Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Biochem Biophys Res Commun ; 666: 1-9, 2023 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-37167718

RESUMO

Lysosomal exocytosis is an essential cellular event for remodeling the extracellular matrix through secreting lysosomal enzymes and developing drug resistance. However, the detailed mechanism underlying the lysosomal exocytosis-driven acquisition of drug resistance is not completely understood. Genetic variations in gefitinib-sensitive (HSC3) and -resistant (HSC3/GR) oral squamous carcinoma cell lines were identified using whole-exome sequencing (WES). The physiological role of the ATP-binding cassette subfamily A member 2 (ABCA2) in gefitinib-induced lysosomal trafficking was evaluated in vitro, through overexpressing ABCA2 and its single nucleotide polymorphisms (SNPs). WES analysis showed that the 554 SNPs harboring 244 genes appeared to be differentially generated depending on gefitinib resistance. Among these genes, ABCA2 was enriched in 24 of 39 gene ontology terms. Two missense SNPs of ABCA2, 4873T  >  A (rs1831123356) and 4873T  >  A, were generated only in gefitinib-sensitive cells. Furthermore, HEK293 cells expressing the wild-type ABCA2 (WT ABCA2) acquired tolerance for gefitinib-induced cytotoxicity by increasing gefitinib sequestration in lysosomes and lysosomal exocytosis. Conversely, cells expressing each ABCA2 SNP exhibited lower efficacy in developing tolerance to gefitinib-induced responses than those expressing WT ABCA2. Notably, HSC3/GR cells were also tolerant to erlotinib and sunitinib but not osimertinib. Furthermore, tolerance for multiple tyrosine kinase inhibitors was attenuated by the deletion of ABCA2. These findings demonstrate that ABCA2 and its SNPs should be considered prominent targets for overcoming multi-drug resistance and enhancing the efficacy of chemotherapeutics.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Antineoplásicos , Carcinoma de Células Escamosas , Gefitinibe , Neoplasias Bucais , Humanos , Antineoplásicos/farmacologia , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/genética , Gefitinibe/farmacologia , Células HEK293 , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/genética , Polimorfismo de Nucleotídeo Único
2.
Biochem Biophys Res Commun ; 627: 152-159, 2022 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-36037747

RESUMO

Lysosomes are emerging as versatile signaling hubs that mediate numerous cellular processes, including the development of drug resistance in cancer cells. Transient receptor potential mucolipin 3 (TRPML3), an endolysosomal Ca2+-permeable channel, is implicated in regulating lysosomal trafficking during endocytosis and autophagy. However, the role of TRPML3 in cancer progression remains unclear. In this study, we focused on identifying key molecules that modulate exosomal release triggered by lysosomal exocytosis during the development of gefitinib resistance in non-small cell lung cancer (NSCLC). We found that the basal release of exosomes and lysosomal exocytosis is higher in the gefitinib-resistant NSCLC cell line HCC827/GR than in the gefitinib-sensitive NSCLC cell line HCC827. Notably, exosomal release and lysosomal exocytosis were associated with an increase in TRPML3 expression. Lysosomal Ca2+ release via TRPML3 was triggered by the gefitinib-mediated elevation of lysosomal pH. Furthermore, TRPML3 deficiency enhanced the gefitinib-mediated increase in sub-G0 cell population, reduction of cell proliferation, and poly (ADP-ribose) polymerase cleavage. These data demonstrated that TRPML3 is a promising modulator of drug resistance. By sensing the elevation of lysosomal pH, it mediates lysosomal Ca2+ release, lysosomal trafficking and exocytosis, and exosomal release. Taken together, our study is the first to report the autonomous defense mechanism developed in NSCLC cells against the small-molecule tyrosine kinase inhibitor gefitinib, leading to acquired drug resistance.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Resistência a Medicamentos , Resistencia a Medicamentos Antineoplásicos , Gefitinibe/farmacologia , Gefitinibe/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Lisossomos/metabolismo
3.
Biochem Biophys Res Commun ; 578: 150-156, 2021 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-34562655

RESUMO

Berberine is a natural isoquinoline alkaloid present in various herbs and is effective against metabolic syndrome in the pre-diabetic stage and high insulin resistance. The present study aimed to determine the effectiveness of WJCPR11, a berberine derivative that is commonly used for diabetes treatment, in ameliorating insulin resistance and diabetes treatment. WJCPR11 promoted adipocyte differentiation to a higher extent than other berberine derivatives and showed no noticeable toxicity in its effective concentration range. It increased the mRNA expression levels and protein abundance of adipogenic markers, including peroxisome proliferator-activated receptor γ (PPARγ), glucose transporter type 4 (GluT4), and fatty acid synthase (FAS), and markedly enhanced the level of adiponectin, a distinct marker of insulin sensitivity. Meanwhile, the mRNA levels of inflammatory markers such as plasminogen activator inhibitor-1 (PAI-1), monocyte chemoattractant protein-1 (MCP-1), and interleukin 6 (IL-6) were reduced after WJCPR11 treatment. Furthermore, the tumor necrosis factor-α (TNF-α)-induced inhibition of adipocyte differentiation and downregulation of glucose uptake were markedly reversed by WJCPR11 treatment. Collectively, the findings of this study indicate that WJCPR11 has great potential for diabetes treatment.


Assuntos
Adipócitos/citologia , Berberina/análogos & derivados , Glucose/metabolismo , Estado Pré-Diabético/tratamento farmacológico , Fator de Necrose Tumoral alfa/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Berberina/farmacologia , Diferenciação Celular/fisiologia , Células Cultivadas , Resistência à Insulina , Camundongos , Estado Pré-Diabético/metabolismo , Estado Pré-Diabético/patologia
4.
Biochem Biophys Res Commun ; 510(3): 409-415, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30711251

RESUMO

The lysosomal Ca2+ permeable channel TRPML1 (MCOLN1) plays key roles in lysosomal membrane trafficking, including the fusion of late endosomes to lysosomes and lysosomal exocytosis, both of which are essential for release of exosomes into the extracellular milieu. Multiple lines of evidence indicate that the contents of adipocyte-derived exosomes mediate diverse cellular responses, including adipogenic differentiation. In this study, we aimed to define the potential roles of TRPML1 in lysosomal membrane trafficking during adipogenesis and in exosomal release. In response to adipogenic stimuli, the endogenous TRPML1 expression in OP9 pre-adipocytes was increased in a time-dependent manner, and the acute deletion of TRPML1 reduced lipid synthesis and expression of differentiation-related marker genes. Notably, mature adipocyte-derived exosomes were found to be necessary for adipogenesis and were dependent on TRPML1-mediated lysosomal exocytosis. Taken together, our findings indicate that TRPML1 mediates diverse roles in adipocyte differentiation and exosomal release. Further, we propose that TRPML1 should be considered as a regulator of obesity-related diseases.


Assuntos
Adipogenia , Exocitose , Exossomos/metabolismo , Lisossomos/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Células Cultivadas , Camundongos , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/biossíntese
5.
Biochem Biophys Res Commun ; 496(2): 693-699, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29366789

RESUMO

Gingivitis, the mildest form of periodontitis, is generally considered a consequence of prolonged exposure of the gingiva to periodontal pathogens. On the other hand, several epidemiologic reports have suggested that other etiologic factors such as oral acidification may also increase the susceptibility of the periodontium to destruction. However, the pathologic mechanism underlying the effects of oral acidification on the gingiva is still largely unknown. In this study, we analyzed molecular pathways mediating the influence of the acidic environment on human gingival fibroblasts (HGFs). Acidic extracellular pH caused biphasic increase of intracellular Ca2+ level ([Ca2+]i) through activation of ovarian cancer G protein-coupled receptor 1, phospholipase C, and Ca2+ release from the endoplasmic reticulum, but not through voltage-gated Ca2+ channels or extracellular Ca2+ influx via transient receptor potential cation channel subfamily V member 1. The acidic environment was also transiently cytotoxic for HGFs; however, the activation of pro-apoptotic proteins poly (ADP-ribose) polymerase-1 and BAX was not observed. Furthermore, we found that intracellular matrix metalloproteinase 1 was consistently upregulated in HGFs grown in regular medium, but significantly reduced in the acidic medium, which depended on [Ca2+]i increase, lysosomal pH homeostasis, and Ca2+-dependent protease calpain. Considering that HGFs, essential for oral wound healing, in the in vitro culture system are placed in wound repair-like conditions, our findings provide important insights into molecular mechanisms underlying HGF functional impairment and chronic damage to the gingiva caused by the acidic intraoral environment.


Assuntos
Cálcio/metabolismo , Calpaína/metabolismo , Fibroblastos/citologia , Gengiva/citologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Linhagem Celular , Fibroblastos/metabolismo , Gengiva/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Lisossomos/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Fosfolipases Tipo C/metabolismo , Cicatrização
6.
EMBO Rep ; 17(2): 266-78, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26682800

RESUMO

Mutations in TRPML1 cause the lysosomal storage disease mucolipidosis type IV (MLIV). The role of TRPML1 in cell function and how the mutations cause the disease are not well understood. Most studies focus on the role of TRPML1 in constitutive membrane trafficking to and from the lysosomes. However, this cannot explain impaired neuromuscular and secretory cells' functions that mediate regulated exocytosis. Here, we analyzed several forms of regulated exocytosis in a mouse model of MLIV and, opposite to expectations, we found enhanced exocytosis in secretory glands due to enlargement of secretory granules in part due to fusion with lysosomes. Preliminary exploration of synaptic vesicle size, spontaneous mEPSCs, and glutamate secretion in neurons provided further evidence for enhanced exocytosis that was rescued by re-expression of TRPML1 in neurons. These features were not observed in Niemann-Pick type C1. These findings suggest that TRPML1 may guard against pathological fusion of lysosomes with secretory organelles and suggest a new approach toward developing treatment for MLIV.


Assuntos
Exocitose , Lisossomos/metabolismo , Mucolipidoses/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Vesículas Secretórias/metabolismo , Animais , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores , Ácido Glutâmico/metabolismo , Camundongos , Potenciais Pós-Sinápticos em Miniatura , Mucolipidoses/genética , Neurônios/metabolismo , Neurônios/fisiologia , Doença de Niemann-Pick Tipo C/genética , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
7.
Biochem Biophys Res Commun ; 482(4): 1073-1079, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27913299

RESUMO

ß-lapachone (ß-L) is a substrate of reduced nicotinamide adenine dinucleotide (NADH): quinone oxidoreductase 1 (NQO1). NQO1 reduces quinones to hydroquinones using NADH as an electron donor and consequently increases the intracellular NAD+/NADH ratio. The activation of NQO1 by ß-L has beneficial effects on several metabolic syndromes, such as obesity, hypertension, and renal injury. However, the effect of ß-L on bone metabolism remains unclear. Here, we show that ß-L might be a potent inhibitor of receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis. ß-L inhibited osteoclast formation in a dose-dependent manner and also reduced the expression of osteoclast differentiation marker genes, such as tartrate-resistant acid phosphatase (Acp5 or TRAP), cathepsin K (CtsK), the d2 isoform of vacuolar ATPase V0 domain (Atp6v0d2), osteoclast-associated receptor (Oscar), and dendritic cell-specific transmembrane protein (Dc-stamp). ß-L treatment of RANKL-induced osteoclastogenesis significantly increased the cellular NAD+/NADH ratio and resulted in the activation of 5' AMP-activated protein kinase (AMPK), a negative regulator of osteoclast differentiation. In addition, ß-L treatment led to significant suppression of the expression of peroxisome proliferator-activated receptor gamma (PPARγ) and peroxisome proliferator-activated receptor gamma coactivator 1ß (PGC1ß), which can stimulate osteoclastogenesis. ß-L treatment downregulated c-Fos and nuclear factor of activated T-cells 1 (NFATc1), which are master transcription factors for osteoclastogenesis. Taken together, the results demonstrated that ß-L inhibits RANKL-induced osteoclastogenesis and could be considered a potent inhibitor of RANKL-mediated bone diseases, such as postmenopausal osteoporosis, rheumatoid arthritis, and periodontitis.


Assuntos
Naftoquinonas/química , Osteoclastos/citologia , Ligante RANK/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Doenças Ósseas/metabolismo , Diferenciação Celular , Sobrevivência Celular , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , NAD/química , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/metabolismo , Osteogênese , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
8.
Mol Cell ; 32(3): 439-48, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18995841

RESUMO

The receptor-evoked Ca(2+) signal includes activation of the store-operated channels (SOCs) TRPCs and the Orais. Although both are gated by STIM1, it is not known how STIM1 gates the channels and whether STIM1 gates the TRPCs and Orais by the same mechanism. Here, we report the molecular mechanism by which STIM1 gates TRPC1, which involves interaction between two conserved, negatively charged aspartates in TRPC1((639)DD(640)) with the positively charged STIM1((684)KK(685)) in STIM1 polybasic domain. Charge swapping and functional analysis revealed that exact orientation of the charges on TRPC1 and STIM1 are required, but all positive-negative charge combinations on TRPC1 and STIM1, except STIM1((684)EE(685))+TRPC1((639)RR(640)), are functional as long as they are reciprocal, indicating that STIM1 gates TRPC1 by intermolecular electrostatic interaction. Similar gating was observed with TRPC3((697)DD(698)). STIM1 gates Orai1 by a different mechanism since the polybasic and S/P domains of STIM1 are not required for activation of Orai1 by STIM1.


Assuntos
Canais de Cálcio/fisiologia , Cálcio/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Canais de Cátion TRPC/fisiologia , Sequência de Aminoácidos , Biotinilação , Linhagem Celular , Membrana Celular/fisiologia , Retículo Endoplasmático/fisiologia , Humanos , Ativação do Canal Iônico , Rim , Proteínas de Membrana/química , Dados de Sequência Molecular , Mutação , Proteínas de Neoplasias/química , Proteína ORAI1 , Eletricidade Estática , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/química , Canais de Cátion TRPC/genética , Transfecção
9.
Adv Exp Med Biol ; 898: 47-66, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27161224

RESUMO

The Ca(2+) second messenger is initiated at ER/PM junctions and propagates into the cell interior to convey the receptor information. The signal is maintained by Ca(2+) influx across the plasma membrane through the Orai and TRPC channels. These Ca(2+) influx channels form complexes at ER/PM junctions with the ER Ca(2+) sensor STIM1, which activates the channels. The function of STIM1 is modulated by other STIM isoforms like STIM1L, STIM2 and STIM2.1/STIM2ß and by SARAF, which mediates the Ca(2+)-dependent inhibition of Orai channels. The ER/PM junctions are formed at membrane contact sites by tethering proteins that generate several types of ER/PM junctions, such as PI(4,5)P2-poor and PI(4,5)P2-rich domains. This chapter discusses several properties of the TRPC channels, the Orai channels and the STIMs, their key interacting proteins and how interaction of the STIMs with the channels gates their activity. The chapter closes by highlighting open questions and potential future directions in this field.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canais de Cátion TRPC/metabolismo , Cálcio/metabolismo , Membrana Celular/metabolismo , Humanos , Ativação do Canal Iônico , Molécula 1 de Interação Estromal
10.
Ann Neurol ; 75(1): 77-87, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24114805

RESUMO

OBJECTIVE: Autosomal dominant cortical myoclonus and epilepsy (ADCME) is characterized by distal, fairly rhythmic myoclonus and epilepsy with variable severity. We have previously mapped the disease locus on chromosome 2p11.1-q12.2 by genome-wide linkage analysis. Additional pedigrees affected by similar forms of epilepsy have been associated with chromosomes 8q, 5p, and 3q, but none of the causing genes has been identified. We aim to identify the mutant gene responsible for this form of epilepsy. METHODS: Genes included in the ADCME critical region were directly sequenced. Coimmunoprecipitation, immunofluorescent, and electrophysiologic approaches to transfected human cells have been utilized for testing the functional significance of the identified mutation. RESULTS: Here we show that mutation in the α2 -adrenergic receptor subtype B (α2B -AR) is associated with ADCME by identifying a novel in-frame insertion/deletion in 2 Italian families. The mutation alters several conserved residues of the third intracellular loop, hampering neither the α2B -AR plasma membrane localization nor the arrestin-mediated internalization capacity, but altering the binding with the scaffolding protein spinophilin upon neurotransmitter activation. Spinophilin, in turn, regulates interaction of G protein coupled receptors with regulator of G protein signaling proteins. Accordingly, the mutant α2B -AR increases the epinephrine-stimulated calcium signaling. INTERPRETATION: The identified mutation is responsible for ADCME, as the loss of α2B -AR/spinophilin interaction causes a gain of function effect. This work implicates for the first time the α-adrenergic system in human epilepsy and opens new ways of understanding the molecular pathway of epileptogenesis, widening the spectrum of possible therapeutic targets.


Assuntos
Epilepsias Mioclônicas/diagnóstico , Epilepsias Mioclônicas/genética , Mutação/genética , Receptores Adrenérgicos alfa 2/genética , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Epilepsias Mioclônicas/fisiopatologia , Potenciais Somatossensoriais Evocados/fisiologia , Feminino , Células HeLa , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Receptores Adrenérgicos alfa 2/química , Xenopus laevis
11.
J Nat Prod ; 78(9): 2167-74, 2015 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26308264

RESUMO

Harpagoside (HAR) is a natural compound isolated from Harpagophytum procumbens (devil's claw) that is reported to have anti-inflammatory effects; however, these effects have not been investigated in the context of bone development. The current study describes for the first time that HAR inhibits receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclastogenesis in vitro and suppresses inflammation-induced bone loss in a mouse model. HAR also inhibited the formation of osteoclasts from mouse bone marrow macrophages (BMMs) in a dose-dependent manner as well as the activity of mature osteoclasts, including filamentous actin (F-actin) ring formation and bone matrix breakdown. This involved a HAR-induced decrease in extracellular signal-regulated kinase (ERK) and c-jun N-terminal kinase (JNK) phosphorylation, leading to the inhibition of Syk-Btk-PLCγ2-Ca(2+) in RANKL-dependent early signaling, as well as the activation of c-Fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1), which resulted in the down-regulation of various target genes. Consistent with these in vitro results, HAR blocked lipopolysaccharide (LPS)-induced bone loss in an inflammatory osteoporosis model. However, HAR did not prevent ovariectomy-mediated bone erosion in a postmenopausal osteoporosis model. These results suggest that HAR is a valuable agent against inflammation-related bone disorders but not osteoporosis induced by hormonal abnormalities.


Assuntos
Glicosídeos/farmacologia , Osteoclastos/efeitos dos fármacos , Piranos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Regulação para Baixo/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Glicosídeos/química , Inflamação/metabolismo , Mediadores da Inflamação , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estrutura Molecular , Fosfolipase C gama , Proteínas Proto-Oncogênicas c-fos/genética , Piranos/química , Ligante RANK/farmacologia , Receptor Ativador de Fator Nuclear kappa-B
12.
BMC Complement Altern Med ; 15: 226, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26169673

RESUMO

BACKGROUND: Portulaca oleracea (PO) has been widely used as traditional medicine because of its pharmacological activities. However, the effects of PO on osteoclasts that modulate bone homeostasis are still elusive. METHODS: In this study, we examined the effects of PO ethanol extract (POEE) on receptor activator of nuclear factor-κB ligand (RANKL)-mediated Ca(2+) mobilization, nuclear factor of activated T-cell c1 (NFATc1) amplification, tartrate-resistant acid phosphatase-positive (TRAP+) multinucleated cell (MNC) formation, and cytotoxicity. RESULTS: Our results demonstrated that POEE suppressed RANKL-induced Ca(2+) oscillations by inhibition of Ca(2+) release from internal Ca(2+) stores, resulting in reduction of NFATc1 amplification. Notably, POEE attenuated RANKL-mediated cytotoxicity and cleavage of polyadenosine 5'-diphosphate-ribose polymerase (PARP), resulted in enhanced formation of TRAP+ MNCs. CONCLUSIONS: These results present in vitro effects of POEE on RANKL-mediated osteoclastogenesis and suggest the possible use of PO in treating bone disorders, such as osteopetrosis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Extratos Vegetais , Portulaca/química , Receptor Ativador de Fator Nuclear kappa-B , Animais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Extratos Vegetais/toxicidade , Receptor Ativador de Fator Nuclear kappa-B/efeitos dos fármacos , Receptor Ativador de Fator Nuclear kappa-B/metabolismo
13.
Handb Exp Pharmacol ; 223: 1035-54, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24961979

RESUMO

Ca(2+) signaling entails receptor-stimulated Ca(2+) release from the ER stores that serves as a signal to activate Ca(2+) influx channels present at the plasma membrane, the store-operated Ca(2+) channels (SOCs). The two known SOCs are the Orai and TRPC channels. The SOC-dependent Ca(2+) influx mediates and sustains virtually all Ca(2+)-dependent regulatory functions. The signal that transmits the Ca(2+) content of the ER stores to the plasma membrane is the ER resident, Ca(2+)-binding protein STIM1. STIM1 is a multidomain protein that clusters and dimerizes in response to Ca(2+) store depletion leading to activation of Orai and TRPC channels. Activation of the Orais by STIM1 is obligatory for their function as SOCs, while TRPC channels can function as both STIM1-dependent and STIM1-independent channels. Here we discuss the different mechanisms by which STIM1 activates the Orai and TRPC channels, the emerging specific and non-overlapping physiological functions of Ca(2+) influx mediated by the two channel types, and argue that the TRPC channels should be the preferred therapeutic target to control the toxic effect of excess Ca(2+) influx.


Assuntos
Canais de Cálcio/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Cálcio/metabolismo , Humanos , Proteína ORAI1 , Molécula 1 de Interação Estromal
14.
Int Immunopharmacol ; 136: 112284, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38823179

RESUMO

Cathepsin B (CTSB) and inflammatory cytokines are critical in initiating and developing pancreatitis. Calcineurin, a central calcium (Ca2+)-responsive signaling molecule, mediates acinar cell death and inflammatory responses leading to pancreatitis. However, the detailed mechanisms for regulating CTSB activity and inflammatory cytokine production are unknown. Myricetin (MC) exhibits various biological activities, including anti-inflammatory effects. Here, we aimed to investigate MC effects on pancreatitis and the underlying mechanisms. Prophylactic and therapeutic MC treatment ameliorated the severity of cerulein-, L-arginine-, and PDL-induced acute pancreatitis (AP). The inhibition of CTSB activity by MC was mediated via decreased calcineurin activity and macrophage infiltration, not neutrophils infiltration, into the pancreas. Additionally, calcineurin activity inhibition by MC prevented the phosphorylation of Ca2+/CaM-dependent protein kinase kinase 2 (CaMKK2) during AP, resulting in the inhibition of CaMKIV phosphorylation and adenosine monophosphate-activated protein kinase (AMPK) dephosphorylation. Furthermore, MC reduced nuclear factor-κB activation by modulating the calcineurin-CaMKIV-IKKα/ß-Iκ-Bα and calcineurin-AMPK-sirtuin1 axes, resulting in reduced production of tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6. Our results showed that MC alleviated AP severity by inhibiting acinar cell death and inflammatory responses, suggesting that MC as a calcineurin and CaMKK2 signaling modulator may be a potential treatment for AP.


Assuntos
Calcineurina , Catepsina B , Citocinas , Flavonoides , Camundongos Endogâmicos C57BL , Pancreatite , Animais , Pancreatite/tratamento farmacológico , Pancreatite/imunologia , Pancreatite/patologia , Pancreatite/induzido quimicamente , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Citocinas/metabolismo , Catepsina B/metabolismo , Camundongos , Masculino , Calcineurina/metabolismo , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/farmacologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Ceruletídeo , NF-kappa B/metabolismo , Pâncreas/patologia , Pâncreas/efeitos dos fármacos , Pâncreas/imunologia , Transdução de Sinais/efeitos dos fármacos , Arginina/metabolismo , Modelos Animais de Doenças , Proteínas Quinases Ativadas por AMP/metabolismo
15.
Traffic ; 12(2): 232-45, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21054717

RESUMO

Polarized Ca(2+) signals in secretory epithelial cells are determined by compartmentalized localization of Ca(2+) signaling proteins at the apical pole. Recently the ER Ca(2+) sensor STIM1 (stromal interaction molecule 1) and the Orai channels were shown to play a critical role in store-dependent Ca(2+) influx. STIM1 also gates the transient receptor potential-canonical (TRPC) channels. Here, we asked how cell stimulation affects the localization, recruitment and function of the native proteins in polarized cells. Inhibition of Orai1, STIM1, or deletion of TRPC1 reduces Ca(2+) influx and frequency of Ca(2+) oscillations. Orai1 localization is restricted to the apical pole of the lateral membrane. Surprisingly, cell stimulation does not lead to robust clustering of native Orai1, as is observed with expressed Orai1. Unexpectedly, cell stimulation causes polarized recruitment of native STIM1 to both the apical and lateral regions, thus to regions with and without Orai1. Accordingly, STIM1 and Orai1 show only 40% colocalization. Consequently, STIM1 shows higher colocalization with the basolateral membrane marker E-cadherin than does Orai1, while Orai1 showed higher colocalization with the tight junction protein ZO1. TRPC1 is expressed in both apical and basolateral regions of the plasma membrane. Co-IP of STIM1/Orai1/IP(3) receptors (IP(3) Rs)/TRPCs is enhanced by cell stimulation and disrupted by 2-aminoethoxydiphenyl borate (2APB). The polarized localization and recruitment of these proteins results in preferred Ca(2+) entry that is initiated at the apical pole. These findings reveal that in addition to Orai1, STIM1 likely regulates other Ca(2+) permeable channels, such as the TRPCs. Both channels contribute to the frequency of [Ca(2+) ] oscillations and thus impact critical cellular functions.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Glicoproteínas de Membrana/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/genética , Membrana Celular/metabolismo , Camundongos , Proteína ORAI1 , Transporte Proteico , Transdução de Sinais , Análise de Célula Única/métodos , Molécula 1 de Interação Estromal
16.
Exp Dermatol ; 22(11): 759-61, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24103002

RESUMO

Ultraviolet B (UVB) radiation induces photoageing by upregulating the expression of matrix metalloproteinases (MMPs) in human skin cells. Dihydroavenanthramide D (DHAvD) is a synthetic analog to naturally occurring avenanthramide, which is the active component in oats. Although anti-inflammatory, anti-atherosclerotic and antioxidant effects have been reported, the antiphotoageing effects of DHAvD are yet to be understood. In this study, we investigated the inhibitory effects of DHAvD on UVB-induced production of reactive oxygen species (ROS) and expression of MMPs, and its molecular mechanism in UVB-irradiated human dermal fibroblasts. Western blot and real-time PCR analyses revealed that DHAvD inhibited UVB-induced MMP-1 and MMP-3 expression. It also significantly blocked UVB-induced ROS generation in fibroblasts. Additionally, DHAvD attenuated UVB-induced phosphorylation of MAPKs, activation of NF-κB and AP-1. DHAvD regulates UVB-irradiated MMP expression by inhibiting ROS-mediated MAPK/NF-κB and AP-1 activation. DHAvD may be a useful candidate for preventing UV light-induced skin photoageing.


Assuntos
Fibroblastos/efeitos dos fármacos , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Envelhecimento da Pele/efeitos dos fármacos , Pele/efeitos dos fármacos , Pele/efeitos da radiação , ortoaminobenzoatos/química , Anti-Inflamatórios/química , Antioxidantes/química , Núcleo Celular/metabolismo , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Humanos , Fosforilação , Espécies Reativas de Oxigênio , Transdução de Sinais , Pele/metabolismo , Raios Ultravioleta
17.
Korean J Physiol Pharmacol ; 17(5): 427-33, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24227944

RESUMO

Receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis is accompanied by intracellular Ca(2+) mobilization in a form of oscillations, which plays essential roles by activating sequentially Ca(2+)/calmodulin-dependent protein kinase, calcineurin and NFATc1, necessary in the osteoclast differentiation. However, it is not known whether Ca(2+) mobilization which is evoked in RANKL-independent way induces to differentiate into osteoclasts. In present study, we investigated Ca(2+) mobilization induced by aluminum fluoride (AlF4 (-)), a G-protein activator, with or without RANKL and the effects of AlF4 (-) on the osteoclastogenesis in primary cultured mouse bone marrow-derived macrophages (BMMs). We show here that AlF4 (-) induces intracellular Ca(2+) concentration ([Ca(2+)]i) oscillations, which is dependent on extracellular Ca(2+) influx. Notably, co-stimulation of AlF4 (-) with RANKL resulted in enhanced NFATc1 expression and formation of tartrate-resistant acid phosphatase (TRAP) positive multinucleated cells. Additionally, we confirmed that mitogen-activated protein kinase (MAPK) is also activated by AlF4 (-). Taken together, these results demonstrate that G-protein would be a novel modulator responsible for [Ca(2+)]i oscillations and MAPK activation which lead to enhancement of RANKL-mediated osteoclastogenesis.

18.
Korean J Physiol Pharmacol ; 17(1): 65-71, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23440520

RESUMO

The transient receptor potential melastatin type 7 (TRPM7) channel is a widely expressed non-selective cation channel with fusion to the C-terminal alpha kinase domain and regarded as a key regulator of whole body Mg(2+) homeostasis in mammals. However, the roles of TRPM7 during osteoclastogenesis in RAW264.7 cells and bone marrow-derived monocyte/macrophage precursor cells (BMMs) are not clear. In the present study, we investigate the roles of TRPM7 in osteoclastogenesis using methods of small interfering RNA (siRNA), RT-PCR, patch-clamp, and calcium imaging. RANKL (receptor activator of NF-κB ligand) stimulation did not affect the TRPM7 expression and TRPM7-mediated current was activated in HEK293, RAW264.7, and BMM cells by the regulation of Mg(2+). Knock-down of TRPM7 by siTRPM7 reduced intracellular Ca(2+) concentration ([Ca(2+)](i)) increases by 0 mM [Mg(2+)](e) in HEK293 cells and inhibited the generation of RANKL-induced Ca(2+) oscillations in RAW264.7 cells. Finally, knock-down of TRPM7 suppressed RANKL-mediated osteoclastogenesis such as activation and translocation of NFATc1, formation of multinucleated cells, and the bone resorptive activity, sequentially. These results suggest that TRPM7 plays an essential role in the RANKL-induced [Ca(2+)](i) oscillations that triggers the late stages of osteoclastogenesis.

19.
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
20.
Tuberc Respir Dis (Seoul) ; 85(2): 147-154, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34847639

RESUMO

BACKGROUND: The expression of calcium signaling pathway molecules is altered in various carcinomas, which are related to the proliferation and altered characteristics of cancer cells. However, changes in calcium signaling in anti-cancer drugresistant cells (bearing a T790M mutation in epidermal growth factor receptor [EGFR]) remain unclear. METHODS: Afatinib-mediated changes in the level of store-operated Ca2+ entry (SOCE)-related proteins and intracellular Ca2+ level in non-small cell lung cancer cells with T790M mutation in the EGFR gene were analyzed using western blot and ratiometric assays, respectively. Afatinib-mediated autophagic flux was evaluated by measuring the cleavage of LC3B-II. Flow cytometry and cell proliferation assays were conducted to assess cell apoptosis and proliferation. RESULTS: The levels of SOCE-mediating proteins (ORAI calcium release-activated calcium modulator 1 [ORAI1], stromal interaction molecule 1 [STIM1], and sarco/endoplasmic reticulum Ca2+ ATPase [SERCA2]) decreased after afatinib treatment in non-small cell lung cancer cells, whereas the levels of SOCE-related proteins did not change in gefitinibresistant non-small cell lung cancer cells (PC-9/GR; bearing a T790M mutation in EGFR ). Notably, the expression level of SOCE-related proteins in PC-9/GR cells was reduced also responding to afatinib in the absence of extracellular Ca2+. Moreover, extracellular Ca2+ influx through the SOCE was significantly reduced in PC-9 cells pre-treated with afatinib than in the control group. Additionally, afatinib was found to decrease the level of SOCE-related proteins through autophagic degradation, and the proliferation of PC-9GR cells was significantly inhibited by a lack of extracellular Ca2+. CONCLUSION: Extracellular Ca2+ plays important role in afatinib-mediated autophagic degradation of SOCE-related proteins in cells with T790M mutation in the EGFR gene and extracellular Ca2+ is essential for determining anti-cancer drug efficacy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA