Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 33(2): ar20, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34985939

RESUMO

Positioning organelles at the right place and time is critical for their function and inheritance. In budding yeast, mitochondrial and nuclear positioning require the anchoring of mitochondria and dynein to the cell cortex by clusters of Num1. We have previously shown that mitochondria drive the assembly of cortical Num1 clusters, which then serve as anchoring sites for mitochondria and dynein. When mitochondrial inheritance is inhibited, mitochondrial-driven assembly of Num1 in buds is disrupted and defects in dynein-mediated spindle positioning are observed. Using a structure-function approach to dissect the mechanism of mitochondria-dependent dynein anchoring, we found that the EF hand-like motif (EFLM) of Num1 and its ability to bind calcium are required to bias dynein anchoring on mitochondria-associated Num1 clusters. Consistently, when the EFLM is disrupted, we no longer observe defects in dynein activity following inhibition of mitochondrial inheritance. Thus, the Num1 EFLM functions to bias dynein anchoring and activity in nuclear inheritance subsequent to mitochondrial inheritance. We hypothesize that this hierarchical integration of organelle positioning pathways by the Num1 EFLM contributes to the regulated order of organelle inheritance during the cell cycle.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Motivos EF Hand/fisiologia , Proteínas de Saccharomyces cerevisiae/metabolismo , Transporte Biológico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas do Citoesqueleto/fisiologia , Dineínas/metabolismo , Motivos EF Hand/genética , Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Organelas/fisiologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Fuso Acromático/metabolismo
2.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-34830487

RESUMO

Neuronal calcium sensor-1 (NCS-1) is a four-EF-hand ubiquitous signaling protein modulating neuronal function and survival, which participates in neurodegeneration and carcinogenesis. NCS-1 recognizes specific sites on cellular membranes and regulates numerous targets, including G-protein coupled receptors and their kinases (GRKs). Here, with the use of cellular models and various biophysical and computational techniques, we demonstrate that NCS-1 is a redox-sensitive protein, which responds to oxidizing conditions by the formation of disulfide dimer (dNCS-1), involving its single, highly conservative cysteine C38. The dimer content is unaffected by the elevation of intracellular calcium levels but increases to 10-30% at high free zinc concentrations (characteristic of oxidative stress), which is accompanied by accumulation of the protein in punctual clusters in the perinuclear area. The formation of dNCS-1 represents a specific Zn2+-promoted process, requiring proper folding of the protein and occurring at redox potential values approaching apoptotic levels. The dimer binds Ca2+ only in one EF-hand per monomer, thereby representing a unique state, with decreased α-helicity and thermal stability, increased surface hydrophobicity, and markedly improved inhibitory activity against GRK1 due to 20-fold higher affinity towards the enzyme. Furthermore, dNCS-1 can coordinate zinc and, according to molecular modeling, has an asymmetrical structure and increased conformational flexibility of the subunits, which may underlie their enhanced target-binding properties. In HEK293 cells, dNCS-1 can be reduced by the thioredoxin system, otherwise accumulating as protein aggregates, which are degraded by the proteasome. Interestingly, NCS-1 silencing diminishes the susceptibility of Y79 cancer cells to oxidative stress-induced apoptosis, suggesting that NCS-1 may mediate redox-regulated pathways governing cell death/survival in response to oxidative conditions.


Assuntos
Sinalização do Cálcio/genética , Receptor Quinase 1 Acoplada a Proteína G/genética , Neoplasias/genética , Proteínas Sensoras de Cálcio Neuronal/genética , Neurônios/metabolismo , Neuropeptídeos/genética , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Dimerização , Dissulfetos/química , Motivos EF Hand/genética , Células HEK293 , Humanos , Cinética , Neoplasias/patologia , Proteínas Sensoras de Cálcio Neuronal/antagonistas & inibidores , Neurônios/química , Neuropeptídeos/antagonistas & inibidores , Oxirredução , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética , Zinco/metabolismo
3.
Cell Calcium ; 80: 46-55, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30953998

RESUMO

Two highly conserved structural motifs observed in members of the EF-hand family of calcium binding proteins. The motifs provide a supporting scaffold for the Ca2+ binding loops and contribute to the hydrophobic core of the EF-hand domain. Each structural motif represents a cluster of three amino acids called cluster I ('black' cluster) and cluster II ('grey' cluster). Cluster I is more conserved and mostly incorporates aromatic amino acids. In contrast, cluster II is noticeably less conserved and includes a mix of aromatic, hydrophobic, and polar amino acids of different sizes. In the human calcium binding S100 P protein, these 'black' and 'gray' clusters include residues F15, F71, and F74 and L33, L58, and K30, respectively. To evaluate the effects of these clusters on structure and functionality of human S100 P, we have performed Ala scanning. The resulting mutants were studied by a multiparametric approach that included circular dichroism, scanning calorimetry, dynamic light scattering, chemical crosslinking, and fluorescent probes. Spectrofluorimetric Ca2+-titration of wild type S100 P showed that S100 P dimer has 1-2 strong calcium binding sites (K1 = 4 × 106 M-1) and two cooperative low affinity (K2 = 4 × 104 M-1) binding sites. Similarly, the S100 P mutants possess two types of calcium binding sites. This analysis revealed that the alanine substitutions in the clusters I and II caused comparable changes in the S100 P functional properties. However, analysis of heat- or GuHCl-induced unfolding of these proteins showed that the alanine substitutions in the cluster I caused notably more pronounced decrease in the protein stability compared to the changes caused by alanine substitutions in the cluster II. Opposite to literature data, the F15 A substitution did not cause the S100 P dimer dissociation, indicating that F15 is not crucial for dimer stability. Overall, similar to parvalbumins, the S100 P cluster I is more important for protein conformational stability than the cluster II.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Sequência de Aminoácidos , Aminoácidos Aromáticos/genética , Sítios de Ligação/genética , Proteínas de Ligação ao Cálcio/genética , Dicroísmo Circular , Difusão Dinâmica da Luz , Motivos EF Hand/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias/genética , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estabilidade Proteica , Relação Estrutura-Atividade
4.
Parasitol Res ; 117(4): 1035-1041, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29404745

RESUMO

Giardia intestinalis is a parasite that inhabits the small intestine of humans and other mammals, causing a disease that can manifest itself with acute diarrhea. This parasite is an early divergent eukaryote with a compact genome and a life cycle composed of two distinct cell types: the trophozoite, the replicative form, and the cyst, the infectious form. Signal transduction pathways implicated in differentiation processes of G. intestinalis are largely unknown. Calcium, considered an essential messenger in cell signaling, has been shown to regulate a myriad of key cell processes including metabolism, motility, and exocytosis, among other important functions, through calcium-binding proteins (CaBPs). The most important and largest family of CaBPs is the EF-hand protein family. To investigate the nature of calcium signaling pathways present in this protozoan, an in silico analysis of the genome to identify genes encoding EF-hand proteins was undertaken. Twenty-eight sequences containing EF-hand domains were found; most of which have only a pair of domains, and half of the sequences were divergent or unique to Giardia. In addition, the transcription pattern for eight genes encoding EF-hand proteins was assessed during encystation. It was found that all the genes were differentially transcribed suggesting a different function in this process. The in silico results suggest that in G. intestinalis, calcium is involved in the regulation of protein phosphorylation through kinases and phosphatases.


Assuntos
Sinalização do Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Motivos EF Hand/genética , Giardia lamblia/genética , Animais , Cálcio/química , Sinalização do Cálcio/fisiologia , Genoma de Protozoário/genética , Giardia lamblia/metabolismo , Giardíase/parasitologia , Humanos , Fosforilação , Trofozoítos/metabolismo
5.
Cancer Res ; 76(15): 4535-45, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27262171

RESUMO

EF hand (EFh) domain-containing proteins have been implicated in malignant progression, but their precise functional contributions are uncertain. Here, we report evidence that the EFh protein IBA2 promotes the proliferation of breast cancer cells by facilitating their transit through the G1-S cell-cycle transition. Mechanistic investigations revealed that IBA2 acted at the transcriptional level to promote the expression of the critical cell-cycle regulator cyclin D1. Clinically, we found that levels of IBA2 were significantly upregulated in breast cancer specimens, where its expression correlated positively with histologic grade. Our results suggest a key role for IBA2 in mammary tumorigenesis. Cancer Res; 76(15); 4535-45. ©2016 AACR.


Assuntos
Neoplasias da Mama/genética , Ciclina D1/metabolismo , Motivos EF Hand/genética , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Cell Biochem ; 115(3): 557-65, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24123053

RESUMO

Phospholipase C-η (PLCη) enzymes are a class of phosphatidylinositol 4,5-bisphosphate-hydrolyzing enzymes involved in intracellular signaling. PLCη2 can sense Ca(2+) (stimulated by ∼1 µM free Ca(2+) ) suggesting that it can amplify transient Ca(2+) signals. PLCη enzymes possess an EF-hand domain composed of two EF-loops; a canonical 12-residue loop (EF-loop 1) and a non-canonical 13-residue loop (EF-loop 2). Ca(2+) -binding to synthetic peptides corresponding to EF-loops 1 and 2 of PLCη2 and EF-loop 1 of calmodulin (as a control) was examined by 2D-[(1) H,(1) H] TOCSY NMR. Both PLCη2 EF-loop peptides bound Ca(2+) in a similar manner to that of the canonical calmodulin EF-loop 1, particularly at their N-terminus. A molecular model of the PLCη2 EF-hand domain, constructed based upon the structure of calmodulin, suggested both EF-loops may participate in Ca(2+) -binding. To determine whether the EF-hand is responsible for Ca(2+) -sensing, inositol phosphate accumulation was measured in COS7 cells transiently expressing wild-type or mutant PLCη2 proteins. Addition of 70 µM monensin (a Na(+) /H(+) antiporter that increases intracellular Ca(2+) ) induced a 4- to 7-fold increase in wild-type PLCη2 activity. In permeabilized cells, PLCη2 exhibited a ∼4-fold increase in activity in the presence of 1 µM free Ca(2+) . The D256A (EF-loop1) mutant exhibited a ∼10-fold reduction in Ca(2+) -sensitivity and was not activated by monensin, highlighting the involvement of EF-loop 1 in Ca(2+) -sensing. Involvement of EF-loop 2 was examined using D292A, H296A, Q297A, and E304A mutants. Interestingly, the monensin responses and Ca(2+) -sensitivities were largely unaffected by the mutations, indicating that the non-canonical EF-loop 2 is not involved in Ca(2+) -sensing.


Assuntos
Cálcio/metabolismo , Modelos Moleculares , Fosfoinositídeo Fosfolipase C/química , Conformação Proteica , Sequência de Aminoácidos , Animais , Células COS , Cálcio/química , Calmodulina/química , Chlorocebus aethiops , Motivos EF Hand/genética , Humanos , Inositol/farmacologia , Mutação/genética , Fosfoinositídeo Fosfolipase C/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais , Relação Estrutura-Atividade
7.
J Biol Chem ; 289(2): 827-37, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24285542

RESUMO

HER2 is overexpressed in 20­25% of breast cancers. Overexpression of HER2 is an adverse prognostic factor and correlates with decreased patient survival. HER2 stimulates breast tumorigenesis via a number of intracellular signaling molecules, including PI3K/AKT and MAPK/ERK.S100A14,one member of the S100 protein family, is significantly associated with outcome of breast cancer patients. Here, for the first time, we show that S100A14 and HER2 are coexpressed in invasive breast cancer specimens,andthere is a significant correlation between the expression levels of the two proteins by immunohistochemistry. S100A14 and HER2 are colocalized in plasma membrane of breast cancer tissue cells and breast cancer cell lines BT474 and SK-BR3. We demonstrate that S100A14 binds directly to HER2 by co-immunoprecipitation and pull-down assays. Further study shows that residues 956­1154 of the HER2 intracellular domain and residue 83 of S100A14 are essential for the two proteins binding.Moreover,we observe a decrease of HER2 phosphorylation, downstream signaling, and HER2-stimulated cell proliferation in S100A14-silenced MCF-7, BT474, and SK-BR3 cells. Our findings suggest that S100A14 functions as a modulator of HER2 signaling and provide mechanistic evidence for its role in breast cancer progression.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Carcinoma Ductal de Mama/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Ligação ao Cálcio/genética , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Motivos EF Hand/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Metástase Linfática , Células MCF-7 , Microscopia Confocal , Pessoa de Meia-Idade , Ligação Proteica , Receptor ErbB-2/genética
8.
Parasitol Res ; 111(4): 1707-13, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22773043

RESUMO

In trematodes, there is a family of proteins which combine EF-hand-containing domains with dynein light chain (DLC)-like domains. A member of this family from the liver fluke, Fasciola hepatica-FhCaBP4-has been identified and characterised biochemically. FhCaBP4 has an N-terminal domain containing two imperfect EF-hand sequences and a C-terminal dynein light chain-like domain. Molecular modelling predicted that the two domains are joined by a flexible linker. Native gel electrophoresis demonstrated that FhCaBP4 binds to calcium, manganese, barium and strontium ions, but not to magnesium or zinc ions. The hydrophobic, fluorescent probe 8-anilinonaphthalene-1-sulphonate bound more tightly to FhCaBP4 in the presence of calcium ions. This suggests that the protein undergoes a conformational change on ion binding which increases the number of non-polar residues on the surface. FhCaBP4 was protected from limited proteolysis by the calmodulin antagonist W7, but not by trifluoperazine or praziquantel. Protein-protein cross-linking experiments showed that FhCaBP4 underwent calcium ion-dependent dimerisation. Since DLCs are commonly dimeric, it is likely that FhCaBP4 dimerises through this domain. The molecular model reveals that the calcium ion-binding site is located close to a key sequence in the DLC-like domain, suggesting a plausible mechanism for calcium-dependent dimerisation.


Assuntos
Motivos de Aminoácidos , Proteínas de Ligação ao Cálcio/genética , Fasciola hepatica/genética , Fasciola hepatica/metabolismo , Sequência de Aminoácidos , Animais , Dineínas/genética , Motivos EF Hand/genética , Metais/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Análise de Sequência de DNA
9.
Proc Natl Acad Sci U S A ; 109(22): 8682-7, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22586105

RESUMO

Orai1 and stromal interaction molecule (STIM)1 are critical components of Ca(2+) release-activated Ca(2+) (CRAC) channels. Orai1 is a pore subunit of CRAC channels, and STIM1 acts as an endoplasmic reticulum (ER) Ca(2+) sensor that detects store depletion. Upon store depletion after T-cell receptor stimulation, STIM1 translocates and coclusters with Orai1 at sites of close apposition of the plasma membrane (PM) and the ER membrane. However, the molecular components of these ER-PM junctions remain poorly understood. Using affinity protein purification, we uncovered junctate as an interacting partner of Orai1-STIM1 complex. Furthermore, we identified a Ca(2+)-binding EF-hand motif in the ER-luminal region of junctate. Mutation of this EF-hand domain of junctate impaired its Ca(2+) binding and resulted in partial activation of CRAC channels and clustering of STIM1 independently of store depletion. In addition to the known mechanisms of STIM1 clustering (i.e., phosphoinositide and Orai1 binding), our study identifies an alternate mechanism to recruit STIM1 into the ER-PM junctions via binding to junctate. We propose that junctate, a Ca(2+)-sensing ER protein, is a structural component of the ER-PM junctions where Orai1 and STIM1 cluster and interact in T cells.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Cálcio/metabolismo , Canais de Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Membrana Celular/metabolismo , Motivos EF Hand/genética , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Células Jurkat , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/genética , Microscopia de Fluorescência , Oxigenases de Função Mista/genética , Proteínas Musculares/genética , Mutação , Proteínas de Neoplasias/genética , Proteína ORAI1 , Ligação Proteica , Transporte Proteico , Molécula 1 de Interação Estromal
10.
Int J Biochem Cell Biol ; 44(3): 480-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22249026

RESUMO

The SPARC family of proteins represents a diverse group of proteins that modulate cell interaction with the extracellular milieu. The eight members of the SPARC protein family are modular in nature. Each shares a follistatin-like domain and an extracellular calcium binding E-F hand motif. In addition, each family member is secreted into the extracellular space. Some of the shared activities of this family include, regulation of extracellular matrix assembly and deposition, counter-adhesion, effects on extracellular protease activity, and modulation of growth factor/cytokine signaling pathways. Recently, several SPARC family members have been implicated in human disease pathogenesis. This review discusses recent advances in the understanding of the functional roles of the SPARC family of proteins in development and disease.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias/metabolismo , Osteonectina/metabolismo , Animais , Adesão Celular , Citocinas/metabolismo , Motivos EF Hand/genética , Proteínas da Matriz Extracelular/genética , Folistatina/genética , Humanos , Neoplasias/patologia , Neovascularização Patológica , Osteonectina/genética , Dobramento de Proteína , Multimerização Proteica
11.
Genes Cells ; 16(4): 416-26, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21392185

RESUMO

The interaction between calcineurin B homologous protein 2 (CHP2) and Na(+) /H(+) exchanger 1 (NHE1), two membrane proteins, is essential for protecting cells from serum deprivation-induced death. Although four putative EF-hands in CHP2 had been predicted for years, Ca²(+) -binding activities of these motifs have not been tested yet, their role in this process remain poorly understood. To identify Ca²(+) -binding motifs required for the stable formation of CHP2/NHE1 complexes, we developed a mutagenesis-based assay in PS120 cells. We found that (45) Ca²(+) bond to two EF-hand motifs (EF3 and 4) of CHP2 proteins with high affinity. Complex formation between CHP2 and the CHP2 binding domain of NHE1 resulted in a marked increase in the Ca²(+) -binding affinity of CHP2. Co-immunoprecipitation and distribution of GFP-tagged CHP2-EF3m/4m also indicated that Ca²(+) affected the membrane location of CHP2 to interact with NHE1. The C-terminal region of CHP2 contains a nuclear export sequence (NES). When the six leucines of NES were mutated to alanines, the resulting CHP2 protein was predominantly localized to the nucleus. Furthermore, mutation of the NES resulted in enhanced proliferation and oncogenic potential of HeLa cells. Together, these results show that calcium and NES control the subcellular distribution of CHP2 and then distinctively regulate cell proliferation.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Proliferação de Células , Motivos EF Hand/genética , Sinais de Localização Nuclear/química , Trocadores de Sódio-Hidrogênio/metabolismo , Sequência de Aminoácidos , Proteínas de Ligação ao Cálcio/química , Linhagem Celular Transformada , Células HeLa/metabolismo , Humanos , Dados de Sequência Molecular , Sinais de Localização Nuclear/fisiologia , Alinhamento de Sequência , Trocador 1 de Sódio-Hidrogênio
12.
Oral Dis ; 17(3): 283-90, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21029261

RESUMO

OBJECTIVE: S100A2, a Ca(2+) -binding protein with two EF-hands, is a tumor suppressor in oral cancer. Helix III flanking the C-terminal EF-hand is implicated to participate in the interaction of S100A2 and its target(s). The aim of this study was to examine if the coding sequence polymorphism S100A2_185G>A, leading to the peptide 62 substitution of asparagine (AAC, A allele) for serine (AGC, G allele) in helix III, had modulation effects on S100A-mediated tumor suppression. SUBJECTS AND METHODS: We sequenced the coding sequence of S100A2 gene in normal oral keratinocytes (NOKs), dysplastic oral keratinocytes (DOKs), eight oral cancer lines, and 54 pairwise oral cancer specimens. We also compared the in vitro anti-tumor effect of wildtype (G allele) and variant (A allele) S100A2 expression using cell proliferation, migration, invasion, and colony formation assays. RESULTS: With the exception of CAL27 and SCC-15 cancer lines being heterozygotes of A and G alleles, the remaining oral cells were homozygotic in G alleles. No alterations of anti-growth, anti-migration, anti-invasion, and anti-colony formation were observed between variant and wildtype cells. Moreover, no minor S100A2_185A allele was detected in 54-pairwise clinical specimens. CONCLUSION: The coding sequence polymorphism S100A2_185G>A had no regulatory role in S100A2-mediated tumor suppression in oral cancer.


Assuntos
Adenina , Carcinoma de Células Escamosas/genética , Fatores Quimiotáticos/genética , Guanina , Neoplasias Bucais/genética , Fases de Leitura Aberta/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas S100/genética , Adulto , Idoso , Alelos , Substituição de Aminoácidos/genética , Asparagina/genética , Linhagem Celular Tumoral , Células Cultivadas , Motivos EF Hand/genética , Éxons/genética , Feminino , Genótipo , Sequências Hélice-Alça-Hélice/genética , Heterozigoto , Humanos , Células KB , Queratinócitos/patologia , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético/genética , Serina/genética , Proteínas Supressoras de Tumor/genética
13.
Int J Biochem Cell Biol ; 42(11): 1816-22, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20674765

RESUMO

The function of protein phosphatases with EF-hand domains (PPEF) in mammals is not known. Large-scale expression profiling experiments suggest that PPEF expression may correlate with stress protective responses, cell survival, growth, proliferation, or neoplastic transformation. Apoptosis signal regulating kinase-1 (ASK1) is a MAP kinase kinase kinase implicated in cancer, cardiovascular and neurodegenerative diseases. ASK1 is activated by oxidative stress and induces pro-apoptotic or inflammatory signalling, largely via sustained activation of MAP kinases p38 and/or JNK. We identify human PPEF2 as a novel interacting partner and a negative regulator of ASK1. In COS-7 or HEK 293A cells treated with H(2)O(2), expression of PPEF2 abrogated sustained activation of p38 and one of the JNK p46 isoforms, and prevented ASK1-dependent caspase-3 cleavage and activation. PPEF2 efficiently suppressed H(2)O(2)-induced activation of ASK1. Overexpessed as well as endogenous ASK1 co-immunoprecipitated with PPEF2. PPEF2 was considerably more potent both as a suppressor of ASK1 activation and as its interacting partner as compared to protein phosphatase 5 (PP5), a well-known negative regulator of ASK1. PPEF2 was found to form complexes with endogenous Hsp70 and to a lesser extent Hsp90, which are also known interacting partners of PP5. These data identify, for the first time, a possible downstream signalling partner of a mammalian PPEF phosphatase, and suggest that, despite structural divergence, PPEF and PP5 phosphatases may share common interacting partners and functions.


Assuntos
MAP Quinase Quinase Quinase 5/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Animais , Células COS , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Chlorocebus aethiops , Motivos EF Hand/genética , Motivos EF Hand/fisiologia , Humanos , Peróxido de Hidrogênio/farmacologia , Immunoblotting , Imunoprecipitação , MAP Quinase Quinase Quinase 5/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Fosfoproteínas Fosfatases/genética , Ligação Proteica
14.
J Clin Invest ; 117(11): 3540-50, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17965774

RESUMO

Changes in cytoplasmic Ca2+ levels regulate a variety of fundamental cellular functions in virtually all cells. In nonexcitable cells, a major pathway of Ca2+ entry involves receptor-mediated depletion of intracellular Ca2+ stores followed by the activation of store-operated calcium channels in the plasma membrane. We have established a mouse line expressing an activating EF hand motif mutant of stromal interaction molecule 1 (Stim1), an ER receptor recently identified as the Ca2+ sensor responsible for activation of Ca2+ release-activated (CRAC) channels in T cells, whose function in mammalian physiology is not well understood. Mice expressing mutant Stim1 had macrothrombocytopenia and an associated bleeding disorder. Basal intracellular Ca2+ levels were increased in platelets, which resulted in a preactivation state, a selective unresponsiveness to immunoreceptor tyrosine activation motif-coupled agonists, and increased platelet consumption. In contrast, basal Ca2+ levels, but not receptor-mediated responses, were affected in mutant T cells. These findings identify Stim1 as a central regulator of platelet function and suggest a cell type-specific activation or composition of the CRAC complex.


Assuntos
Cálcio/metabolismo , Motivos EF Hand/genética , Hemorragia , Glicoproteínas de Membrana/metabolismo , Mutação , Ativação Plaquetária , Trombocitopenia , Animais , Medula Óssea/patologia , Canais de Cálcio/metabolismo , Fibrose/patologia , Hemorragia/genética , Hemorragia/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos , Ativação Plaquetária/genética , Glicoproteínas da Membrana de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Esplenomegalia/metabolismo , Molécula 1 de Interação Estromal , Linfócitos T/citologia , Linfócitos T/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo
15.
Mol Biochem Parasitol ; 146(1): 10-23, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16297461

RESUMO

A full-length cDNA encoding the Fasciola gigantica calcium-binding protein 1 (FgCaBP1) was cloned from an adult stage cDNA expression library in an immunoscreen using rabbit immune serum against the parasite's excretion/secretion antigens. The deduced amino acid sequence showed 96.3% identity to Fh22CBP of Fasciola hepatica. During development in the mammalian host FgCaBP1 RNA was detected in metacercariae, juveniles and adults and was exclusively localized to the tegumental cell bodies. Immune serum of a rabbit infected with F. gigantica detected recombinant FgCaBP1 starting from the sixth week of infection. Immune sera of mice infected with Schistosoma mansoni and Schistosoma mekongi cross-reacted with recombinant FgCaBP1 in immunoblots. Recombinant FgCaBP1 showed calcium and magnesium-binding activity by a mobility shift during non-denaturing PAGE in the presence of Ca2+ or Mg2+, respectively. A polyclonal mouse anti-rFgCaBP1 antiserum detected the native protein as a major component of the parasite's tegumental antigens in immunoblots and as a strictly tegumental antigen in tissue cross-sections of adult and juvenile parasites. Comparative sequence analysis of homologs from Fasciola and Schistosoma present in the GenBank database revealed sequence signatures specific to these trematode proteins and thereby indicates their origin from a single ancestor. FgCaBP1 contains two adjacent, N-terminal located EF-hands and a C-terminal located domain similar to dynein light chain type 1. Independent structure predictions of the two domains suggest that they will fold according to the already determined structures of the EF-hand motif and the dynein light chain type 1 proteins.


Assuntos
Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/genética , Fasciola/metabolismo , Platelmintos/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/farmacologia , Proteínas de Ligação ao Cálcio/imunologia , Proteínas de Ligação ao Cálcio/isolamento & purificação , Proteínas de Transporte/química , Bovinos , Clonagem Molecular/métodos , DNA Complementar/química , DNA Complementar/genética , Bases de Dados Genéticas , Proteínas de Drosophila/química , Dineínas , Motivos EF Hand/genética , Ensaio de Desvio de Mobilidade Eletroforética , Fasciola/genética , Fasciola/crescimento & desenvolvimento , Fasciola/imunologia , Fasciolíase/imunologia , Fasciolíase/parasitologia , Sequências Hélice-Alça-Hélice/genética , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Filogenia , Platelmintos/classificação , Platelmintos/genética , Coelhos , Proteínas Recombinantes/isolamento & purificação , Alinhamento de Sequência , Análise de Sequência de Proteína
16.
Exp Cell Res ; 307(1): 204-11, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15922740

RESUMO

Allograft Inflammatory Factor-1 (AIF-1) is a cytoplasmic calcium-binding protein expressed in vascular smooth muscle cells (VSMC) in response to injury or cytokine stimulation. AIF-1 contains a partially conserved EF-hand calcium-binding domain, and participates in VSMC activation by activation of Rac1 and induction of Granulocyte-Colony Stimulating Factor (G-CSF) expression; however, the mechanism whereby AIF-1 mediates these effects is presently uncharacterized. To determine if calcium binding plays a functional role in AIF-1 activity, a single site-specific mutation was made in the EF-hand calcium-binding domain to abrogate binding of calcium (AIF-1DeltaA), which was confirmed by calcium overlay. Functionally, similar to wild-type AIF-1, AIF-1DeltaA was able to polymerize F-actin in vitro. However, in contrast to wild-type AIF-1, over-expression of AIF-1DeltaA was unable to increase migration or proliferation of primary human VSMC. Further, it was unable to activate Rac1, or induce G-CSF expression to the degree as wild-type AIF-1. Taken together, modification of the wild-type EF-hand domain and native calcium-binding activity results in a loss of AIF-1 function. We conclude that appropriate calcium-binding potential is critical in AIF-1-mediated effects on VSMC pathophysiology, and that AIF-1 activity is mediated by Rac1 activation and G-CSF expression.


Assuntos
Substituição de Aminoácidos , Proteínas de Ligação ao Cálcio/genética , Cálcio/metabolismo , Motivos EF Hand/genética , Músculo Liso Vascular/metabolismo , Western Blotting , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/metabolismo , Movimento Celular/genética , Proliferação de Células , Proteínas de Ligação a DNA , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Proteínas dos Microfilamentos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Mutagênese Sítio-Dirigida , Retroviridae/genética , Transdução Genética , Proteínas rac1 de Ligação ao GTP/metabolismo
17.
Biochemistry ; 44(19): 7305-14, 2005 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-15882069

RESUMO

S100B is a dimeric Ca(2+)-binding protein that undergoes a 90 +/- 3 degrees rotation of helix 3 in the typical EF-hand domain (EF2) upon the addition of calcium. The large reorientation of this helix is a prerequisite for the interaction between each subunit of S100B and target proteins such as the tumor suppressor protein, p53. In this study, Tb(3+) was used as a probe to examine how binding of a 22-residue peptide derived from the C-terminal regulatory domain of p53 affects the rate of Ca(2+) ion dissociation. In competition studies with Tb(3+), the dissociation rates of Ca(2+) (k(off)) from the EF2 domains of S100B in the absence and presence of the p53 peptide was determined to be 60 and 7 s(-)(1), respectively. These data are consistent with a previously reported result, which showed that that target peptide binding to S100B enhances its calcium-binding affinity [Rustandi et al. (1998) Biochemistry 37, 1951-1960]. The corresponding Ca(2+) association rate constants for S100B, k(on), for the EF2 domains in the absence and presence of the p53 peptide are 1.1 x 10(6) and 3.5 x 10(5) M(-)(1) s(-)(1), respectively. These two association rate constants are significantly below the diffusion control ( approximately 10(9) M(-)(1) s(-)(1)) and likely involve both Ca(2+) ion association and a Ca(2+)-dependent structural rearrangement, which is slightly different when the target peptide is present. EF-hand calcium-binding mutants of S100B were engineered at the -Z position (EF-hand 1, E31A; EF-hand 2, E72A; both EF-hands, E31A + E72A) and examined to further understand how specific residues contribute to calcium binding in S100B in the absence and presence of the p53 peptide.


Assuntos
Cálcio/química , Motivos EF Hand/genética , Fatores de Crescimento Neural/química , Fragmentos de Peptídeos/química , Proteínas S100/química , Proteína Supressora de Tumor p53/química , Proteínas Supressoras de Tumor/química , Alanina/genética , Sequência de Aminoácidos , Animais , Ácido Glutâmico/genética , Humanos , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Ratos , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/genética , Proteínas S100/metabolismo , Térbio/química , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
18.
Biochemistry ; 43(35): 11175-86, 2004 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-15366927

RESUMO

Apoptosis-linked gene-2 (ALG-2) encodes a 22 kDa Ca(2+)-binding protein of the penta EF-hand family that is required for programmed cell death in response to various apoptotic agents. Here, we demonstrate that ALG-2 mRNA and protein are down-regulated in human uveal melanoma cells compared to their progenitor cells, normal melanocytes. The down regulation of ALG-2 may provide melanoma cells with a selective advantage. ALG-2 and its putative target molecule, Alix/AIP1, are localized primarily in the cytoplasm of melanocytes and melanoma cells independent of the intracellular Ca(2+) concentration or the activation of apoptosis. Cross-linking and analytical centrifugation studies support a single-species dimer conformation of ALG-2, also independent of Ca(2+) concentration. However, binding of Ca(2+) to both EF-1 and EF-3 is necessary for ALG-2 interaction with Alix/AIP1 as demonstrated using surface plasmon resonance spectroscopy. Mutations in EF-5 result in reduced target interaction without alteration in Ca(2+) affinity. The addition of N-terminal ALG-2 peptides, residues 1-22 or residues 7-17, does not alter the interaction of ALG-2 or an N-terminal deletion mutant of ALG-2 with Alix/AIP1, as might be expected from a model derived from the crystal structure of ALG-2. Fluorescence studies of ALG-2 demonstrate that an increase in surface hydrophobicity is primarily due to Ca(2+) binding to EF-3, while Ca(2+) binding to EF-1 has little effect on surface exposure of hydrophobic residues. Together, these data indicate that gross surface hydrophobicity changes are insufficient for target recognition.


Assuntos
Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Motivos EF Hand , Melanoma/metabolismo , Neoplasias Uveais/metabolismo , Proteínas Reguladoras de Apoptose , Sítios de Ligação/genética , Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Células Cultivadas , Dimerização , Regulação para Baixo/genética , Motivos EF Hand/genética , Complexos Endossomais de Distribuição Requeridos para Transporte , Regulação Neoplásica da Expressão Gênica , Humanos , Melanócitos/citologia , Melanócitos/metabolismo , Melanoma/genética , Melanoma/patologia , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Estrutura Terciária de Proteína/genética , Ressonância de Plasmônio de Superfície , Neoplasias Uveais/genética , Neoplasias Uveais/patologia
19.
Eur J Biochem ; 271(18): 3785-93, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15355355

RESUMO

Guanylate cyclase-activating proteins (GCAPs) are neuronal calcium sensors that activate membrane bound guanylate cyclases (EC 4.6.1.2.) of vertebrate photoreceptor cells when cytoplasmic Ca2+ decreases during illumination. GCAPs contain four EF-hand Ca2+-binding motifs, but the first EF-hand is nonfunctional. It was concluded that for GCAP-2, the loss of Ca2+-binding ability of EF-hand 1 resulted in a region that is crucial for targeting guanylate cyclase [Ermilov, A.N., Olshevskaya, E.V. & Dizhoor, A.M. (2001) J. Biol. Chem.276, 48143-48148]. In this study we tested the consequences of mutations in EF-hand 1 of GCAP-1 with respect to Ca2+ binding, Ca2+-induced conformational changes and target activation. When the nonfunctional first EF-hand in GCAP-1 is replaced by a functional EF-hand the chimeric mutant CaM-GCAP-1 bound four Ca2+ and showed similar Ca2+-dependent changes in tryptophan fluorescence as the wild-type. CaM-GCAP-1 neither activated nor interacted with guanylate cyclase. Size exclusion chromatography revealed that the mutant tended to form inactive dimers instead of active monomers like the wild-type. Critical amino acids in EF-hand 1 of GCAP-1 are cysteine at position 29 and proline at position 30, as changing these to glycine was sufficient to cause loss of target activation without a loss of Ca2+-induced conformational changes. The latter mutation also promoted dimerization of the protein. Our results show that EF-hand 1 in wild-type GCAP-1 is critical for providing the correct conformation for target activation.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Mutação , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/isolamento & purificação , Cromatografia em Gel , Cisteína/metabolismo , Dimerização , Motivos EF Hand/genética , Escherichia coli/genética , Proteínas Ativadoras de Guanilato Ciclase , Transdução de Sinal Luminoso , Dados de Sequência Molecular , Prolina/metabolismo , Conformação Proteica , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Espectrometria de Fluorescência
20.
Genomics ; 84(1): 10-22, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15203200

RESUMO

The EF-hand superfamily of calcium binding proteins includes the S100, calcium binding protein, and troponin subfamilies. This study represents a genome, structure, and expression analysis of the S100 protein family, in mouse, human, and rat. We confirm the high level of conservation between mammalian sequences but show that four members, including S100A12, are present only in the human genome. We describe three new members of the S100 family in the three species and their locations within the S100 genomic clusters and propose a revised nomenclature and phylogenetic relationship between members of the EF-hand superfamily. Two of the three new genes were induced in bone-marrow-derived macrophages activated with bacterial lipopolysaccharide, suggesting a role in inflammation. Normal human and murine tissue distribution profiles indicate that some members of the family are expressed in a specific manner, whereas others are more ubiquitous. Structure-function analysis of the chemotactic properties of murine S100A8 and human S100A12, particularly within the active hinge domain, suggests that the human protein is the functional homolog of the murine protein. Strong similarities between the promoter regions of human S100A12 and murine S100A8 support this possibility. This study provides insights into the possible processes of evolution of the EF-hand protein superfamily. Evolution of the S100 proteins appears to have occurred in a modular fashion, also seen in other protein families such as the C2H2-type zinc-finger family.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Motivos EF Hand/genética , Evolução Molecular , Genoma , Filogenia , Animais , Compostos de Boro/metabolismo , Quimiotaxia/genética , Quimiotaxia/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Metacrilatos/metabolismo , Metilmetacrilatos/metabolismo , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/metabolismo , Especificidade de Órgãos/genética , Ratos , Dedos de Zinco/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA