Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(5): 2389-2415, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38224453

RESUMO

DNA damage represents a challenge for cells, as this damage must be eliminated to preserve cell viability and the transmission of genetic information. To reduce or eliminate unscheduled chemical modifications in genomic DNA, an extensive signaling network, known as the DNA damage response (DDR) pathway, ensures this repair. In this work, and by means of a proteomic analysis aimed at studying the STIM1 protein interactome, we have found that STIM1 is closely related to the protection from endogenous DNA damage, replicative stress, as well as to the response to interstrand crosslinks (ICLs). Here we show that STIM1 has a nuclear localization signal that mediates its translocation to the nucleus, and that this translocation and the association of STIM1 to chromatin increases in response to mitomycin-C (MMC), an ICL-inducing agent. Consequently, STIM1-deficient cell lines show higher levels of basal DNA damage, replicative stress, and increased sensitivity to MMC. We show that STIM1 normalizes FANCD2 protein levels in the nucleus, which explains the increased sensitivity of STIM1-KO cells to MMC. This study not only unveils a previously unknown nuclear function for the endoplasmic reticulum protein STIM1 but also expands our understanding of the genes involved in DNA repair.


Assuntos
Núcleo Celular , Dano ao DNA , Molécula 1 de Interação Estromal , Cromatina/genética , Reparo do DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Mitomicina/farmacologia , Proteômica , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Humanos , Núcleo Celular/metabolismo , Proteínas de Neoplasias/metabolismo
2.
J Biol Chem ; 295(50): 17071-17082, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33023909

RESUMO

Stromal interaction molecule 1 (STIM1) plays a pivotal role in store-operated Ca2+ entry (SOCE), an essential mechanism in cellular calcium signaling and in maintaining cellular calcium balance. Because O-GlcNAcylation plays pivotal roles in various cellular function, we examined the effect of fluctuation in STIM1 O-GlcNAcylation on SOCE activity. We found that both increase and decrease in STIM1 O-GlcNAcylation impaired SOCE activity. To determine the molecular basis, we established STIM1-knockout HEK293 (STIM1-KO-HEK) cells using the CRISPR/Cas9 system and transfected STIM1 WT (STIM1-KO-WT-HEK), S621A (STIM1-KO-S621A-HEK), or T626A (STIM1-KO-T626A-HEK) cells. Using these cells, we examined the possible O-GlcNAcylation sites of STIM1 to determine whether the sites were O-GlcNAcylated. Co-immunoprecipitation analysis revealed that Ser621 and Thr626 were O-GlcNAcylated and that Thr626 was O-GlcNAcylated in the steady state but Ser621 was not. The SOCE activity in STIM1-KO-S621A-HEK and STIM1-KO-T626A-HEK cells was lower than that in STIM1-KO-WT-HEK cells because of reduced phosphorylation at Ser621 Treatment with the O-GlcNAcase inhibitor Thiamet G or O-GlcNAc transferase (OGT) transfection, which increases O-GlcNAcylation, reduced SOCE activity, whereas treatment with the OGT inhibitor ST045849 or siOGT transfection, which decreases O-GlcNAcylation, also reduced SOCE activity. Decrease in SOCE activity due to increase and decrease in O-GlcNAcylation was attributable to reduced phosphorylation at Ser621 These data suggest that both decrease in O-GlcNAcylation at Thr626 and increase in O-GlcNAcylation at Ser621 in STIM1 lead to impairment of SOCE activity through decrease in Ser621 phosphorylation. Targeting STIM1 O-GlcNAcylation could provide a promising treatment option for the related diseases, such as neurodegenerative diseases.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Acilação , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas de Neoplasias/genética , Fosforilação , Serina , Molécula 1 de Interação Estromal/genética
3.
Int J Mol Sci ; 21(18)2020 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-32916960

RESUMO

STIM1 is an endoplasmic reticulum (ER) protein that modulates the activity of a number of Ca2+ transport systems. By direct physical interaction with ORAI1, a plasma membrane Ca2+ channel, STIM1 activates the ICRAC current, whereas the binding with the voltage-operated Ca2+ channel CaV1.2 inhibits the current through this latter channel. In this way, STIM1 is a key regulator of Ca2+ signaling in excitable and non-excitable cells, and altered STIM1 levels have been reported to underlie several pathologies, including immunodeficiency, neurodegenerative diseases, and cancer. In both sporadic and familial Alzheimer's disease, a decrease of STIM1 protein levels accounts for the alteration of Ca2+ handling that compromises neuronal cell viability. Using SH-SY5Y cells edited by CRISPR/Cas9 to knockout STIM1 gene expression, this work evaluated the molecular mechanisms underlying the cell death triggered by the deficiency of STIM1, demonstrating that STIM1 is a positive regulator of ITPR3 gene expression. ITPR3 (or IP3R3) is a Ca2+ channel enriched at ER-mitochondria contact sites where it provides Ca2+ for transport into the mitochondria. Thus, STIM1 deficiency leads to a strong reduction of ITPR3 transcript and ITPR3 protein levels, a consequent decrease of the mitochondria free Ca2+ concentration ([Ca2+]mit), reduction of mitochondrial oxygen consumption rate, and decrease in ATP synthesis rate. All these values were normalized by ectopic expression of ITPR3 in STIM1-KO cells, providing strong evidence for a new mode of regulation of [Ca2+]mit mediated by the STIM1-ITPR3 axis.


Assuntos
Sinalização do Cálcio , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mitocôndrias/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Técnicas de Inativação de Genes , Humanos , Proteínas de Neoplasias/genética , Molécula 1 de Interação Estromal/genética
4.
Sci Rep ; 10(1): 6580, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313105

RESUMO

Tumor invasion requires efficient cell migration, which is achieved by the generation of persistent and polarized lamellipodia. The generation of lamellipodia is supported by actin dynamics at the leading edge where a complex of proteins known as the WAVE regulatory complex (WRC) promotes the required assembly of actin filaments to push the front of the cell ahead. By using an U2OS osteosarcoma cell line with high metastatic potential, proven by a xenotransplant in zebrafish larvae, we have studied the role of the plasma membrane Ca2+ channel ORAI1 in this process. We have found that epidermal growth factor (EGF) triggered an enrichment of ORAI1 at the leading edge, where colocalized with cortactin (CTTN) and other members of the WRC, such as CYFIP1 and ARP2/3. ORAI1-CTTN co-precipitation was sensitive to the inhibition of the small GTPase RAC1, an upstream activator of the WRC. RAC1 potentiated ORAI1 translocation to the leading edge, increasing the availability of surface ORAI1 and increasing the plasma membrane ruffling. The role of ORAI1 at the leading edge was studied in genetically engineered U2OS cells lacking ORAI1 expression that helped us to prove the key role of this Ca2+ channel on lamellipodia formation, lamellipodial persistence, and cell directness, which are required for tumor cell invasiveness in vivo.


Assuntos
Cortactina/genética , Proteína ORAI1/genética , Osteossarcoma/genética , Pseudópodes/genética , Proteínas rac1 de Ligação ao GTP/genética , Citoesqueleto de Actina/genética , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular/genética , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Pseudópodes/metabolismo
5.
J Mol Med (Berl) ; 96(10): 1061-1079, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30088035

RESUMO

STIM1 is an endoplasmic reticulum protein with a role in Ca2+ mobilization and signaling. As a sensor of intraluminal Ca2+ levels, STIM1 modulates plasma membrane Ca2+ channels to regulate Ca2+ entry. In neuroblastoma SH-SY5Y cells and in familial Alzheimer's disease patient skin fibroblasts, STIM1 is cleaved at the transmembrane domain by the presenilin-1-associated γ-secretase, leading to dysregulation of Ca2+ homeostasis. In this report, we investigated expression levels of STIM1 in brain tissues (medium frontal gyrus) of pathologically confirmed Alzheimer's disease patients, and observed that STIM1 protein expression level decreased with the progression of neurodegeneration. To study the role of STIM1 in neurodegeneration, a strategy was designed to knock-out the expression of STIM1 gene in the SH-SY5Y neuroblastoma cell line by CRISPR/Cas9-mediated genome editing, as an in vitro model to examine the phenotype of STIM1-deficient neuronal cells. It was proved that, while STIM1 is not required for the differentiation of SH-SY5Y cells, it is absolutely essential for cell survival in differentiating cells. Differentiated STIM1-KO cells showed a significant decrease of mitochondrial respiratory chain complex I activity, mitochondrial inner membrane depolarization, reduced mitochondrial free Ca2+ concentration, and higher levels of senescence as compared with wild-type cells. In parallel, STIM1-KO cells showed a potentiated Ca2+ entry in response to depolarization, which was sensitive to nifedipine, pointing to L-type voltage-operated Ca2+ channels as mediators of the upregulated Ca2+ entry. The stable knocking-down of CACNA1C transcripts restored mitochondrial function, increased mitochondrial Ca2+ levels, and dropped senescence to basal levels, demonstrating the essential role of the upregulation of voltage-operated Ca2+ entry through Cav1.2 channels in STIM1-deficient SH-SY5Y cell death. KEY MESSAGES: STIM1 protein expression decreases with the progression of neurodegeneration in Alzheimer's disease. STIM1 is essential for cell viability in differentiated SH-SY5Y cells. STIM1 deficiency triggers voltage-regulated Ca2+ entry-dependent cell death. Mitochondrial dysfunction and senescence are features of STIM1-deficient differentiated cells.


Assuntos
Doença de Alzheimer/genética , Canais de Cálcio Tipo L/fisiologia , Cálcio/fisiologia , Proteínas de Neoplasias/fisiologia , Molécula 1 de Interação Estromal/fisiologia , Idoso , Idoso de 80 Anos ou mais , Morte Celular , Linhagem Celular Tumoral , Humanos , Córtex Pré-Frontal/fisiologia
6.
Cell Signal ; 40: 44-52, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28866365

RESUMO

STIM1, the endoplasmic reticulum Ca2+ sensor that modulates the activity of plasma membrane Ca2+ channels, becomes phosphorylated at ERK1/2 target sites during Ca2+ store depletion triggered by thapsigargin or epidermal growth factor (EGF). This ERK1/2-dependent phosphorylation regulates STIM1 localization and dissociation from microtubules, and it is known that enhances the binding to ORAI1, a store-operated Ca2+ entry (SOCE) channel, leading to the activation of this Ca2+ influx pathway. However, there remained some evidence of a role for SOCE in the activation of ERK1/2, and here we assessed the contribution of SOCE to ERK1/2 activation by generating a STIM1-deficient cell line by CRISPR/Cas9 genome editing of the STIM1 locus in prostate cancer PC3 cells. The genomic modification consisted of a 16 base-pair insertion in exon 5 of both alleles, therefore abrogating STIM1 synthesis. STIM1-KO cells did show a striking decrease in Ca2+ influx in response to thapsigargin or EGF, a result that demonstrates that SOCE mediates Ca2+ entry in PC3 cells during stimulation with EGF. Moreover, identical levels of total ERK1/2 were found in STIM1-KO cells and the parental cell line, and ERK1/2 activation was fully activated in KO cells, both in the presence and in the absence of extracellular Ca2+, a result that supports that STIM1 and SOCE are not required for ERK1/2 activation. This activation was sensitive to Src kinase inhibition, but not to CAMKII nor PKC inhibition, a result that sets STIM1 and SOCE as downstream targets of the axis Src-Raf-MEK-ERK, rather than upstream regulators.


Assuntos
Canais de Cálcio/genética , Cálcio/metabolismo , Proteínas de Membrana/genética , Neoplasias da Próstata/genética , Molécula 1 de Interação Estromal/genética , Sistemas CRISPR-Cas/genética , Canais de Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Técnicas de Inativação de Genes , Humanos , Proteínas Sensoras de Cálcio Intracelular , Sistema de Sinalização das MAP Quinases/genética , Masculino , Microtúbulos/genética , Microtúbulos/metabolismo , Neoplasias da Próstata/patologia , Molécula 1 de Interação Estromal/metabolismo , Quinases da Família src/antagonistas & inibidores
7.
Sci Rep ; 7(1): 383, 2017 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-28341841

RESUMO

Cell motility and migration requires the reorganization of the cortical cytoskeleton at the leading edge of cells and extracellular Ca2+ entry is essential for this reorganization. However the molecular nature of the regulators of this pathway is unknown. This work contributes to understanding the role of STIM1 and ORAI1 in the promotion of membrane ruffling by showing that phospho-STIM1 localizes at the leading edge of cells, and that both phospho-STIM1 and ORAI1 co-localize with cortactin (CTTN), a regulator of the cytoskeleton at membrane ruffling areas. STIM1-KO and ORAI1-KO cell lines were generated by CRISPR/Cas9 genome editing in U2OS cells. In both cases, KO cells presented a notable reduction of store-operated Ca2+ entry (SOCE) that was rescued by expression of STIM1-mCherry and ORAI1-mCherry. These results demonstrated that SOCE regulates membrane ruffling at the leading edge of cells. Moreover, endogenous ORAI1 and overexpressed ORAI1-GFP co-immunoprecipitated with endogenous CTTN. This latter result, in addition to the KO cells' phenotype, the preservation of ORAI1-CTTN co-localization during ruffling, and the inhibition of membrane ruffling by the Ca2+-channel inhibitor SKF96365, further supports a functional link between SOCE and membrane ruffling.


Assuntos
Sinalização do Cálcio , Membrana Celular/metabolismo , Movimento Celular , Cortactina/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Animais , Linhagem Celular , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos
8.
Cell Signal ; 27(3): 545-54, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25562429

RESUMO

STIM1 is a Ca(2+) sensor of the endoplasmic reticulum (ER) that triggers the activation of plasma membrane Ca(2+) channels upon depletion of Ca(2+) levels within the ER. During thapsigargin-triggered Ca(2+) store depletion, ERK1/2 phosphorylates STIM1 at Ser575, Ser608, and Ser621. This phosphorylation plays a role in the regulation of STIM1 dissociation from the microtubule plus-end binding protein EB1, an essential step for STIM1 activation by thapsigargin. However, little is known regarding the physiological role of this phosphorylation. Because IGF-1 triggers the activation of the RAF-MEK-ERK and the phosphoinositide pathways, the role of STIM1 phosphorylation in IGF-1 stimulation was studied. There was found to be phosphorylation of ERK1/2 in both the presence and the absence of extracellular Ca(2+), demonstrating that Ca(2+) influx is not essential for ERK1/2 activation. In parallel, IGF-1 triggered STIM1 phosphorylation at the aforementioned sites, an effect that was blocked by PD0325901, a MEK1/2 inhibitor used to block ERK1/2 activation. Also, STIM1-GFP was found in clusters upon IGF-1 stimulation, and STIM1-S575A/S608A/S621A-GFP strongly reduced this multimerization. Interestingly, phospho-STIM1 was mainly found in clusters when cells were treated with IGF-1, and IGF-1 triggered the dissociation of STIM1 from EB1, similarly to what has been observed for thapsigargin, suggesting that STIM1 mediates the IGF-1 signaling pathway. A study of IGF-1-stimulated NFAT translocation was therefore performed, finding that STIM1-S575A/S608A/S621A blocked this translocation, as did the fusion protein STIM1-EB1, confirming that both STIM1 phosphorylation and STIM1-EB1 dissociation are required for IGF-1-triggered Ca(2+)-dependent signaling, and demonstrating that STIM1 phosphorylation plays a role as a downstream effector of the RAF-MEK-ERK pathway and an upstream activator of Ca(2+) entry.


Assuntos
Fator de Crescimento Insulin-Like I/farmacologia , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Benzamidas/farmacologia , Cálcio/metabolismo , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Proteínas de Neoplasias/genética , Fosforilação/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Molécula 1 de Interação Estromal , Tapsigargina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Biochim Biophys Acta ; 1853(1): 233-43, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25447552

RESUMO

STIM1 is a key regulator of store-operated calcium entry (SOCE), and therefore a mediator of Ca²âº entry-dependent cellular events. Phosphorylation of STIM1 at ERK1/2 target sites has been described as enhancing STIM1 activation during intracellular Ca²âº emptying triggered by the inhibition of the sarco(endo)plasmic Ca²âº -ATPase with thapsigargin. However, no physiological function is known for this specific phosphorylation. The present study examined the role of STIM1 phosphorylation in cell signaling triggered by EGF. Using a human endometrial adenocarcinoma cell line (Ishikawa cells) EGF or H-Ras(G12V), an active mutant of H-Ras, was found to trigger STIM1 phosphorylation at residues Ser575, Ser608, and Ser621, and this process was sensitive to PD0325901, an inhibitor of ERK1/2. Both, ERK1/2 activation and STIM1 phosphorylation took place in the absence of extracellular Ca²âº, indicating that both events are upstream steps for Ca²âºentry activation. Also, EGF triggered the dissociation of STIM1 from EB1 (a regulator of microtubule plus-ends) in a manner similar to that reported for the activation of STIM1 by thapsigargin. Migration of the Ishikawa cells was impaired when STIM1 phosphorylation was targeted by Ser-to-Ala substitution mutation of ERK1/2 target sites. This effect was also observed with the Ca²âº channel blocker SKF96365. Phosphomimetic mutation of STIM1 restored the migration to levels similar to that found for STIM1-wild type. Finally, the increased vimentin expression and relocalization of E-cadherin triggered by EGF were largely inhibited by targeting STIM1 phosphorylation, while STIM1-S575E/S608E/S621E normalized the profiles of these two EMT markers.


Assuntos
Movimento Celular , Fator de Crescimento Epidérmico/farmacologia , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Benzamidas/farmacologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Humanos , Imidazóis/farmacologia , Fosforilação , Molécula 1 de Interação Estromal
10.
J Cell Sci ; 126(Pt 14): 3170-80, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23687376

RESUMO

STIM1 (stromal interaction molecule 1) is a key regulator of store-operated calcium entry (SOCE). Upon depletion of Ca(2+) concentration within the endoplasmic reticulum (ER), STIM1 relocalizes at ER-plasma membrane junctions, activating store-operated calcium channels (SOCs). Although the molecular details for STIM1-SOC binding is known, the regulation of SOCE remains largely unknown. A detailed list of phosphorylated residues within the STIM1 sequence has been reported. However, the molecular pathways controlling this phosphorylation and its function are still under study. Using phosphospecific antibodies, we demonstrate that ERK1/2 mediates STIM1 phosphorylation at Ser575, Ser608 and Ser621 during Ca(2+) store depletion, and that Ca(2+) entry and store refilling restore phosphorylation to basal levels. This phosphorylation occurs in parallel to the dissociation from end-binding protein 1 (EB1), a regulator of growing microtubule ends. Although Ser to Ala mutation of residues 575, 608 and 621 showed a constitutive binding to EB1 even after Ca(2+) store depletion, Ser to Glu mutation of these residues (to mimic the phosphorylation profile attained after store depletion) triggered full dissociation from EB1. Given that wild-type STIM1 and STIM1(S575E/S608E/S621E) activate SOCE similarly, a model is proposed to explain how ERK1/2-mediated phosphorylation of STIM1 regulates SOCE. This regulation is based on the phosphorylation of STIM1 to trigger dissociation from EB1 during Ca(2+) store depletion, an event that is fully reversed by Ca(2+) entry and store refilling.


Assuntos
Canais de Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/fisiologia , Proteínas de Neoplasias/metabolismo , Cálcio/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/genética , Microscopia Confocal , Mutação/genética , Proteínas de Neoplasias/genética , Fosforilação/genética , Ligação Proteica/genética , Molécula 1 de Interação Estromal , Transgenes/genética
11.
J Cell Sci ; 123(Pt 18): 3084-93, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20736304

RESUMO

Store-operated calcium entry (SOCE) is an important Ca2+ entry pathway that regulates many cell functions. Upon store depletion, STIM1, a transmembrane protein located in the endoplasmic reticulum (ER), aggregates and relocates close to the plasma membrane (PM) where it activates store-operated calcium channels (SOCs). Although STIM1 was early defined as a phosphoprotein, the contribution of the phosphorylation has been elusive. In the present work, STIM1 was found to be a target of extracellular-signal-regulated kinases 1 and 2 (ERK1/2) in vitro, and we have defined the ERK1/2-phosphorylated sites on the STIM1 sequence. Using HEK293 cells stably transfected for the expression of tagged STIM1, we found that alanine substitution mutants of ERK1/2 target sites reduced SOCE significantly, suggesting that phosphorylation of these residues are required to fully accomplish SOCE. Indeed, the ERK1/2 inhibitors PD184352 and PD0325901 decreased SOCE in transfected cells. Conversely, 12-O-tetradecanoylphorbol-13-acetate, which activates ERK1/2, enhanced SOCE in cells expressing wild-type tagged STIM1, but did not potentiate Ca2+ influx in cells expressing serine to alanine mutations in ERK1/2 target sites of STIM1. Alanine substitution mutations decreased Ca2+ influx without disturbing the aggregation of STIM1 upon store depletion and without affecting the relocalization in ER-PM punctae. However, our results suggest that STIM1 phosphorylation at ERK1/2 target sites can modulate SOCE by altering STIM1 binding to SOCs, because a significant decrease in FRET efficiency was observed between alanine substitution mutants of STIM1-GFP and ORAI1-CFP.


Assuntos
Cálcio/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Transporte Biológico , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Humanos , Proteínas de Membrana/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Fosforilação , Molécula 1 de Interação Estromal
12.
Reproduction ; 138(2): 211-21, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19470709

RESUMO

Calcium waves represent one of the most important intracellular signaling events in oocytes at fertilization required for the exit from metaphase arrest and the resumption of the cell cycle. The molecular mechanism ruling this signaling has been described in terms of the contribution of intracellular calcium stores to calcium spikes. In this work, we considered the possible contribution of store-operated calcium entry (SOCE) to this signaling, by studying the localization of the protein STIM1 in oocytes. STIM1 has been suggested to play a key role in the recruitment and activation of plasma membrane calcium channels, and we show here that mature mouse oocytes express this protein distributed in discrete clusters throughout their periphery in resting cells, colocalizing with the endoplasmic reticulum marker calreticulin. However, immunolocalization of the endogenous STIM1 showed considerable redistribution over larger areas or patches covering the entire periphery of the oocyte during Ca(2+) store depletion induced with thapsigargin or ionomycin. Furthermore, pharmacological activation of endogenous phospholipase C induced a similar pattern of redistribution of STIM1 in the oocyte. Finally, fertilization of mouse oocytes revealed a significant and rapid relocalization of STIM1, similar to that found after pharmacological Ca(2+) store depletion. This particular relocalization supports a role for STIM1 and SOCE in the calcium signaling during early stages of fertilization.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Fertilização/fisiologia , Glicoproteínas de Membrana/análise , Oócitos/metabolismo , Animais , Transporte Biológico , Biomarcadores/análise , Canais de Cálcio/metabolismo , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Calreticulina/análise , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Retículo Endoplasmático/metabolismo , Ativação Enzimática , Feminino , Fertilização in vitro , Imunofluorescência , Ionomicina/farmacologia , Ionóforos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Molécula 1 de Interação Estromal , Tapsigargina/farmacologia , Fosfolipases Tipo C/metabolismo
13.
Reprod Biomed Online ; 17(5): 652-61, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18983749

RESUMO

The adverse effects of reactive oxygen species (ROS) on many aspects of reproduction are well documented. However, much less is known regarding the contribution of culture media to the oxidative stress of gametes during assisted reproductive techniques. This study measured the generation of ROS by culture media during IVF procedures and its effects on human oocytes. Commercially supplied culture media generated ROS at various rates, depending on the composition, whereas follicular fluid generated ROS at a much lower level. The incubation of cumulus-oocyte complexes (COC) in culture media induced marked lipid peroxidation compared with levels found in freshly retrieved COC. This plasma membrane damage, measured with the quenching of cis-parinaric acid fluorescence assay, was attenuated by supplementation of the medium with alpha-tocopherol or catalase. Moreover, there was an association between ROS production by culture medium and thiolic content consumption within the oocytes, suggesting that the intracellular reduced glutathione pool was partially depleted during in-vitro manipulation. The results show that culture medium could damage oocytes (and consequently embryo development) depending on their composition, and it is proposed that current IVF protocols could be revised in order to decrease ROS generation.


Assuntos
Meios de Cultura/efeitos adversos , Oócitos/metabolismo , Estresse Oxidativo , Técnicas de Reprodução Assistida/efeitos adversos , Adulto , Meios de Cultura/química , Células do Cúmulo/metabolismo , Feminino , Glutationa/metabolismo , Humanos , Técnicas In Vitro , Peroxidação de Lipídeos , Espécies Reativas de Oxigênio/metabolismo
14.
Antioxid Redox Signal ; 10(1): 31-42, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17956188

RESUMO

Hydrogen sulfide (H(2)S) concentration can be maintained in cell cultures within the range reported for rat brain by repetitive pulses of sodium hydrogen sulfide. Less than 2 h exposure to H(2)S concentrations within 50 and 120 microM (i.e., within the upper segment of the reported physiological range of H(2)S in rat brain), produces a large shift of the intracellular calcium homeostasis in cerebellar granule neurons (CGN) in culture, leading to a large and sustained increase of cytosolic calcium concentration. Only 1 h exposure to H(2)S concentrations within 100 and 300 microM raises intracellular calcium to the neurotoxic range, with nearly 50% cell death after 2 h. L-type Ca(2+) channels antagonists nimodipine and nifedipine block both the H(2)S-induced rise of cytosolic calcium and cell death. The N-methyl-D-aspartate receptor antagonists (+)-MK-801 and DL-2-amino-5-phosphonovaleric acid afforded a nearly complete protection against H(2)S-induced CGN death and largely attenuated the rise of cytosolic calcium. Thus, H(2)S-induced rise of cytosolic calcium eventually reaches the neurotoxic cytosolic calcium range, leading to glutamate-induced excitotoxic CGN death. The authors conclude that H(2)S is a major modulator of calcium homeostasis in neurons as it induces activation of Ca(2+) entry through L-type Ca(2+) channels, and thereby of neuronal activity.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Citosol/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , 2-Amino-5-fosfonovalerato/farmacologia , Animais , Sequência de Bases , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Cerebelo/citologia , Cerebelo/enzimologia , Cerebelo/metabolismo , Cistationina beta-Sintase/genética , Cistationina gama-Liase/genética , Citosol/metabolismo , Primers do DNA , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Nifedipino/farmacologia , Nimodipina/farmacologia , RNA Mensageiro/genética , Ratos , Ratos Wistar
15.
Biochemistry ; 45(11): 3794-804, 2006 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-16533063

RESUMO

Exposure of myosin subfragment 1 (S1) to 3-morpholinosydnonimine (SIN-1) produced a time-dependent inhibition of the F-actin-stimulated S1 Mg(2+)-ATPase activity, reaching 50% inhibition with 46.7 +/- 8.3 microM SIN-1 for 8.7 microM S1, that is, at a SIN-1/S1 molar ratio of approximately 5.5. The inhibition was due to the peroxynitrite produced by SIN-1 decomposition because (1) decomposed SIN-1 was found to have no effect on S1 ATPase activity, (2) addition of SIN-1 in the presence of superoxide dismutase and catalase fully prevented inhibition by SIN-1, and (3) micromolar pulses of chemically synthesized peroxynitrite produced inhibition of F-actin-stimulated S1 Mg(2+)-ATPase activity. In parallel, SIN-1 produced the inhibition of the nonphysiological Ca(2+)-dependent and K(+)/EDTA-dependent S1 ATPase activity of S1 and, therefore, suggested that the inhibition of F-actin-stimulated S1 Mg(2+)-ATPase activity is produced by the oxidation of highly reactive cysteines of S1 (Cys(707) and Cys(697)), located close to the catalytic center. This point was further confirmed by the titration of S1 cysteines with 5,5'-dithiobis(2-nitrobenzoic acid) and by the parallel decrease of Cys(707) labeling by 5-(iodoacetamido)fluorescein, and it was reinforced by the fact that other common protein modifications produced by peroxynitrite, for example, protein carbonyl and nitrotyrosine formation, were barely detected at the concentrations of SIN-1 that produced more than 50% inhibition of the F-actin-stimulated S1 Mg(2+)-ATPase activity. Differential scanning calorimetry of S1 (untreated and treated with different SIN-1 concentrations) pointed out that SIN-1, at concentrations that generate micromolar peroxynitrite fluxes, impaired the ability of ADP.V(1) to induce the intermediate catalytic transition state and also produced the partial unfolding of S1 that leads to an enhanced susceptibility of S1 to trypsin digestion, which can be fully protected by 2 mM GSH.


Assuntos
Adenosina Trifosfatases/metabolismo , Subfragmentos de Miosina/antagonistas & inibidores , Subfragmentos de Miosina/metabolismo , Ácido Peroxinitroso/farmacologia , Actinas/metabolismo , Difosfato de Adenosina/metabolismo , Animais , Catálise , Cisteína/metabolismo , Relação Dose-Resposta a Droga , Estabilidade Enzimática , Magnésio/metabolismo , Magnésio/farmacologia , Molsidomina/análogos & derivados , Molsidomina/metabolismo , Molsidomina/farmacologia , Oxirredução , Ácido Peroxinitroso/metabolismo , Dobramento de Proteína , Coelhos , Miosinas de Músculo Esquelético/antagonistas & inibidores , Temperatura , Fatores de Tempo , Tirosina/análogos & derivados , Tirosina/metabolismo
16.
J Neurosci Methods ; 149(1): 82-9, 2005 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-15975662

RESUMO

While the intraluminal thread technique to induce middle cerebral artery occlusion is widely used in animal models of focal cerebral ischemia, it has several drawbacks. The present study describes a new technique involving transfemoral selective intraluminal wiring, and evaluates its technical feasibility, effectiveness, and safety. Twenty-four Wistar rats were used in this work: two for a vascular anatomy study and 22 subjected to middle cerebral artery occlusion for 1 h by our new transfemoral selective "intraluminal wiring" technique. After 24 h of reperfusion, the animals were evaluated neurologically, and then were sacrificed. Macroscopic, histological (2,3,5-triphenyltetrazolium chloride (TTC), hematoxylin-eosin and TUNEL), and biochemical (DNA fragmentation and caspase-3 activity) studies were performed to assess the extent of brain damage produced by focal ischemia. Technical success was obtained in all 22 animals. Signs of focal ischemia and reperfusion, such as necrosis and apoptosis, were detected in the middle cerebral artery territory. No subarachnoid hemorrhage was noticed in any animal. Transfemoral selective intraluminal wiring appears to be a reliable, safe, and minimally invasive technique to induce transient focal cerebral ischemia in rats.


Assuntos
Arteriopatias Oclusivas/complicações , Arteriopatias Oclusivas/diagnóstico , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/etiologia , Artéria Cerebral Média/cirurgia , Procedimentos Cirúrgicos Vasculares/instrumentação , Procedimentos Cirúrgicos Vasculares/métodos , Animais , Cateterismo Periférico/efeitos adversos , Cateterismo Periférico/métodos , Modelos Animais de Doenças , Artéria Femoral/cirurgia , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Fatores de Tempo , Procedimentos Cirúrgicos Vasculares/efeitos adversos
17.
Biochim Biophys Acta ; 1711(1): 33-40, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15904661

RESUMO

In this paper, we show further evidences for the existence of store-operated calcium entry in differentiated skeletal muscle C2C12 myotubes after Ca2+ depletion in sarcoplasmic reticulum, using thapsigargin, a potent sarcoplasmic reticulum Ca2+-ATPase inhibitor, caffeine as ryanodine receptor activator, and ATP which activates purinergic receptors. The quenching of fura 2 fluorescence emission by Mn2+ also provided evidences for store-operated calcium entry because this quenching was accelerated when sarcoplasmic reticulum was depleted of Ca2+. Ca2+ entry was sensitive to Ni2+, La3+, Gd3+ and 2-aminoethyl diphenyl borate but resistant to nifedipine, thus excluding L-type Ca2+ channels in this type of calcium entry. Our data obtained using ATP for store depletion suggest that the level of Ca2+ in internal stores could play a role in the regulation of store-operated calcium channel activity in this cell type.


Assuntos
Cálcio/metabolismo , Músculo Esquelético/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cafeína/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Estimulantes do Sistema Nervoso Central/farmacologia , Humanos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Free Radic Biol Med ; 32(1): 46-55, 2002 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11755316

RESUMO

A sustained increase of intracellular free [Ca(2+)] ([Ca(2+)](i)) has been shown to be an early event of neuronal cell death induced by peroxynitrite (ONOO(-)). In this paper, chronic exposure to ONOO(-) has been simulated by treatment of rat brain synaptosomes or plasma membrane vesicles with repetitive pulses of ONOO(-) during at most 50 min, which efficiently produced nitrotyrosine formation in several membrane proteins (including the Ca(2+)-ATPase). The plasma membrane Ca(2+)-ATPase activity at near-physiological conditions (pH 7, submicromolar Ca(2+), and millimolar Mg(2+)-ATP concentrations), which plays a major role in the control of synaptic [Ca(2+)](i), can be more than 75% inhibited by a sustained exposure to micromolar ONOO(-) (e.g., to 100 pulses of 10 microM ONOO(-)). This inhibition is irreversible and mostly due to a decreased V(max), and to the 2-fold increase of the K(0.5) for Ca(2+) stimulation and about 5-fold increase of the K(M) for Mg(2+)-ATP. [Ca(2+)](i) increases to >400 nM when synaptosomes are subjected to this treatment. Reduced glutathione can afford only partial protection against the inhibition produced by micromolar ONOO(-) pulses. Therefore, inhibition of the plasma membrane Ca(2+)-pump activity during chronic exposure to ONOO(-) may account by itself for a large and sustained increase of intracellular [Ca(2+)](i) in synaptic nerve terminals.


Assuntos
ATPases Transportadoras de Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Ácido Peroxinitroso/farmacologia , Sinaptossomos/efeitos dos fármacos , Animais , Encéfalo/citologia , Encéfalo/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/fisiologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Ratos , Ratos Wistar , Sinaptossomos/metabolismo , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA