Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Development ; 144(3): 357-364, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28143842

RESUMO

The 6th EMBO conference on the Molecular and Cellular Basis of Regeneration and Tissue Repair took place in Paestum (Italy) on the 17th-21st September, 2016. The 160 scientists who attended discussed the importance of cellular and tissue plasticity, biophysical aspects of regeneration, the diverse roles of injury-induced immune responses, strategies to reactivate regeneration in mammals, links between regeneration and ageing, and the impact of non-mammalian models on regenerative medicine.


Assuntos
Regeneração/fisiologia , Cicatrização/fisiologia , Envelhecimento/fisiologia , Animais , Fenômenos Biomecânicos , Sistema Nervoso Central/fisiologia , Fenômenos Eletrofisiológicos , Coração/fisiologia , Humanos , Modelos Biológicos , Regeneração/imunologia , Medicina Regenerativa/tendências , Transdução de Sinais , Cicatrização/imunologia , Ferimentos e Lesões/imunologia , Ferimentos e Lesões/fisiopatologia
2.
Nucleic Acids Res ; 46(1): 267-278, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29165708

RESUMO

Proper chromosome segregation is crucial for preserving genomic integrity, and errors in this process cause chromosome mis-segregation, which may contribute to cancer development. Sister chromatid separation is triggered by Separase, an evolutionary conserved protease that cleaves the cohesin complex, allowing the dissolution of sister chromatid cohesion. Here we provide evidence that Separase participates in genomic stability maintenance by controlling replication fork speed. We found that Separase interacted with the replication licensing factors MCM2-7, and genome-wide data showed that Separase co-localized with MCM complex and cohesin. Unexpectedly, the depletion of Separase increased the fork velocity about 1.5-fold and caused a strong acetylation of cohesin's SMC3 subunit and altered checkpoint response. Notably, Separase silencing triggered genomic instability in both HeLa and human primary fibroblast cells. Our results show a novel mechanism for fork progression mediated by Separase and thus the basis for genomic instability associated with tumorigenesis.


Assuntos
Replicação do DNA , DNA/química , Instabilidade Genômica , Conformação de Ácido Nucleico , Separase/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Cromátides/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos , DNA/genética , DNA/metabolismo , Células HeLa , Humanos , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Modelos Genéticos , Ligação Proteica , Interferência de RNA , Separase/genética , Coesinas
3.
Hum Mol Genet ; 24(15): 4296-305, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25948553

RESUMO

Defective expression of frataxin is responsible for the inherited, progressive degenerative disease Friedreich's Ataxia (FRDA). There is currently no effective approved treatment for FRDA and patients die prematurely. Defective frataxin expression causes critical metabolic changes, including redox imbalance and ATP deficiency. As these alterations are known to regulate the tyrosine kinase Src, we investigated whether Src might in turn affect frataxin expression. We found that frataxin can be phosphorylated by Src. Phosphorylation occurs primarily on Y118 and promotes frataxin ubiquitination, a signal for degradation. Accordingly, Src inhibitors induce accumulation of frataxin but are ineffective on a non-phosphorylatable frataxin-Y118F mutant. Importantly, all the Src inhibitors tested, some of them already in the clinic, increase frataxin expression and rescue the aconitase defect in frataxin-deficient cells derived from FRDA patients. Thus, Src inhibitors emerge as a new class of drugs able to promote frataxin accumulation, suggesting their possible use as therapeutics in FRDA.


Assuntos
Ataxia de Friedreich/genética , Proteínas de Ligação ao Ferro/biossíntese , Quinases da Família src/genética , Trifosfato de Adenosina/deficiência , Trifosfato de Adenosina/genética , Inibidores Enzimáticos/farmacologia , Ataxia de Friedreich/tratamento farmacológico , Ataxia de Friedreich/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Ligação ao Ferro/genética , Oxirredução , Ubiquitinação/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo , Frataxina
4.
Hum Mol Genet ; 23(21): 5814-26, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24925315

RESUMO

Saposin (Sap) C is an essential cofactor for the lysosomal degradation of glucosylceramide (GC) by glucosylceramidase (GCase) and its functional impairment underlies a rare variant form of Gaucher disease (GD). Sap C promotes rearrangement of lipid organization in lysosomal membranes favoring substrate accessibility to GCase. It is characterized by six invariantly conserved cysteine residues involved in three intramolecular disulfide bonds, which make the protein remarkably stable to acid environment and degradation. Five different mutations (i.e. p.C315S, p.342_348FDKMCSKdel, p.L349P, p.C382G and p.C382F) have been identified to underlie Sap C deficiency. The molecular mechanism by which these mutations affect Sap C function, however, has not been delineated in detail. Here, we characterized biochemically and functionally four of these gene lesions. We show that all Sap C mutants are efficiently produced, and exhibit lipid-binding properties, modulatory behavior on GCase activity and subcellular localization comparable with those of the wild-type protein. We then delineated the structural rearrangement of these mutants, documenting that most proteins assume diverse aberrant disulfide bridge arrangements, which result in a substantial diminished half-life, and rapid degradation via autophagy. These findings further document the paramount importance of disulfide bridges in the stability of Sap C and provide evidence that accelerated degradation of the Sap C mutants is the underlying pathogenetic mechanism of Sap C deficiency.


Assuntos
Doença de Gaucher/genética , Doença de Gaucher/metabolismo , Lisossomos/metabolismo , Mutação , Saposinas/genética , Saposinas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Expressão Gênica , Humanos , Espectrometria de Massas , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Estabilidade Proteica , Transporte Proteico , Proteólise , Saposinas/química , Saposinas/deficiência , Alinhamento de Sequência
5.
Mol Ther ; 23(5): 885-895, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25669433

RESUMO

Although in the last decades the molecular underpinnings of the cell cycle have been unraveled, the acquired knowledge has been rarely translated into practical applications. Here, we investigate the feasibility and safety of triggering proliferation in vivo by temporary suppression of the cyclin-dependent kinase inhibitor, p21. Adeno-associated virus (AAV)-mediated, acute knockdown of p21 in intact skeletal muscles elicited proliferation of multiple, otherwise quiescent cell types, notably including satellite cells. Compared with controls, p21-suppressed muscles exhibited a striking two- to threefold expansion in cellularity and increased fiber numbers by 10 days post-transduction, with no detectable inflammation. These changes partially persisted for at least 60 days, indicating that the muscles had undergone lasting modifications. Furthermore, morphological hyperplasia was accompanied by 20% increases in maximum strength and resistance to fatigue. To assess the safety of transiently suppressing p21, cells subjected to p21 knockdown in vitro were analyzed for γ-H2AX accumulation, DNA fragmentation, cytogenetic abnormalities, ploidy, and mutations. Moreover, the differentiation competence of p21-suppressed myoblasts was investigated. These assays confirmed that transient suppression of p21 causes no genetic damage and does not impair differentiation. Our results establish the basis for further exploring the manipulation of the cell cycle as a strategy in regenerative medicine.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Animais , Ciclo Celular/genética , Diferenciação Celular/genética , Proliferação de Células , Aberrações Cromossômicas , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dependovirus/classificação , Dependovirus/genética , Fibroblastos , Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Reporter , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Camundongos , Contração Muscular/genética , Mutação , Interferência de RNA , RNA Interferente Pequeno/genética , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Sorogrupo , Transdução Genética
6.
J Cell Sci ; 126(Pt 23): 5477-89, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24046450

RESUMO

Nemaline myopathy (NM) is a congenital myopathy with an estimated incidence of 150,000 live births. It is caused by mutations in thin filament components, including nebulin, which accounts for about 50% of the cases. The identification of NM cases with nonsense mutations resulting in loss of the extreme C-terminal SH3 domain of nebulin suggests an important role of the nebulin SH3 domain, which is further supported by the recent demonstration of its role in IGF-1-induced sarcomeric actin filament formation through targeting of N-WASP to the Z-line. To provide further insights into the functional significance of the nebulin SH3 domain in the Z-disk and to understand the mechanisms by which truncations of nebulin lead to NM, we took two approaches: (1) an affinity-based proteomic screening to identify novel interaction partners of the nebulin SH3 domain; and (2) generation and characterization of a novel knockin mouse model with a premature stop codon in the nebulin gene, eliminating its C-terminal SH3 domain (NebΔSH3 mouse). Surprisingly, detailed analyses of NebΔSH3 mice revealed no structural or histological skeletal muscle abnormalities and no changes in gene expression or localization of interaction partners of the nebulin SH3 domain, including myopalladin, palladin, zyxin and N-WASP. Also, no significant effect on peak isometric stress production, passive tensile stress or Young's modulus was found. However, NebΔSH3 muscle displayed a slightly altered force-frequency relationship and was significantly more susceptible to eccentric contraction-induced injury, suggesting that the nebulin SH3 domain protects against eccentric contraction-induced injury and possibly plays a role in fine-tuning the excitation-contraction coupling mechanism.


Assuntos
Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Animais , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Módulo de Elasticidade/fisiologia , Acoplamento Excitação-Contração/fisiologia , Feminino , Expressão Gênica , Humanos , Contração Isométrica/fisiologia , Masculino , Camundongos , Proteínas Musculares/química , Proteínas Musculares/deficiência , Proteínas Musculares/metabolismo , Músculo Esquelético/patologia , Miopatias da Nemalina/genética , Miopatias da Nemalina/metabolismo , Miopatias da Nemalina/patologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Resistência à Tração/fisiologia , Suporte de Carga/fisiologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Zixina/genética , Zixina/metabolismo
7.
J Autoimmun ; 58: 78-89, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25623267

RESUMO

T lymphocytes from patients with Systemic Lupus Erythematosus (SLE) display multiple abnormalities, including increased cell activation, abnormal cell death by apoptosis and impairment of autophagy pathway. In the present study we report the presence of specific antibodies to D4GDI, a small GTPase family inhibitor, in a significant percentage (46%) of SLE patient sera. We also found a significant association between the presence of these autoantibodies and hematologic manifestations occurring in these patients. Investigating the possible implication of anti-D4GDI autoantibodies in SLE pathogenesis or progression, we found that these antibodies were capable of binding D4GDI expressed at the lymphocyte surface and triggering a series of subcellular events, including Rho GTPase activation. These antibodies were also able to induce autophagy in T cells from both healthy donors and SLE patients, but only those negative to these antibodies. We can conclude that anti-D4GDI autoantibodies could be capable of triggering important responses in T cells such as cytoskeleton remodeling and autophagy pathway and that, in SLE patients, the chronic exposure to these specific autoantibodies could lead to the selection of autophagy-resistant T cell clones contributing to the pathogenesis of the disease.


Assuntos
Autoanticorpos/sangue , Lúpus Eritematoso Sistêmico/imunologia , Linfócitos T/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Inibidor beta de Dissociação do Nucleotídeo Guanina rho/imunologia , Adulto , Idoso , Autofagia/genética , Citoesqueleto/metabolismo , Progressão da Doença , Feminino , Humanos , Células Jurkat , Masculino , Pessoa de Meia-Idade , Ligação Proteica/genética , RNA Interferente Pequeno/genética , Linfócitos T/imunologia , Adulto Jovem , Inibidor beta de Dissociação do Nucleotídeo Guanina rho/genética
8.
Differentiation ; 87(1-2): 83-99, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24703763

RESUMO

Hydra is a freshwater hydrozoan polyp that constantly renews its two tissue layers thanks to three distinct stem cell populations that cannot replace each other, epithelial ectodermal, epithelial endodermal, and multipotent interstitial. These adult stem cells, located in the central body column, exhibit different cycling paces, slow for the epithelial, fast for the interstitial. To monitor the changes in cell cycling in Hydra, we established a fast and efficient flow cytometry procedure, which we validated by confirming previous findings, as the Nocodazole-induced reversible arrest of cell cycling in G2/M, and the mitogenic signal provided by feeding. Then to dissect the cycling and differentiation behaviors of the interstitial stem cells, we used the AEP_cnnos1 and AEP_Icy1 transgenic lines that constitutively express GFP in this lineage. For the epithelial lineages we used the sf-1 strain that rapidly eliminates the fast cycling cells upon heat-shock and progressively becomes epithelial. This study evidences similar cycling patterns for the interstitial and epithelial stem cells, which all alternate between the G2 and S-phases traversing a minimal G1-phase. We also found interstitial progenitors with a shorter G2 that pause in G1/G0. At the animal extremities, most cells no longer cycle, the epithelial cells terminally differentiate in G2 and the interstitial progenitors in G1/G0. At the apical pole ~80% cells are post-mitotic differentiated cells, reflecting the higher density of neurons and nematocytes in this region. We discuss how the robust G2 pausing of stem cells, maintained over weeks of starvation, may contribute to regeneration.


Assuntos
Células-Tronco Adultas/citologia , Diferenciação Celular/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Hydra/citologia , Animais , Células Epiteliais/citologia , Neurônios/fisiologia , Regeneração/genética
9.
Biochim Biophys Acta ; 1833(6): 1443-53, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23485397

RESUMO

HIPK2 (homeodomain-interacting protein kinase-2) binds to and phosphorylates, at Ser and Thr residues, a large number of targets involved in cell division and cell fate decision in response to different physiological or stress stimuli. Inactivation of HIPK2 has been observed in human and mouse cancers supporting its role as a tumor suppressor. Despite the biological relevance of this kinase, very little is known on how HIPK2 becomes catalytically active. Based on sequence homologies, HIPK2 has been taxonomically classified as a subfamily member of the dual-specificity tyrosine-regulated kinases (DYRKs) and the activation-loop Y354 of HIPK2 has been found phosphorylated in different cells; however, the relevance of this Y phosphorylation is presently unknown. Here, we show that HIPK2, which is extensively phosphorylated at S/T sites throughout its functional domains, becomes catalytically active by autophosphorylation at the activation-loop Y354. In particular, we found that, in analogy to DYRKs, HIPK2-Y354 phosphorylation is an autocatalytic event and its prevention, through Y354 substitution with non-phosphorylatable amino acids or by using the kinase inhibitor purvalanol A, induces a strong reduction of the HIPK2 S/T-kinase activity on different substrates. Interestingly, at variance from DYRKs, inhibition of HIPK2-Y354 phosphorylation induces a strong out-of-target Y-kinase activity in cis and a strong cytoplasmic relocalization of the kinase. Together, these results demonstrate that the catalytic activity, substrate specificity, and subcellular localization of HIPK2 are regulated by autophosphorylation of its activation-loop Y354.


Assuntos
Proteínas de Transporte/metabolismo , Citoplasma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Treonina/metabolismo , Animais , Western Blotting , Proteínas de Transporte/genética , Cromatografia Líquida , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Camundongos , Fosforilação , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Frações Subcelulares , Especificidade por Substrato , Espectrometria de Massas em Tandem , Treonina/genética , Tirosina/metabolismo
10.
Biochim Biophys Acta ; 1833(1): 110-21, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23103755

RESUMO

Phosphorylation and nitration of protein tyrosine residues are thought to play a role in signaling pathways at the nerve terminal and to affect functional properties of proteins involved in the synaptic vesicle (SV) exo-endocytotic cycle. We previously demonstrated that the tyrosine residues in the C-terminal domain of the SV protein Synaptophysin (SYP) are targets of peroxynitrite (PN). Here, we have characterized the association between SYP and c-src tyrosine kinase demonstrating that phosphorylation of Tyr(273) in the C-terminal domain of SYP is crucial in mediating SYP binding to and activation of c-src. SYP forms a complex with Dynamin I (DynI), a GTPase required for SV endocytosis, which may be regulated by tyrosine phosphorylation of SYP. We here report that, in rat brain synaptosomes treated with PN, the formation of SYP/DynI complex was impaired. Noteworthy, we found that DynI was also modified by PN. DynI tyrosine phosphorylation was down-regulated in a dose-dependent manner, while DynI tyrosine nitration increased. Using mass spectrometry analysis, we identified Tyr(354) as one nitration site in DynI. In addition, we tested DynI self-assembly and GTPase activity, which are enhanced by c-src-dependent tyrosine phosphorylation of DynI, and found that both were inhibited by PN. Our results suggest that the site-specific tyrosine residue modifications may modulate the association properties of SV proteins and serve as a regulator of DynI function via control of self-assembly, thus influencing the physiology of the exo-endocytotic cycle.


Assuntos
Dinamina I/metabolismo , Dinamina I/fisiologia , Vesículas Sinápticas/metabolismo , Sinaptofisina/metabolismo , Sinaptofisina/fisiologia , Sequência de Aminoácidos , Animais , Dinamina I/química , Dinamina I/genética , Endocitose/genética , Endocitose/fisiologia , Exocitose/genética , Exocitose/fisiologia , Técnicas In Vitro , Dados de Sequência Molecular , Nitratos/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Ratos , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Vesículas Sinápticas/fisiologia , Sinaptofisina/química , Sinaptofisina/genética , Tirosina/metabolismo , Tirosina/fisiologia
11.
Neurobiol Dis ; 66: 1-18, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24561067

RESUMO

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare leukodystrophy caused by mutations in the gene encoding MLC1, a membrane protein mainly expressed in astrocytes in the central nervous system. Although MLC1 function is unknown, evidence is emerging that it may regulate ion fluxes. Using biochemical and proteomic approaches to identify MLC1 interactors and elucidate MLC1 function we found that MLC1 interacts with the vacuolar ATPase (V-ATPase), the proton pump that regulates endosomal acidity. Because we previously showed that in intracellular organelles MLC1 directly binds Na, K-ATPase, which controls endosomal pH, we studied MLC1 endosomal localization and trafficking and MLC1 effects on endosomal acidity and function using human astrocytoma cells overexpressing wild-type (WT) MLC1 or MLC1 carrying pathological mutations. We found that WT MLC1 is abundantly expressed in early (EEA1(+), Rab5(+)) and recycling (Rab11(+)) endosomes and uses the latter compartment to traffic to the plasma membrane during hyposmotic stress. We also showed that WT MLC1 limits early endosomal acidification and influences protein trafficking in astrocytoma cells by stimulating protein recycling, as revealed by FITC-dextran measurement of endosomal pH and transferrin protein recycling assay, respectively. WT MLC1 also favors recycling to the plasma-membrane of the TRPV4 cation channel which cooperates with MLC1 to activate calcium influx in astrocytes during hyposmotic stress. Although MLC disease-causing mutations differentially affect MLC1 localization and trafficking, all the mutated proteins fail to influence endosomal pH and protein recycling. This study demonstrates that MLC1 modulates endosomal pH and protein trafficking suggesting that alteration of these processes contributes to MLC pathogenesis.


Assuntos
Astrócitos/metabolismo , Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Transporte Proteico , Animais , Encéfalo/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Endossomos/efeitos dos fármacos , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Proteínas de Membrana/genética , Estresse Oxidativo , Transporte Proteico/efeitos dos fármacos , Ratos , Canais de Cátion TRPV/metabolismo , Transferrina/metabolismo , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo
12.
Cancer Cell ; 10(6): 473-86, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17157788

RESUMO

Che-1 is a RNA polymerase II-binding protein involved in the transcription of E2F target genes and induction of cell proliferation. Here we show that Che-1 contributes to DNA damage response and that its depletion sensitizes cells to anticancer agents. The checkpoint kinases ATM/ATR and Chk2 interact with Che-1 and promote its phosphorylation and accumulation in response to DNA damage. These Che-1 modifications induce a specific recruitment of Che-1 on the TP53 and p21 promoters. Interestingly, it has a profound effect on the basal expression of p53, which is preserved following DNA damage. Notably, Che-1 contributes to the maintenance of the G2/M checkpoint induced by DNA damage. These findings identify a mechanism by which checkpoint kinases regulate responses to DNA damage.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Genes p53 , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Animais , Antineoplásicos/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia , Divisão Celular , Quinase do Ponto de Checagem 2 , Inibidor de Quinase Dependente de Ciclina p21/genética , Dano ao DNA , Fase G2 , Humanos , Camundongos , Células NIH 3T3 , Fosforilação , Regiões Promotoras Genéticas , Transcrição Gênica
13.
Nucleic Acids Res ; 40(3): 1106-17, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21984412

RESUMO

DNA ligase I-deficient 46BR.1G1 cells show a delay in the maturation of replicative intermediates resulting in the accumulation of single- and double-stranded DNA breaks. As a consequence the ataxia telangiectasia mutated protein kinase (ATM) is constitutively phosphorylated at a basal level. Here, we use 46BR.1G1 cells as a model system to study the cell response to chronic replication-dependent DNA damage. Starting from a proteomic approach, we demonstrate that the phosphorylation level of factors controlling constitutive and alternative splicing is affected by the damage elicited by DNA ligase I deficiency. In particular, we show that SRSF1 is hyperphosphorylated in 46BR.1G1 cells compared to control fibroblasts. This hyperphosphorylation can be partially prevented by inhibiting ATM activity with caffeine. Notably, hyperphosphorylation of SRSF1 affects the subnuclear distribution of the protein and the alternative splicing pattern of target genes. We also unveil a modulation of SRSF1 phosphorylation after exposure of MRC-5V1 control fibroblasts to different exogenous sources of DNA damage. Altogether, our observations indicate that a relevant aspect of the cell response to DNA damage involves the post-translational regulation of splicing factor SRSF1 which is associated with a shift in the alternative splicing program of target genes to control cell survival or cell death.


Assuntos
Dano ao DNA , Replicação do DNA , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Processamento Alternativo , Linhagem Celular Transformada , DNA Ligase Dependente de ATP , DNA Ligases/genética , Humanos , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Fosforilação , Proteômica , Proteínas de Ligação a RNA/análise , Proteínas de Ligação a RNA/genética , Fatores de Processamento de Serina-Arginina , Estresse Fisiológico/genética
14.
bioRxiv ; 2023 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-37609214

RESUMO

The WRN protein mutated in the hereditary premature aging disorder Werner syndrome plays a vital role in handling, processing, and restoring perturbed replication forks. One of its most abundant partners, Replication Protein A (RPA), has been shown to robustly enhance WRN helicase activity in specific cases when tested in vitro. However, the significance of RPA-binding to WRN at replication forks in vivo has remained largely unexplored. In this study, we have identified several conserved phosphorylation sites in the acidic domain of WRN that are targeted by Casein Kinase 2 (CK2). Surprisingly, these phosphorylation sites are essential for the interaction between WRN and RPA, both in vitro and in human cells. By characterizing a CK2-unphosphorylatable WRN mutant that lacks the ability to bind RPA, we have determined that the WRN-RPA complex plays a critical role in fork recovery after replication stress whereas the WRN-RPA interaction is not necessary for the processing of replication forks or preventing DNA damage when forks stall or collapse. When WRN fails to bind RPA, fork recovery is impaired, leading to the accumulation of single-stranded DNA gaps in the parental strands, which are further enlarged by the structure-specific nuclease MRE11. Notably, RPA-binding by WRN and its helicase activity are crucial for countering the persistence of G4 structures after fork stalling. Therefore, our findings reveal for the first time a novel role for the WRN-RPA interaction to facilitate fork restart, thereby minimizing G4 accumulation at single-stranded DNA gaps and suppressing accumulation of unreplicated regions that may lead to MUS81-dependent double-strand breaks requiring efficient repair by RAD51 to prevent excessive DNA damage.

15.
J Proteome Res ; 11(5): 2666-83, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22452640

RESUMO

14-3-3s are phosphoserine/phosphotreonine binding proteins that play pivotal roles as regulators of multiple cellular processes in eukaryotes. The flagellated protozoan parasite Giardia duodenalis, the causing agent of giardiasis, is a valuable simplified eukaryotic model. A single 14-3-3 isoform (g14-3-3) is expressed in Giardia, and it is directly involved in the differentiation of the parasite into cyst. To define the overall functions of g14-3-3, the protein interactome has been investigated. A transgenic G. duodenalis strain was engineered to express a FLAG-tagged g14-3-3 under its own promoter. Affinity chromatography coupled with tandem mass spectrometry analysis have been used to purify and identify FLAG-g14-3-3-associated proteins from trophozoites and encysting parasites. A total of 314 putative g14-3-3 interaction partners were identified, including proteins involved in several pathways. Some interactions seemed to be peculiar of one specific stage, while others were shared among the different stages. Furthermore, the interaction of g14-3-3 with the giardial homologue of the CDC7 protein kinase (gCDC7) was characterized, leading to the identification of a multiprotein complex containing not only g14-3-3 and gCDC7 but also a newly identified and highly divergent homologue of DBF4, the putative regulatory subunit of gCDC7. The relevance of g14-3-3 interactions in G. duodenalis biology was discussed.


Assuntos
Proteínas 14-3-3/metabolismo , Giardia lamblia/metabolismo , Mapas de Interação de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas 14-3-3/genética , Motivos de Aminoácidos , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatografia de Afinidade , DNA de Protozoário/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Giardia lamblia/genética , Imunoprecipitação , Ligantes , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Organismos Geneticamente Modificados/genética , Organismos Geneticamente Modificados/metabolismo , Regiões Promotoras Genéticas , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/genética , Proteoma/análise , Proteínas de Protozoários/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Espectrometria de Massas em Tandem , Transfecção
16.
J Biol Chem ; 286(10): 8644-8654, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21209082

RESUMO

Proliferation of mammalian cardiomyocytes stops rapidly after birth and injured hearts do not regenerate adequately. High cyclin-dependent kinase inhibitor (CKI) levels have been observed in cardiomyocytes, but their role in maintaining cardiomyocytes in a post-mitotic state is still unknown. In this report, it was investigated whether CKI knockdown by RNA interference induced cardiomyocyte proliferation. We found that triple transfection with p21(Waf1), p27(Kip1), and p57(Kip2) siRNAs induced both neonatal and adult cardiomyocyte to enter S phase and increased the nuclei/cardiomyocyte ratio; furthermore, a subpopulation of cardiomyocytes progressed beyond karyokynesis, as assessed by the detection of mid-body structures and by straight cardiomyocyte counting. Intriguingly, cardiomyocyte proliferation occurred in the absence of overt DNA damage and aberrant mitotic figures. Finally, CKI knockdown and DNA synthesis reactivation correlated with a dramatic change in adult cardiomyocyte morphology that may be a prerequisite for cell division. In conclusion, CKI expression plays an active role in maintaining cardiomyocyte withdrawal from the cell cycle.


Assuntos
Ciclo Celular/fisiologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Animais Recém-Nascidos , Linhagem Celular , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Dano ao DNA/fisiologia , Técnicas de Silenciamento de Genes , Camundongos , Proteínas Musculares/genética , Miócitos Cardíacos/citologia , Interferência de RNA , Ratos , Ratos Wistar
17.
J Biol Chem ; 286(6): 4471-84, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21135098

RESUMO

The flagellated protozoan Giardia duodenalis is a parasite of the upper part of the small intestine of mammals, including humans, and an interesting biological model. Giardia harbors a single 14-3-3 isoform, a multifunctional protein family, that is modified at the C terminus by polyglycylation, an unusual post-translational modification consisting of the covalent addition of one or multiple glycines on the γ-carboxyl groups of specific glutamic acids. Polyglycylation affects the intracellular localization of g14-3-3, as the shortening of the polyglycine chain is correlated with a partial relocalization of 14-3-3 inside the nuclei during encystation. In this work we demonstrate that the gTTLL3, a member of the tubulin tyrosine ligase-like family, is the enzyme responsible for the 14-3-3 polyglycylation. We also identify two metallopeptidases of the M20 family, here termed gDIP1 (giardial dipeptidase 1) and gDIP2, as enzymes able to shorten the g14-3-3 polyglycine tail both in vivo and in vitro. Finally, we show that the ectopic expression of gDIP2 alters the g14-3-3 localization and strongly hampers the cyst formation. In conclusion, we have identified a polyglycylase and two deglycylases that act in concert to modulate the stage-dependent glycylation status of the multifunctional regulatory g14-3-3 protein in G. duodenalis.


Assuntos
Proteínas 14-3-3/metabolismo , Carboxipeptidases/metabolismo , Giardia/metabolismo , Metaloproteases/metabolismo , Metaloproteases/fisiologia , Peptídeo Sintases/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas 14-3-3/genética , Animais , Carboxipeptidases/genética , Giardia/genética , Glicina/genética , Glicina/metabolismo , Metaloproteases/genética , Peptídeo Sintases/genética , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética
18.
Electrophoresis ; 33(2): 307-15, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22222975

RESUMO

This work presents the proteome profile of cultured human skin fibroblasts established from a patient affected by DNA ligase I (Lig I) deficiency syndrome, a rare disorder characterized by immunodeficiency, growth retardation and sun sensitivity. 2-DE (in the 3-10 and 4-7 pH ranges) was the separation technique used for the production of maps. MALDI-TOF/MS and LC-MS/MS were the mass spectrometry platforms applied for the identification of proteins in gel spots. A total of 154 proteins, including 41 never detected before in skin fibroblasts with this approach, were identified in gel spots analyzed. This newly generated extensive database provides for the first time a global picture of abundant proteins in 46BR.1G1 skin fibroblasts. While being relevant to the particular disorder considered, these results may be regarded as an intriguing starting point on the way to achieve a reference map of the proteins highly expressed in an inherited syndrome with defect in DNA replication and repair pathways.


Assuntos
DNA Ligases/deficiência , Fibroblastos/metabolismo , Proteoma/análise , Linhagem Celular Transformada , Cromatografia Líquida , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/química , DNA Ligase Dependente de ATP , Eletroforese em Gel Bidimensional , Ribonucleoproteínas Nucleares Heterogêneas/análise , Ribonucleoproteínas Nucleares Heterogêneas/química , Humanos , Concentração de Íons de Hidrogênio , Dobramento de Proteína , Proteínas/análise , Proteínas/química , Proteínas/classificação , Proteoma/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
19.
J Cell Biol ; 176(6): 807-18, 2007 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-17353358

RESUMO

In adult vertebrates, most cells are not in the cell cycle at any one time. Physiological nonproliferation states encompass reversible quiescence and permanent postmitotic conditions such as terminal differentiation and replicative senescence. Although these states appear to be attained and maintained quite differently, they might share a core proliferation-restricting mechanism. Unexpectedly, we found that all sorts of nonproliferating cells can be mitotically reactivated by the sole suppression of histotype-specific cyclin-dependent kinase (cdk) inhibitors (CKIs) in the absence of exogenous mitogens. RNA interference-mediated suppression of appropriate CKIs efficiently triggered DNA synthesis and mitosis in established and primary terminally differentiated skeletal muscle cells (myotubes), quiescent human fibroblasts, and senescent human embryo kidney cells. In serum-starved fibroblasts and myotubes alike, cell cycle reactivation was critically mediated by the derepression of cyclin D-cdk4/6 complexes. Thus, both temporary and permanent growth arrest must be actively maintained by the constant expression of CKIs, whereas the cell cycle-driving cyclins are always present or can be readily elicited. In principle, our findings could find wide application in biotechnology and tissue repair whenever cell proliferation is limiting.


Assuntos
Ciclo Celular/fisiologia , Proliferação de Células , Proteínas Inibidoras de Quinase Dependente de Ciclina/antagonistas & inibidores , Animais , Diferenciação Celular , Células Cultivadas , Senescência Celular/fisiologia , Ciclina D3 , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/fisiologia , Ciclinas/metabolismo , Replicação do DNA/fisiologia , Humanos , Camundongos , Fibras Musculares Esqueléticas/citologia , Interferência de RNA
20.
Exp Cell Res ; 317(20): 2958-68, 2011 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21978910

RESUMO

The assumption that cells are temporally organized systems, i.e. showing relevant dynamics of their state variables such as gene expression or protein and metabolite concentration, while tacitly given for granted at the molecular level, is not explicitly taken into account when interpreting biological experimental data. This conundrum stems from the (undemonstrated) assumption that a cell culture, the actual object of biological experimentation, is a population of billions of independent oscillators (cells) randomly experiencing different phases of their cycles and thus not producing relevant coordinated dynamics at the population level. Moreover the fact of considering reproductive cycle as by far the most important cyclic process in a cell resulted in lower attention given to other rhythmic processes. Here we demonstrate that growing yeast cells show a very repeatable and robust cyclic variation of the concentration of proteins with different cellular functions. We also report experimental evidence that the mechanism governing this basic oscillator and the cellular entrainment is resistant to external chemical constraints. Finally, cell growth is accompanied by cyclic dynamics of medium pH. These cycles are observed in batch cultures, different from the usual continuous cultures in which yeast metabolic cycles are known to occur, and suggest the existence of basic, spontaneous, collective and synchronous behaviors of the cell population as a whole.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , DNA Helicases/metabolismo , Proteínas Ribossômicas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Ciclo Celular/genética , Proliferação de Células , Cicloeximida/farmacologia , Concentração de Íons de Hidrogênio , Proteínas Ribossômicas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Vanadatos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA