Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Physiol Rev ; 100(4): 1599-1619, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32666886

RESUMO

Sentrin/small ubiquitin-like modifier (SUMO) is protein modification pathway that regulates multiple biological processes, including cell division, DNA replication/repair, signal transduction, and cellular metabolism. In this review, we will focus on recent advances in the mechanisms of disease pathogenesis, such as cancer, diabetes, seizure, and heart failure, which have been linked to the SUMO pathway. SUMO is conjugated to lysine residues in target proteins through an isopeptide linkage catalyzed by SUMO-specific activating (E1), conjugating (E2), and ligating (E3) enzymes. In steady state, the quantity of SUMO-modified substrates is usually a small fraction of unmodified substrates due to the deconjugation activity of the family Sentrin/SUMO-specific proteases (SENPs). In contrast to the complexity of the ubiquitination/deubiquitination machinery, the biochemistry of SUMOylation and de-SUMOylation is relatively modest. Specificity of the SUMO pathway is achieved through redox regulation, acetylation, phosphorylation, or other posttranslational protein modification of the SUMOylation and de-SUMOylation enzymes. There are three major SUMOs. SUMO-1 usually modifies a substrate as a monomer; however, SUMO-2/3 can form poly-SUMO chains. The monomeric SUMO-1 or poly-SUMO chains can interact with other proteins through SUMO-interactive motif (SIM). Thus SUMO modification provides a platform to enhance protein-protein interaction. The consequence of SUMOylation includes changes in cellular localization, protein activity, or protein stability. Furthermore, SUMO may join force with ubiquitin to degrade proteins through SUMO-targeted ubiquitin ligases (STUbL). After 20 yr of research, SUMO has been shown to play critical roles in most, if not all, biological pathways. Thus the SUMO enzymes could be targets for drug development to treat human diseases.


Assuntos
Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Imunidade Adaptativa/fisiologia , Animais , Doenças Cardiovasculares/metabolismo , Diabetes Mellitus/metabolismo , Humanos , Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética
2.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34789577

RESUMO

Recognition of invading pathogens by Toll-like receptors (TLRs) activates innate immunity through signaling pathways that involved multiple protein kinases and phosphatases. We previously demonstrated that somatic nuclear autoantigenic sperm protein (sNASP) binds to TNF receptor-associated factor 6 (TRAF6) in the resting state. Upon TLR4 activation, a signaling complex consisting of TRAF6, sNASP, interleukin (IL)-1 receptor-associated kinase 4, and casein kinase 2 (CK2) is formed. CK2 then phosphorylates sNASP to release phospho-sNASP (p-sNASP) from TRAF6, initiating downstream signaling pathways. Here, we showed that protein phosphatase 4 (PP4) is the specific sNASP phosphatase that negatively regulates TLR4-induced TRAF6 activation and its downstream signaling pathway. Mechanistically, PP4 is directly recruited by phosphorylated sNASP to dephosphorylate p-sNASP to terminate TRAF6 activation. Ectopic expression of PP4 specifically inhibited sNASP-dependent proinflammatory cytokine production and downstream signaling following bacterial lipopolysaccharide (LPS) treatment, whereas silencing PP4 had the opposite effect. Primary macrophages and mice infected with recombinant adenovirus carrying a gene encoding PP4 (Ad-PP4) showed significant reduction in IL-6 and TNF-α production. Survival of Ad-PP4-infected mice was markedly increased due to a better ability to clear bacteria in a sepsis model. These results indicate that the serine/threonine phosphatase PP4 functions as a negative regulator of innate immunity by regulating the binding of sNASP to TRAF6.


Assuntos
Autoantígenos/metabolismo , Caseína Quinase II/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Autoantígenos/genética , Caseína Quinase II/genética , Proteínas de Ciclo Celular/genética , Quimiocinas/metabolismo , Citocinas , Imunidade Inata , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoproteínas Fosfatases/genética , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Fator 6 Associado a Receptor de TNF/genética , Receptor 4 Toll-Like/genética , Receptores Toll-Like/metabolismo
3.
J Biol Chem ; 297(4): 101183, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34509475

RESUMO

Sentrin/small ubiquitin-like modifier (SUMO)-specific protease 2 (SENP2)-deficient mice develop spontaneous seizures in early life because of a marked reduction in M currents, which regulate neuronal membrane excitability. We have previously shown that hyper-SUMOylation of the Kv7.2 and Kv7.3 channels is critically involved in the regulation of the M currents conducted by these potassium voltage-gated channels. Here, we show that hyper-SUMOylation of the Kv7.2 and Kv7.3 proteins reduced binding to the lipid secondary messenger PIP2. CaM1 has been shown to be tethered to the Kv7 subunits via hydrophobic motifs in its C termini and implicated in the channel assembly. Mutation of the SUMOylation sites on Kv7.2 and Kv7.3 specifically resulted in decreased binding to CaM1 and enhanced CaM1-mediated assembly of Kv7.2 and Kv7.3, whereas hyper-SUMOylation of Kv7.2 and Kv7.3 inhibited channel assembly. SENP2-deficient mice exhibited increased acetylcholine levels in the brain and the heart tissue because of increases in the vagal tone induced by recurrent seizures. The SENP2-deficient mice develop seizures followed by a period of sinus pauses or atrioventricular conduction blocks. Chronic administration of the parasympathetic blocker atropine or unilateral vagotomy significantly prolonged the life of the SENP2-deficient mice. Furthermore, we showed that retigabine, an M-current opener, reduced the transcription of SUMO-activating enzyme SAE1 and inhibited SUMOylation of the Kv7.2 and Kv7.3 channels, and also prolonged the life of SENP2-deficient mice. Taken together, the previously demonstrated roles of PIP2, CaM1, and retigabine on the regulation of Kv7.2 and Kv7.3 channel function can be explained by their roles in regulating SUMOylation of this critical potassium channel.


Assuntos
Cisteína Endopeptidases/metabolismo , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Sistemas do Segundo Mensageiro , Sumoilação , Motivos de Aminoácidos , Animais , Encéfalo/metabolismo , Cisteína Endopeptidases/genética , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ3/genética , Camundongos , Camundongos Mutantes , Miocárdio/metabolismo , Convulsões/genética , Convulsões/metabolismo , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo
4.
J Neurochem ; 161(6): 478-491, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35583089

RESUMO

Glutamate N-methyl-d-aspartate (NMDA) receptors (NMDARs) and Kv7/M channels are importantly involved in regulating neuronal activity involved in various physiological and pathological functions. Corticotropin-releasing hormone (CRH)-expressing neurons in the central nucleus of the amygdala (CeA) critically mediate autonomic response during stress. However, the interaction between NMDA receptors and Kv7/M channels in the CRHCeA neurons remains unclear. In this study, we identified rat CRHCeA neurons through the expression of an AAV viral vector-mediated enhanced green fluorescent protein (eGFP) driven by the rat CRH promoter. M-currents carried by Kv7/M channels were recorded using the whole-cell patch-clamp approach in eGFP-tagged CRHCeA neurons in brain slices. Acute exposure to NMDA significantly reduced M-currents recorded from the CRHCeA neurons. NMDA-induced suppression of M-currents was eliminated by chelating intracellular Ca2+ , supplying phosphatidylinositol 4,5-bisphosphate (PIP2) intracellularly, or blocking phosphoinositide3-kinase (PI3K). In contrast, inhibiting protein kinase C (PKC) or calmodulin did not alter NMDA-induced suppression of M-currents. Sustained exposure of NMDA decreased Kv7.3 membrane protein levels and suppressed M-currents, while the Kv7.2 expression levels remained unaltered. Pre-treatment of brain slices with PKC inhibitors alleviated the decreases in Kv7.3 and reduction of M-currents in CRHCeA neurons induced by NMDA. PKC inhibitors did not alter Kv7.2 and Kv7.3 membrane protein levels and M-currents in CRHCeA neurons. These data suggest that transient activation of NMDARs suppresses M-currents through the Ca2+ -dependent PI3K-PIP2 signaling pathway. In contrast, sustained activation of NMDARs reduces Kv7.3 protein expression and suppresses M-currents through a PKC-dependent pathway.


Assuntos
Núcleo Central da Amígdala , Hormônio Liberador da Corticotropina , Animais , Hormônio Liberador da Corticotropina/metabolismo , N-Metilaspartato/farmacologia , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/fisiologia
5.
Mol Cell ; 45(2): 210-21, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22284677

RESUMO

Small ubiquitin-like modifier (SUMO) modification has emerged as an important regulatory mechanism during embryonic development. However, it is not known whether SUMOylation plays a role in the development of the immune system. Here, we show that SUMO-specific protease 1 (SENP1) is essential for the development of early T and B cells. STAT5, a key regulator of lymphoid development, is modified by SUMO-2 and is specifically regulated by SENP1. In the absence of SENP1, SUMO-2 modified STAT5 accumulates in early lymphoid precursors, resulting in a block in its acetylation and subsequent signaling. These results demonstrate a crucial role of SENP1 in the regulation of STAT5 activation during early lymphoid development.


Assuntos
Linfócitos B/citologia , Endopeptidases/fisiologia , Fator de Transcrição STAT5/metabolismo , Linfócitos T/citologia , Animais , Linfócitos B/metabolismo , Linfócitos B/fisiologia , Diferenciação Celular/genética , Cisteína Endopeptidases , Endopeptidases/genética , Endopeptidases/metabolismo , Camundongos , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/metabolismo , Fator de Transcrição STAT5/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Linfócitos T/metabolismo , Linfócitos T/fisiologia
6.
J Biol Chem ; 293(11): 3965-3980, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29352108

RESUMO

Protein SUMOylation has been reported to play a role in innate immune response, but the enzymes, substrates, and consequences of the specific inflammatory signaling events are largely unknown. Reactive oxygen species (ROS) are abundantly produced during macrophage activation and required for Toll-like receptor 4 (TLR4)-mediated inflammatory signaling. Previously, we demonstrated that SENP3 is a redox-sensitive SUMO2/3 protease. To explore any links between reversible SUMOylation and ROS-related inflammatory signaling in macrophage activation, we generated mice with Senp3 conditional knock-out in myeloid cells. In bacterial lipopolysaccharide (LPS)-induced in vitro and in vivo inflammation models, we found that SENP3 deficiency markedly compromises the activation of TLR4 inflammatory signaling and the production of proinflammatory cytokines in macrophages exposed to LPS. Moreover, Senp3 conditional knock-out mice were significantly less susceptible to septic shock. Of note, SENP3 deficiency was associated with impairment in JNK phosphorylation. We found that MKK7, which selectively phosphorylates JNK, is a SENP3 substrate and that SENP3-mediated deSUMOylation of MKK7 may favor its binding to JNK. Importantly, ROS-dependent SENP3 accumulation and MKK7 deSUMOylation rapidly occurred after LPS stimulation. In conclusion, our findings indicate that SENP3 potentiates LPS-induced TLR4 signaling via deSUMOylation of MKK7 leading to enhancement in JNK phosphorylation and the downstream events. Therefore this work provides novel mechanistic insights into redox regulation of innate immune responses.


Assuntos
Imunidade Inata/imunologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , MAP Quinase Quinase 7/metabolismo , Macrófagos/patologia , Peptídeo Hidrolases/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Células Cultivadas , Cisteína Endopeptidases , Citocinas/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , MAP Quinase Quinase 7/genética , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Sumoilação
7.
Mol Cell ; 43(2): 180-91, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21777808

RESUMO

Activation of NF-κB, pivotal for immunity and oncogenesis, is tightly controlled by multiple feedback mechanisms. In response to DNA damage, SUMOylation of NEMO (NF-κB essential modulator) is critical for NF-κB activation; however, the SUMO proteases and feedback mechanisms involved remain unknown. Here we show that among the six known Sentrin/SUMO-specific proteases (SENPs), only SENP2 can efficiently associate with NEMO, deSUMOylate NEMO, and inhibit NF-κB activation induced by DNA damage. We further show that NF-κB induces SENP2 (and SENP1) transcription selectively in response to genotoxic stimuli, which involves ataxia telangiectasia mutated (ATM)-dependent histone methylation of SENP2 promoter κB regions and NF-κB recruitment. SENP2 null cells display biphasic NEMO SUMOylation and activation of IKK and NF-κB, and higher resistance to DNA damage-induced cell death. Our study establishes a self-attenuating feedback mechanism selective to DNA damage-induced signaling to limit NF-κB-dependent cell survival responses.


Assuntos
Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , NF-kappa B/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular , Células Cultivadas , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Metilação , Proteínas Serina-Treonina Quinases/metabolismo , Sumoilação , Proteínas Supressoras de Tumor/metabolismo
9.
Mol Cell ; 39(3): 333-45, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20705237

RESUMO

The replication protein A complex (RPA) plays a crucial role in DNA replication and damage response. However, it is not known whether this complex is regulated by the SUMOylation pathway. Here, we show that the 70 kDa subunit of RPA (RPA70) associates with a Sentrin/SUMO-specific protease, SENP6, in the nucleus to maintain RPA70 in a hypoSUMOylated state during S phase. Campothecin (CPT), an inducer of replication stress, dissociates SENP6 from RPA70, allowing RPA70 to be modified by a small ubiquitin-like modifier 2/3 (SUMO-2/3). RPA70 SUMOylation facilitates recruitment of Rad51 to the DNA damage foci to initiate DNA repair through homologous recombination (HR). Cell lines that expressed a RPA70 mutant that cannot be SUMOylated are defective in HR and have a marked increase in sensitivity to CPT. These results demonstrate that SUMOylation status of RPA70 plays a critical role in the regulation of DNA repair through homologous recombination.


Assuntos
Reparo do DNA/fisiologia , DNA Polimerase Dirigida por DNA/metabolismo , Complexos Multienzimáticos/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Fase S/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Linhagem Celular , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Replicação do DNA/fisiologia , DNA Polimerase Dirigida por DNA/genética , Humanos , Complexos Multienzimáticos/genética , Mutação , Recombinação Genética/fisiologia , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Ubiquitinas/genética
10.
Mol Cell ; 38(2): 191-201, 2010 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-20417598

RESUMO

SUMO-specific protease 2 (SENP2) has a broad de-SUMOylation activity in vitro. However, the biological function of SENP2 is largely unknown. Here, we show that deletion of SENP2 gene in mouse causes defects in the embryonic heart and reduces the expression of Gata4 and Gata6, which are essential for cardiac development. SENP2 regulates transcription of Gata4 and Gata6 mainly through alteration of occupancy of Pc2/CBX4, a polycomb repressive complex 1 (PRC1) subunit, on its promoters. We demonstrate that Pc2/CBX4 is a target of SENP2 in vivo and that SUMOylation is essential for Pc2/CBX4-mediated PRC1 recruitment to methylated histone 3 at K27 (H3K27me3). In SENP2 null embryos, SUMOylated Pc2/CBX4 accumulates and Pc2/CBX4 occupancy on the promoters of PcG target genes is markedly increased, leading to repression of Gata4 and Gata6 transcription. Our results reveal a critical role for de-SUMOylation in the regulation of PcG target gene expression.


Assuntos
Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Complexos Multienzimáticos/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Cisteína Endopeptidases , Embrião de Mamíferos , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/genética , Proteínas do Grupo Polycomb , Proteínas Repressoras , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética
11.
J Card Fail ; 22(6): 433-8, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27079675

RESUMO

BACKGROUND: Anthracycline chemotherapy is associated with an increased risk of developing heart failure (HF). The current standard for detecting HF or cardiotoxicity during chemotherapy involves episodic cardiac imaging typically at prescribed intervals and there are limited studies examining techniques beyond measuring left ventricular (LV) function. This study explores whether cardiac biomarkers troponin I (TnI) and B-type natriuretic peptide (BNP) could be part of a screening strategy for early detection of the development of cardiotoxicity in patients undergoing anthracycline chemotherapy. METHODS AND RESULTS: Patients were enrolled from a single medical center. Cardiac biomarkers (TnI, BNP) were measured before and within 24 hours after completion of anthracycline administration for each cycle of therapy. Cardiac imaging was obtained at baseline and at completion of chemotherapy (commonly at 6 or 12 months) or based on clinical suspicion of a cardiac event. Of the enrolled 109 patients, 11 (10.1%) experienced cardiac events; all of these patients had at least 1 BNP value >100 pg/mL before the cardiac event. Significant reduction in LV ejection fraction as defined for cardiotoxicity occurred in only 3 of 10 patients (30%) with a cardiac event. CONCLUSIONS: The use of cardiac biomarkers, particularly BNP, may allow early detection of cardiotoxicity related to anthracycline chemotherapy.


Assuntos
Antraciclinas/efeitos adversos , Cardiopatias/induzido quimicamente , Peptídeo Natriurético Encefálico/sangue , Neoplasias/tratamento farmacológico , Sistemas Automatizados de Assistência Junto ao Leito , Troponina I/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antraciclinas/uso terapêutico , Biomarcadores/sangue , Cardiotoxicidade/sangue , Cardiotoxicidade/diagnóstico , Estudos de Viabilidade , Feminino , Cardiopatias/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Adulto Jovem
12.
J Mol Cell Cardiol ; 79: 115-22, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25446185

RESUMO

Defect in mitochondrial biogenesis and cardiac energy metabolism is a critical contributing factor to cardiac hypertrophy and heart failure. Sentrin/SUMO specific protease 1 (SENP1) mediated regulation of PGC-1α transcriptional activity plays an essential role in mitochondrial biogenesis and mitochondrial function. However, whether SENP1 plays a role in cardiac hypertrophy and failure is unknown. We investigated whether alteration in SENP1 expression affects cardiomyopathy and the underlying mechanism. In our present study, we found that the expression of SENP1 was induced in mouse and human failing hearts associated with induced expression of mitochondrial genes. SENP1 expression in cardiomyocytes was induced by hypertrophic stimuli through calcium/calcineurin-NFAT3. SENP1 regulated mitochondrial gene expression by de-SUMOylation of MEF-2C, which enhanced MEF-2C-mediated PGC-1α transcription. Genetic induction of SENP1 led to mitochondrial dysregulation and cardiac dysfunction in vivo. Our data showed that pathogenesis of cardiomyopathy is attributed by SENP1 mediated regulation of mitochondrial abnormities. SENP1 up-regulation in diseased heart is mediated via calcineurin-NFAT/MEF2C-PGC-1α pathway.


Assuntos
Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Endopeptidases/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Animais , Calcineurina/metabolismo , Cardiomiopatias/genética , Cardiomiopatias/fisiopatologia , Cisteína Endopeptidases , Endopeptidases/genética , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Fatores de Transcrição MEF2/metabolismo , Camundongos , Mitocôndrias/ultraestrutura , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/ultraestrutura , Fatores de Transcrição NFATC/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Sumoilação , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
13.
J Biol Chem ; 289(6): 3288-93, 2014 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-24344126

RESUMO

Sentrin/small ubiquitin-like modifier (SUMO)-specific protease 2 (SENP2) has broad de-SUMOylation activities in vitro, which is essential for embryonic heart development. Here, we show that myostatin, a key factor in skeletal muscle development, is markedly reduced in Senp2(-/-) mouse embryonic fibroblast cells and embryos. SENP2 regulates the transcription of myostatin mainly through de-SUMOylation of MEF2A. Silencing SENP2 can reduce myostatin expression and, therefore, promote myogenesis of skeletal muscle. These results reveal the important role of SENP2 in the regulation of myostatin expression and myogenesis.


Assuntos
Cisteína Endopeptidases/metabolismo , Regulação da Expressão Gênica/fisiologia , Desenvolvimento Muscular/fisiologia , Miostatina/biossíntese , Animais , Linhagem Celular , Cisteína Endopeptidases/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Camundongos , Camundongos Knockout , Miostatina/genética , Sumoilação/fisiologia
14.
Circ Res ; 113(6): 754-64, 2013 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-23989717

RESUMO

The cardiovascular care of cancer patients (cardio-oncology) has emerged as a new discipline in clinical medicine, given recent advances in cancer therapy, and is driven by the cardiovascular complications that occur as a direct result of cancer therapy. Traditional therapies such as anthracyclines and radiation have been recognized for years to have cardiovascular complications. Less expected were the cardiovascular effects of targeted cancer therapies, which were initially thought to be specific to cancer cells and would spare any adverse effects on the heart. Cancers are typically driven by mutations, translocations, or overexpression of protein kinases. The majority of these mutated kinases are tyrosine kinases, though serine/threonine kinases also play key roles in some malignancies. Several agents were developed to target these kinases, but many more are in development. Major successes have been largely restricted to agents targeting human epidermal growth factor receptor-2 (mutated or overexpressed in breast cancer), BCR-ABL (chronic myelogenous leukemia and some cases of acute lymphoblastic leukemia), and c-Kit (gastrointestinal stromal tumor). Other agents targeting more complex malignancies, such as advanced solid tumors, have had successes, but have not extended life to the degree seen with chronic myelogenous leukemia. Years before the first targeted therapy, Judah Folkman correctly proposed that to address solid tumors one had to target the inherent neoangiogenesis. Unfortunately, emerging evidence confirms that angiogenesis inhibitors cause cardiac complications, including hypertension, thrombosis, and heart failure. And therein lies the catch-22. Nevertheless, cardio-oncology has the potential to be transformative as the human cardiomyopathies that arise from targeted therapies can provide insights into the normal function of the heart.


Assuntos
Inibidores da Angiogênese/efeitos adversos , Cardiomiopatias/induzido quimicamente , Animais , Antraciclinas/efeitos adversos , Cardiotoxinas/toxicidade , Humanos
15.
Circ Res ; 112(6): 911-23, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23381569

RESUMO

RATIONALE: Disturbed flow induces proinflammatory and apoptotic responses in endothelial cells, causing them to become dysfunctional and subsequently proatherogenic. OBJECTIVE: Although a possible link between SUMOylation of p53 and ERK5 detected during endothelial apoptosis and inflammation has been suggested, the mechanistic insights, especially under the proatherogenic flow condition, remain largely unknown. METHODS AND RESULTS: SUMOylation of p53 and ERK5 was induced by disturbed flow but not by steady laminar flow. To examine the role of the disturbed flow-induced p53 and ERK5 SUMOylation, we used de-SUMOylation enzyme of sentrin/Small Ubiquitin-like MOdifier (SUMO)-specific protease 2 deficiency (Senp2(+/-)) mice and observed a significant increase in endothelial apoptosis and adhesion molecule expression both in vitro and in vivo. These increases, however, were significantly inhibited in endothelial cells overexpressing p53 and ERK5 SUMOylation site mutants. Senp2(+/-) mice exhibited increased leukocyte rolling along the endothelium, and accelerated formation of atherosclerotic lesions was observed in Senp2(+/-)/Ldlr(-/-), but not in Senp2(+/+)/Ldlr(-/-), mice fed a high-cholesterol diet. Notably, the extent of lesion size in the aortic arch of Senp2(+/-)/Ldlr(-/-) mice was much larger than that in the descending aorta, also suggesting a crucial role of the disturbed flow-induced SUMOylation of proteins, including p53 and ERK5 in atherosclerosis formation. CONCLUSIONS: These data show the unique role of sentrin/SUMO-specific protease 2 on endothelial function under disturbed flow and suggest that SUMOylation of p53 and ERK5 by disturbed flow contributes to the atherosclerotic plaque formation. Molecules involved in this newly discovered signaling will be useful targets for controlling endothelial cells dysfunction and consequently atherosclerosis formation.


Assuntos
Aterosclerose/etiologia , Células Endoteliais/metabolismo , Hemorreologia/fisiologia , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Proteína SUMO-1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/fisiologia , Transplante de Medula Óssea , Moléculas de Adesão Celular/metabolismo , Regulação para Baixo , Selectina E/metabolismo , Endotélio Vascular/metabolismo , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo
16.
Proc Natl Acad Sci U S A ; 109(43): 17466-71, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23045645

RESUMO

Two Sentrin/small ubiquitin-like modifier (SUMO)-specific protease 7 (SENP7) variants are naturally expressed in breast epithelia. Breast cancer (BCa) onset down-regulates the short SENP7 splice variant (SENP7S) and enhances the long transcript (SENP7L). Here, we show that SENP7L induction promotes gene expression profiles that favor aberrant proliferation and initiate epithelial-mesenchymal transition (EMT). SENP7L exhibits an interaction domain for the epigenetic remodeler heterochromatin protein 1 α (HP1α) and isopeptidase activity against SUMO-modified HP1α. Loss of this interaction domain, as observed with SENP7S, favors HP1α SUMOylation. SUMOylated HP1α is enriched at E2F-responsive and mesenchymal gene promoters, silences transcription of these genes, and promotes cellular senescence. Elevated SENP7L renders HP1α hypo-SUMOylated, which relieves transcriptional repression of the same genes and concurrently decreases transcription of epithelial-promoting genes via an HP1α-independent mechanism. Consequently, SENP7L levels correlate with EMT, motility, and invasiveness of BCa cells. Stable knockdown of elevated SENP7L levels lessens the dissemination of highly metastatic BCa cells to the lungs from primary implantation sites in in vivo studies. Thus, differential splicing of the SENP7 regulates either tumor suppression or progression.


Assuntos
Endopeptidases/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Endopeptidases/química , Endopeptidases/genética , Endopeptidases/fisiologia , Perfilação da Expressão Gênica , Humanos , Dados de Sequência Molecular , RNA Mensageiro/genética , Homologia de Sequência de Aminoácidos
17.
EMBO J ; 29(22): 3773-86, 2010 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-20924358

RESUMO

The molecular chaperone heat shock protein 90 (Hsp90) and the co-chaperone/ubiquitin ligase carboxyl terminus of Hsc70-interacting protein (CHIP) control the turnover of client proteins. How this system decides to stabilize or degrade the client proteins under particular physiological or pathological conditions is unclear. We report here a novel client protein, the SUMO2/3 protease SENP3, that is sophisticatedly regulated by CHIP and Hsp90. SENP3 is maintained at a low basal level under non-stress condition due to Hsp90-independent CHIP-mediated ubiquitination. Upon mild oxidative stress, SENP3 undergoes thiol modification, which recruits Hsp90. Hsp90/SENP3 association protects SENP3 from CHIP-mediated ubiquitination and subsequent degradation, but this effect of Hsp90 requires the presence of CHIP. Our data demonstrate for the first time that CHIP and Hsp90 interplay with a client alternately under non-stress and stress conditions, and the choice between stabilization and degradation is made by the redox state of the client. In addition, enhanced SENP3/Hsp90 association is found in cancer. These findings provide new mechanistic insight into how cells regulate the SUMO protease in response to oxidative stress.


Assuntos
Cisteína Endopeptidases/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Cisteína/metabolismo , Estabilidade Enzimática , Células HEK293 , Células HeLa , Humanos , Oxirredução , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Ubiquitinação
18.
J Biol Chem ; 287(53): 44464-70, 2012 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-23152500

RESUMO

Peroxisome proliferator-activated receptor γ (PPARγ) coactivator 1α (PGC-1α) is a master regulator of mitochondrial biogenesis in response to changes in the cellular environment, physiological or pathological status of mammals. PGC-1α is known to be modified by SUMO (Small Ubiquitin-like Modifier). However, it is not known whether SUMOylation could affect the function of PGC-1α in mitochondrial biogenesis and that how PGC-1α SUMOylation is regulated. In this study, we have identified the role of Sentrin/SUMO-specific protease 1 (SENP1) as a specific SUMO protease to regulate SUMOylation status of PGC-1α. More importantly, we have also found that SENP1 promotes PGC-1α transcription activity, which is essential for the expression of mitochondrial genes and subsequently mitochondrial biogenesis. Thus, we reveal that the SUMOylation of PGC-1α controlled by SENP1 plays an important role in mitochondrial biogenesis and function.


Assuntos
Endopeptidases/metabolismo , Transativadores/metabolismo , Animais , Cisteína Endopeptidases , Endopeptidases/genética , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/enzimologia , Mitocôndrias/genética , Mitocôndrias/metabolismo , Renovação Mitocondrial , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Sumoilação , Transativadores/genética , Fatores de Transcrição
19.
EMBO J ; 28(18): 2748-62, 2009 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-19680224

RESUMO

The physiological function of Sentrin/SUMO-specific proteases (SENPs) remains largely unexplored, and little is known about the regulation of SENPs themselves. Here, we show that a modest increase of reactive oxygen species (ROS) regulates SENP3 stability and localization. We found that SENP3 is continuously degraded through the ubiquitin-proteasome pathway under basal condition and that ROS inhibit this degradation. Furthermore, ROS causes SENP3 to redistribute from the nucleoli to the nucleoplasm, allowing it to regulate nuclear events. The stabilization and redistribution of SENP3 correlate with an increase in the transcriptional activity of the hypoxia-inducing factor-1 (HIF-1) under mild oxidative stress. ROS-enhanced HIF-1 transactivation is blocked by SENP3 knockdown. The de-SUMOylating activity of SENP3 is required for ROS-induced increase of HIF-1 transactivation, but the true substrate of SENP3 is the co-activator of HIF-1 alpha, p300, rather than HIF-1 alpha itself. Removing SUMO2/3 from p300 enhances its binding to HIF-1 alpha. In vivo nude mouse xenografts overexpressing SENP3 are more angiogenic. Taken together, our results identify SENP3 as a redox sensor that regulates HIF-1 transcriptional activity under oxidative stress through the de-SUMOylation of p300.


Assuntos
Cisteína Endopeptidases/metabolismo , Proteína p300 Associada a E1A/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Estresse Oxidativo , Proteína SUMO-1/metabolismo , Ativação Transcricional , Animais , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Oxirredução , Espécies Reativas de Oxigênio
20.
Nat Cell Biol ; 8(10): 1171-7, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16998474

RESUMO

Neddylation has an important role in ubiquitin-mediated protein degradation through modification of cullins, which are the main substrates for NEDD8 modification. Here, we show that breast cancer-associated protein 3 (BCA3) is a NEDD8 substrate. BCA3 suppressed NFkappaB-dependent transcription through its ability to bind to p65 and the cyclin D1 promoter in a neddylation-dependent manner. Transcriptional suppression mediated by BCA3 may be attributed to the ability of neddylated BCA3 to recruit SIRT1, a class III histone deacetylase. Silencing of endogenous BCA3 in DU145 and MCF7 cells enhanced NFkappaB transcription and inhibited tumour necrosis factor (TNF)alpha-induced apoptosis. Conversely, BCA3 silencing could be reversed by over-expression of wild-type BCA3 and SENP8, a NEDD8-specific protease, but not by neddylation-deficient BCA3 or a SENP8 mutant. These results provide a crucial link between neddylation and transcriptional regulation by SIRT1, a NAD-dependent histone deacetylase that prolongs life span in yeast and worms.


Assuntos
Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Sirtuínas/metabolismo , Fator de Transcrição RelA/metabolismo , Ubiquitinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose , Neoplasias da Mama , Células COS , Chlorocebus aethiops , Ciclina D , Ciclinas/genética , Ciclinas/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo , Feminino , Inativação Gênica , Células HeLa , Humanos , Masculino , Proteína NEDD8 , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata , Sirtuína 1 , Sirtuínas/genética , Fator de Transcrição RelA/genética , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA