreático , MicroRNAs , Neoplasias Pancreáticas , Humanos , Autofagia , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glucocorticoides/farmacologia , MicroRNAs/genética , Simulação de Acoplamento Molecular , Neoplasias Pancreáticas/patologia , Animais , Neoplasias Pancreáticas
14.
Matrix Biol ; 114: 84-107, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36375776

RESUMO

Several collagen subtypes are involved in pancreatic ductal adenocarcinoma (PDAC) desmoplasia, which constrains therapeutic efficacy. We evaluated collagen type VIII alpha 1 chain (COL8A1), whose function in PDAC is currently unknown. We identified COL8A1 expression in 7 examined PDAC cell lines by microarray analysis, western blotting, and RT‒qPCR. Higher COL8A1 expression occurred in 2 gemcitabine-resistant PDAC cell lines; pancreas tissue (n=15) from LSL-KrasG12D/+; p48-Cre mice with advanced PDAC predisposition; and PDAC parenchyma and stroma of a patient tissue microarray (n=82). Bioinformatic analysis confirmed higher COL8A1 expression in PDAC patient tissue available from TCGA (n=183), GTEx (n=167), and GEO (n=261) databases. siRNA or lentiviral sh-mediated COL8A1 inhibition in PDAC cells reduced migration, invasion and gemcitabine resistance and resulted in lower cytidine deaminase and thymidine kinase 2 expression and was rescued by COL8A1-secreting cancer-associated fibroblasts (CAFs). The activation of COL8A1 expression involved cJun/AP-1, as demonstrated by CHIP assay and siRNA inhibition. Downstream of COL8A1, activation of ITGB1 and DDR1 receptors and PI3K/AKT and NF-κB signaling occurred, as detected by expression, adhesion and EMSA binding studies. Orthotopic transplantation of PDAC cells with downregulated COL8A1 expression resulted in reduced tumor xenograft growth and lower gemcitabine resistance but was prevented by cotransplantation of COL8A1-secreting CAFs. Most importantly, COL8A1 expression in PDAC patient tissues from our clinic (n=84) correlated with clinicopathological data, and we confirmed these findings by the use of patient data (n=177) from the TCGA database. These findings highlight COL8A1 expression in tumor and stromal cells as a new biomarker for PDAC progression.


Assuntos
Adenocarcinoma , Carcinoma Ductal Pancreático , Colágeno Tipo VIII , Neoplasias Pancreáticas , Animais , Humanos , Camundongos , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosfatidilinositol 3-Quinases , RNA Interferente Pequeno , Colágeno Tipo VIII/metabolismo , Neoplasias Pancreáticas
15.
Brain Sci ; 12(10)2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36291321

RESUMO

An essential task of any system is to distinguish between itself and its environment, between what is inside and that which is outside [...].

16.
Biomed Pharmacother ; 153: 113511, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36076598

RESUMO

Silver has been in clinical use since ancient times and silver nanoparticles (AgNPs) have attracted attention in cancer therapy. We investigated the mechanisms by which AgNPs inhibit pancreatic ductal adenocarcinoma (PDAC). AgNPs were synthesized and 3 human PDAC and 2 nonmalignant primary cell lines were treated with AgNPs. MTT, MAPK, colony, spheroid and scratch assays, Western blotting, TEM, annexin V, 7-AAD, and H2DCFDA staining, FACS analysis, mRNA array and bioinformatics analyses, tumor xenograft transplantation, and immunohistochemistry of the treated cells were performed. We found that minimal AgNPs amounts selectively eradicated PDAC cells within a few hours. AgNPs inhibited cell migration and spheroid and colony formation, damaged mitochondria, and induced paraptosis-like cell death with the presence of cytoplasmic vacuoles, dilation of the ER and mitochondria, ROS formation, MAPK activity, and p62 and LC3b expression, whereas effects on the nucleus, DNA fragmentation, or caspases were not detectable. AgNPs strongly decreased tumor xenograft growth without side effects and reduced the expression of markers for proliferation and DNA repair, but upregulated paraptosis markers. The results highlight nanosilver as complementary agent to improve the therapeutic efficacy in pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático , Nanopartículas Metálicas , Neoplasias Pancreáticas , Apoptose , Carcinoma Ductal Pancreático/genética , Morte Celular , Linhagem Celular Tumoral , Humanos , Neoplasias Pancreáticas/patologia , Prata/farmacologia , Prata/uso terapêutico , Neoplasias Pancreáticas
17.
ACS Earth Space Chem ; 6(8): 2041-2052, 2022 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-36016759

RESUMO

Anthropogenic emissions of vanadium (V) into terrestrial and aquatic surface systems now match those of geogenic processes, and yet, the geochemistry of vanadium is poorly described in comparison to other comparable contaminants like arsenic. In oxic systems, V is present as an oxyanion with a +5 formal charge on the V center, typically described as H x VO4 (3-x)-, but also here as V(V). Iron (Fe) and manganese (Mn) (oxy)hydroxides represent key mineral phases in the cycling of V(V) at the solid-solution interface, and yet, fundamental descriptions of these surface-processes are not available. Here, we utilize extended X-ray absorption fine structure (EXAFS) and thermodynamic calculations to compare the surface complexation of V(V) by the common Fe and Mn mineral phases ferrihydrite, hematite, goethite, birnessite, and pyrolusite at pH 7. Inner-sphere V(V) complexes were detected on all phases, with mononuclear V(V) species dominating the adsorbed species distribution. Our results demonstrate that V(V) adsorption is exergonic for a variety of surfaces with differing amounts of terminal -OH groups and metal-O bond saturations, implicating the conjunctive role of varied mineral surfaces in controlling the mobility and fate of V(V) in terrestrial and aquatic systems.

18.
Cell Calcium ; 106: 102640, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36030694

RESUMO

The transient receptor potential cation channel, subfamily M, members 6 and 7 (TRPM6 and TRPM7) are homologous membrane proteins encompassing cation channel units fused to cytosolic serine/threonine-protein kinase domains. Clinical studies and experiments with animal disease models suggested that selective inhibition of TRPM6 and TRPM7 currents might be beneficial for subjects with immune and cardiovascular disorders, tumours and other pathologies, but the suitable pharmacological toolkit remains underdeveloped. The present study identified small synthetic molecules acting specifically on the channel moieties of TRPM6 and TRPM7. Using electrophysiological analysis in conjunction with Ca2+ imaging, we show that iloperidone and ifenprodil inhibit the channel activity of recombinant TRPM6 with IC50 values of 0.73 and 3.33 µM, respectively, without an impact on the TRPM7 channel. We also found that VER155008 suppresses the TRPM7 channel with an IC50 value of 0.11 µM but does not affect TRPM6. Finally, the effects of iloperidone and VER155008 were found to be suitable for blocking native endogenous TRPM6 and TRPM7 in a collection of mouse and human cell models. Hence, the identification of iloperidone, ifenprodil, and VER155008 allows for the first time to selectively manipulate TRPM6 and TRPM7 currents.


Assuntos
Canais de Cátion TRPM , Animais , Humanos , Isoxazóis/farmacologia , Magnésio/metabolismo , Camundongos , Piperidinas/farmacologia , Proteínas Serina-Treonina Quinases , Nucleosídeos de Purina/farmacologia , Canais de Cátion TRPM/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/metabolismo
19.
Cell Calcium ; 106: 102634, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35917684

RESUMO

Amongst the superfamily of transient receptor potential (TRP) channels, TRPV5 and TRPV6 are specialized members that mediate Ca2+-selective transport across epithelial membranes. Intriguingly, fluorescent fusion proteins of TRPV5 or TRPV6 are hardly discernible within the plasma membrane of living cells. Instead, TRPV6 is mostly found in vesicular membrane compartments, indicating either a rapid degradation or cycling of channel-bearing vesicles between endomembrane compartments and the plasma membrane. In TRPV6-expressing cells, brefeldin A, a toxin that blocks the transit between the endoplasmic reticulum and the Golgi apparatus, caused a drop in [Ca2+]i with a half time in the range of 0.5-1 h. Upon wash-out of the toxin, the [Ca2+]i rose to a steady-state level within 2-3 h. Consistently, the synchronized forward trafficking of TRPV6VL-eGFP after brefeldin A wash-out led to a visible accumulation of the protein within the plasma membrane, as shown by confocal and total internal reflection microscopy. Analysis of the internalization route and differentiation of vesicle populations provided evidence for a clathrin-dependent internalization pathway. Most TRPV6VL-bearing vesicles co-stained with Rab5a, a marker protein for early endosomes. Fewer vesicles were co-localized with Rab7a (late endosomes) or with Rab11 (recycling endosomes). From these data, we propose that the lack of plasma membrane visibility of the channel results from a rapid internalization, which in addition to transcriptional regulation, adds a layer of functional channel regulation to modulate transepithelial Ca2+ transport.


Assuntos
Cálcio , Canais de Cátion TRPV , Brefeldina A/metabolismo , Brefeldina A/farmacologia , Cálcio/metabolismo , Membrana Celular/metabolismo , Complexo de Golgi/metabolismo , Canais de Cátion TRPV/metabolismo
20.
Br J Pharmacol ; 179(24): 5290-5304, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35916168

RESUMO

BACKGROUND AND PURPOSE: Ca2+ signalling mediated by the thermosensitive, non-selective, Ca2+ -permeable transient receptor potential channel TRPV3 is assumed to play a critical role in regulating several aspects of skin functions, such as keratinocyte proliferation, differentiation, skin barrier formation and wound healing. Studying the function of TRPV3 in skin homeostasis, however, is still constrained by a lack of potent and selective pharmacological modulators of TRPV3. EXPERIMENTAL APPROACH: By screening an in-house compound library using fluorometric intracellular Ca2+ assays, we identified two chemically related hits. The more potent and efficient TRPV3 activator 2-(2-chloro-3-isopropylcyclopent-2-en-1-yl)-4-methylphenol (KS0365) was further evaluated in fluo-4-assisted Ca2+ assays, different Ca2+ imaging approaches, electrophysiological studies, cytotoxicity and migration assays. KEY RESULTS: KS0365 activated recombinant and native mouse TRPV3 more potently and with a higher efficacy compared with 2-APB and did not activate TRPV2 or TRPV4 channels. The activation of TRPV3 by KS0365 super-additively accelerated the EGF-induced keratinocyte migration, which was inhibited by the TRP channel blocker ruthenium red or by siRNA-mediated TRPV3 knockdown. Moreover, KS0365 induced strong Ca2+ responses in migrating front cells and in leading edges of keratinocytes. CONCLUSIONS AND IMPLICATIONS: The selective TRPV3 activator KS0365 triggers increases in [Ca2+ ]i with most prominent signals in the leading edge and accelerates migration of keratinocytes. TRPV3 activators may promote re-epithelialization upon skin wounding.


Assuntos
Queratinócitos , Canais de Cátion TRPV , Animais , Camundongos , Diferenciação Celular , Movimento Celular , Proliferação de Células , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/fisiologia , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...