Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(49): e2214935119, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36442094

RESUMO

The 53BP1-RIF1 pathway restricts the resection of DNA double-strand breaks (DSBs) and promotes blunt end-ligation by non-homologous end joining (NHEJ) repair. The Shieldin complex is a downstream effector of the 53BP1-RIF1 pathway. Here, we identify a component of this pathway, CCAR2/DBC1, which is also required for restriction of DNA end-resection. CCAR2 co-immunoprecipitates with the Shieldin complex, and knockout of CCAR2 in a BRCA1-deficient cell line results in elevated DSB end-resection, RAD51 loading, and PARP inhibitor (PARPi) resistance. Knockout of CCAR2 is epistatic with knockout of other Shieldin proteins. The S1-like RNA-binding domain of CCAR2 is required for its interaction with the Shieldin complex and for suppression of DSB end-resection. CCAR2 functions downstream of the Shieldin complex, and CCAR2 knockout cells have delayed resolution of Shieldin complex foci. Forkhead-associated (FHA)-dependent targeting of CCAR2 to DSB sites re-sensitized BRCA1-/-SHLD2-/- cells to PARPi. Taken together, CCAR2 is a functional component of the 53BP1-RIF1 pathway, promotes the refill of resected DSBs, and suppresses homologous recombination.


Assuntos
Quebras de DNA de Cadeia Dupla , Inibidores de Poli(ADP-Ribose) Polimerases , Reparo do DNA por Junção de Extremidades , Recombinação Homóloga , DNA
2.
J Biol Chem ; 298(2): 101496, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34921839

RESUMO

Deleted in Breast Cancer 1 (DBC1) is an important metabolic sensor. Previous studies have implicated DBC1 in various cellular functions, notably cell proliferation, apoptosis, histone modification, and adipogenesis. However, current reports about the role of DBC1 in tumorigenesis are controversial and designate DBC1 alternatively as a tumor suppressor or a tumor promoter. In the present study, we report that polyoma small T antigen (PyST) associates with DBC1 in mammalian cells, and this interaction leads to the posttranslational downregulation of DBC1 protein levels. When coexpressed, DBC1 overcomes PyST-induced mitotic arrest and promotes the exit of cells from mitosis. Using both transient and stable modes of PyST expression, we also show that cellular DBC1 is subjected to degradation by LKB1, a tumor suppressor and cellular energy sensor kinase, in an AMP kinase-independent manner. Moreover, LKB1 negatively regulates the phosphorylation as well as activity of the prosurvival kinase AKT1 through DBC1 and its downstream pseudokinase substrate, Tribbles 3 (TRB3). Using both transient transfection and stable cell line approaches as well as soft agar assay, we demonstrate that DBC1 has oncogenic potential. In conclusion, our study provides insight into a novel signaling axis that connects LKB1, DBC1, TRB3, and AKT1. We propose that the LKB1-DBC1-AKT1 signaling paradigm may have an important role in the regulation of cell cycle and apoptosis and consequently tumorigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Antígenos Virais de Tumores , Proteínas de Ciclo Celular , Proteínas do Tecido Nervoso , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose , Carcinogênese , Proteínas de Ciclo Celular/metabolismo , Regulação para Baixo , Humanos , Proteínas do Tecido Nervoso/metabolismo , Sirtuína 1/metabolismo
3.
Mol Cell Neurosci ; 123: 103781, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36122891

RESUMO

The protein DBC1 is the main SIRT1 regulator known so far, and by doing so, it is involved in the regulation of energy metabolism, especially in liver and fat adipose tissue. DBC1 also has an important function in cell cycle progression and regulation in cancer cells, affecting tumorigenesis. We recently showed that during quiescence, non-transformed cells need DBC1 in order to re-enter and progress through the cell cycle. Moreover, we showed that deletion of DBC1 affects cell cycle progression during liver regeneration. This novel concept prompted us to evaluate the role of DBC1 during adult neurogenesis, where transition from quiescence to proliferation in neuronal progenitors is key and tightly regulated. Herein, we analyzed several markers of cell cycle expressed in the dentate gyrus of the hippocampus of controls and DBC1 KO adult mice. Our results suggest a reduced number of neuroblasts therein present, probably due to a decline of neuroblast generation or an impairment in neural differentiation. In agreement with this, we also found that adult DBC1 KO mice had a reduction in the volume of the granule cell layer of the dentate gyrus. Interestingly, behavioral analysis of KO and control mice revealed that deletion of DBC1 parallels to specific cognitive impairments, concerning learning and possibly memory formation. Our results show, for the first time, that DBC1 plays an active role in the nervous system. In particular, specific anatomical and behavioral changes are observed when is absent.


Assuntos
Células-Tronco Neurais , Neurogênese , Camundongos , Animais , Camundongos Knockout , Neurogênese/fisiologia , Hipocampo/metabolismo , Células-Tronco Neurais/metabolismo , Cognição/fisiologia , Giro Denteado , Camundongos Endogâmicos C57BL
4.
Proc Natl Acad Sci U S A ; 117(12): 6509-6520, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32152128

RESUMO

Among all of the Super Elongation Complex (SEC) components, ELL1 (also known as ELL) is the only bona fide elongation factor that directly stimulates transcription elongation by RNA polymerase II. However, the mechanism(s) of functional regulation of ELL1 (referred to as ELL hereafter), through its stabilization, is completely unknown. Here, we report a function of human DBC1 in regulating ELL stability involving HDAC3, p300, and Siah1. Mechanistically, we show that p300-mediated site-specific acetylation increases, whereas HDAC3-mediated deacetylation decreases, ELL stability through polyubiquitylation by the E3 ubiquitin ligase Siah1. DBC1 competes with HDAC3 for the same binding sites on ELL and thus increases its acetylation and stability. Knockdown of DBC1 reduces ELL levels and expression of a significant number of genes, including those involved in glucose metabolism. Consistently, Type 2 diabetes patient-derived peripheral blood mononuclear cells show reduced expression of DBC1 and ELL and associated key target genes required for glucose homeostasis. Thus, we describe a pathway of regulating stability and functions of key elongation factor ELL for expression of diverse sets of genes, including ones that are linked to Type 2 diabetes pathogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína p300 Associada a E1A/metabolismo , Regulação da Expressão Gênica , Histona Desacetilases/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Elongação da Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Acetilação , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sítios de Ligação , Linhagem Celular , Diabetes Mellitus Tipo 2/patologia , Proteína p300 Associada a E1A/genética , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Histona Desacetilases/genética , Humanos , Leucócitos Mononucleares/metabolismo , Mutação , Ligação Proteica , Estabilidade Proteica , Transcrição Gênica , Fatores de Elongação da Transcrição/química , Fatores de Elongação da Transcrição/genética , Ubiquitinação
5.
Cancer Sci ; 111(10): 3416-3425, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33403784

RESUMO

Cell Cycle and Apoptosis Regulator 1 (CCAR1) and Cell Cycle and Apoptosis Regulator 2 (CCAR2) have emerged as key players in physiology and pathophysiology, with critical roles in the DNA damage response, nuclear receptor function, and Wnt signaling, among other activities. Contradictory reports exist on the functional duality of CCAR1 and CCAR2 as either tumor promoters or suppressors, suggesting that CCAR1 and CCAR2 have the hallmarks of gene chameleons. We review herein the mechanistic, preclinical, and human translational findings for CCAR1 and CCAR2, based on available RNA and protein expression data from human studies, The Cancer Genome Atlas (TCGA) data mining, gene knockout mouse models, and cell-based assays. Multiple factors contribute to the divergent activities of CCAR1 and CCAR2, including tissue type, mutation/genetic background, protein-protein interactions, dynamic regulation via posttranslational modifications, and alternative RNA splicing. An array of protein partners interact with CCAR1 and CCAR2 in the context of tumor promotion and suppression, including ß-catenin, androgen receptor, p21Cip1/Waf1, tumor protein p53 (p53), sirtuin 1, and histone deacetylase 3. Genetic changes frequently found in cancer, such as TP53 mutation, also serve as critical determinants of survival outcomes in cancer patients. This review seeks to provide the impetus for further investigation into CCAR1 and CCAR2 as potential master regulators of metabolism, aging, and cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ciclo Celular/genética , Neoplasias/genética , Animais , Modelos Animais de Doenças , Genes Supressores de Tumor , Humanos , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas
6.
J Biol Chem ; 292(38): 15952-15963, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28794159

RESUMO

Bladder cancer (BC) is the sixth most common cancer in the United States and is the number one cause of death among patients with urinary system malignancies. This makes the identification of invasive regulator(s)/effector(s) as the potential therapeutic targets for managing BC a high priority. p63 is a member of the p53 family of tumor suppressor genes/proteins, plays a role in the differentiation of epithelial tissues, and is believed to function as a tumor suppressor. However, it remains unclear whether and how p63 functions in BC cell invasion after tumorigenesis. Here, we show that p63α protein levels were much higher in mouse high-invasive BC tissues than in normal tissues. Our results also revealed that p63α is crucial for heat shock protein 70 (Hsp70) expression and subsequently increases the ability of BC invasion. Mechanistic experiments demonstrated that p63α can transcriptionally up-regulate Hsp70 expression, thereby promoting BC cell invasion via the Hsp70/Wasf3/Wave3/MMP-9 axis. We further show that E2F transcription factor 1 (E2F1) mediates p63α overexpression-induced Hsp70 transcription. We also found that p63α overexpression activates E2F1 transcription, which appears to be stimulated by p63α together with E2F1. Collectively, our results demonstrate that p63α is a positive regulator of BC cell invasion after tumorigenesis, providing significant insights into the biological function of p63α in BC and supporting the notion that p63α might be a potential target for invasive BC therapy.


Assuntos
Fator de Transcrição E2F1/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima , Neoplasias da Bexiga Urinária/patologia , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Fator de Transcrição E2F1/genética , Proteínas de Choque Térmico HSP70/genética , Humanos , Masculino , Camundongos , Invasividade Neoplásica , Transdução de Sinais , Fator de Transcrição Sp1/metabolismo , Transcrição Gênica , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo
7.
Mol Carcinog ; 57(12): 1803-1815, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30175866

RESUMO

The biological role and underlying mechanism of action of zinc-finger protein 326 (ZNF326) in malignant tumors, including breast cancer, are still not clear. In this study, we detected high expression of ZNF326 in breast cancer specimens (60/111, 54.1%) and breast cancer cell lines (7/7); the expression level of ZNF326 was inversely associated with advanced pTNM stage (P = 0.002), positive lymph node metastasis (P = 0.004), poor prognosis in patients with breast cancer (P = 0.0097), and ER/PR/Her2 status (P = 0.013). Meanwhile, the ectopic expression of ZNF326 significantly upregulated MMP7, EMT-related proteins (Snail and Slug), and cell cycle-related proteins (cyclinA2 and cyclinB1); downregulated E-cadherin expression; and promoted the proliferation and invasiveness of breast cancer cells both in vivo and in vitro. Mechanistically, co-immunoprecipitation and immunofluorescence assays both demonstrated that ZNF326 interacted with deleted in breast cancer-1 (DBC1) in breast cancer cells. Additionally, DBC1 knockdown eliminated the up-regulation of MMP7, EMT-related proteins, and cell cycle-related proteins as well as the enhanced proliferation and invasiveness induced by ZNF326. Therefore, we concluded that ZNF326 is highly expressed in breast cancer, is associated with poor prognosis, and plays a vital role in promoting the malignant phenotype of breast cancer cells by interacting with DBC1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/patologia , Proteínas de Transporte/metabolismo , Regulação para Cima , Animais , Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Células MCF-7 , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Estadiamento de Neoplasias , Transplante de Neoplasias , Prognóstico , Análise de Sobrevida
8.
Biochem Biophys Res Commun ; 494(3-4): 511-517, 2017 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-29106957

RESUMO

Deleted in Breast Cancer 1 (DBC1) is a regulatory protein involved in cell metabolism and cancer progression. Nevertheless, the expression and prognostic values of DBC1 in hepatocellular carcinoma (HCC) are still not well understood. The following study investigated the clinical significance and biological function of DBC1 in HCC. Briefly, overexpression of DBC1 at transcriptional and translational levels in human HCC tissues compared to adjacent normal tissues was observed using quantitative real-time polymerase chain reaction (qRT-PCR), western blot (WB) and immunohistochemistry (IHC) approach. Furthermore, upregulated DBC1 was significantly correlated with tumor size (p = 0.005), N stage (p = 0.016), M stage (p = 0.011), tumor differentiation (p < 0.001), and American Joint Committee on Cancer (AJCC) stage (p = 0.001). Moreover, Kaplan-Meier survival analysis revealed that DBC1 was an independent prognosis predictor for disease-free survival (DFS) (p < 0.001) and overall survival (OS) (p < 0.001). In addition, by using Cell Counting Kit-8 (CCK8) assays and colony formation assays, we found that the knockdown of DBC1 significantly suppressed the proliferation of HCC cells in vitro. To conclude, these findings demonstrated that DBC1 was essential in tumorigenesis and proliferation. Moreover, it was identified as a potential therapeutic target for HCC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidade , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidade , Idoso , Idoso de 80 Anos ou mais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , China/epidemiologia , Intervalo Livre de Doença , Humanos , Neoplasias Hepáticas/patologia , Pessoa de Meia-Idade , Terapia de Alvo Molecular/métodos , Prevalência , Prognóstico , Fatores de Risco , Estatística como Assunto , Taxa de Sobrevida , Regulação para Cima
9.
Int J Mol Sci ; 18(5)2017 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-28481291

RESUMO

Brown adipose tissue thermogenesis at the cost of energy is not only important for the development of obesity, but also possesses great promise in anti-obesity treatment. Uncoupling protein 1 (UCP1) expression has been reported to be under control of the intracellular deacetylase SIRT1. Here, we investigated the effect and mechanism of inflammation and sirtuin-1 (SIRT1) activation on the induction of thermogenic genes in immortalized brown adipocytes incubated with LPS or IL1ß and mice with elevated inflammatory tone. In vitro stimulation of brown adipocytes with dibutyryl cyclic adenosine monophosthate (dbcAMP) reduced the expression of deleted in breast cancer-1 (Dbc1) (SIRT1 inhibitor) and increased the Ucp1 expression. Silencing of SIRT1 attenuated dbcAMP induction of Ucp1. In contrast, IL1ß increased the expression of Dbc1 and greatly reduced the induction of Ucp1. Similarly, in vivo studies revealed decreased expression of Ucp1 in brown adipose tissue (BAT) in mice chronically infused with LPS. Resveratrol, a known SIRT1 activator, partly rescued the Ucp1 downregulation by inflammation in both the cell cultures and mice. Here, we describe how the expression of Ucp1 in BAT is controlled via SIRT1 and is reduced under inflammation and can be rescued by SIRT1 activation by resveratrol. We suggest the reduced UCP1 expression under inflammation is mediated by the increased expression of DBC1, which inhibits SIRT1 activity.


Assuntos
Adipócitos Marrons/metabolismo , Regulação para Baixo , Proteínas do Tecido Nervoso/metabolismo , Sirtuína 1/metabolismo , Proteína Desacopladora 1/genética , Adipócitos Marrons/efeitos dos fármacos , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Inflamação/metabolismo , Interleucina-1beta/farmacologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Resveratrol , Sirtuína 1/genética , Estilbenos/farmacologia , Proteína Desacopladora 1/metabolismo
10.
Int J Cancer ; 136(4): 797-809, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24962073

RESUMO

CK2α has diverse effects on the tumorigenesis owing to its kinase activity, which phosphorylates various proteins involved in tumorigenesis. We, therefore, investigated the expression and role of CK2α in the phosphorylation of deleted in breast cancer 1 (DBC1) in gastric carcinomas. We used 187 gastric carcinomas and human gastric cancer cells to investigate the roles and relationship between CK2α and DBC1 in gastric carcinomas. Positive expression of CK2α and phospho-DBC1 predicted shorter overall survival and relapse-free survival by univariate analysis. Especially, CK2α expression was an independent prognostic indicator for gastric carcinoma patients. In gastric carcinoma cells, CK2α was bound to DBC1 and phosphorylated DBC1. The phosphorylation of DBC1 by CK2α was evidenced by co-immunoprecipitation of CK2α and DBC1 in a GST pull-down assay, an in vitro kinase assay, and immunofluorescence staining. Inhibition of both CK2α and DBC1 decreased proliferation and invasive activity of cancer cells. Decreased migration and invasive activity was associated with a downregulation of MMP2, MMP9 and the epithelial-mesenchymal transition. A mutation at the phosphorylation site of DBC1 also downregulated the signals related with the epithelial-mesenchymal transition. Our study demonstrated that CK2α is an independent prognostic indicator for gastric carcinoma patients and is involved in tumorigenesis by regulating the phosphorylation of DBC1. In addition, the blocking of CK2α and DBC1 inhibited the proliferation and invasive potential of gastric cancer cells. Therefore, our study suggests that CK2α-DBC1 pathway might be a new therapeutic target for the treatment of gastric carcinoma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenocarcinoma/enzimologia , Caseína Quinase II/fisiologia , Neoplasias Gástricas/enzimologia , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Movimento Celular , Proliferação de Células , Progressão da Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Fosforilação , Prognóstico , Processamento de Proteína Pós-Traducional , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia
11.
Int J Cancer ; 136(1): 55-64, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24824780

RESUMO

The mutated in colorectal cancer (MCC) is a multifunctional gene showing loss of expression in colorectal and liver cancers. MCC mutations can drive colon carcinogenesis in the mouse and in vitro experiments suggest that loss of MCC function promotes cancer through several important cellular pathways. In particular, the MCC protein is known to regulate beta-catenin (ß-cat) signaling, but the mechanism is poorly understood. Here we show that the ß-cat repressor function of MCC is strongly impaired by the presence of a disease-associated mutation. We also identify deleted in breast cancer 1 (DBC1) as a new MCC interacting partner and regulator of ß-cat signaling. RNA interference experiments show that DBC1 promotes ß-cat transcriptional activity and that the presence of DBC1 is required for MCC-mediated ß-cat repression. In contrast to all other DBC1 interacting partners, MCC does not interact through the DBC1 Leucine Zipper domain but with a glutamic-acid rich region located between the Nudix and EF-hand domains. Furthermore, MCC overexpression relocalizes DBC1 from the nucleus to the cytoplasm and reduces ß-cat K49 acetylation. Treatment of cells with the SIRT1 inhibitor Nicotinamide reverses MCC-induced deacetylation of ß-cat K49. These data suggest that the cytoplasmic MCC-DBC1 interaction sequesters DBC1 away from the nucleus, thereby removing a brake on DBC1 nuclear targets, such as SIRT1. This study provides new mechanistic insights into the DBC1-MCC axis as a new APC independent ß-cat inhibitory pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citoplasma/metabolismo , beta Catenina/genética , Acetilação , Transporte Ativo do Núcleo Celular , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Sequência de Aminoácidos , Sítios de Ligação , Núcleo Celular , Neoplasias Colorretais , Sequência Conservada , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Células HCT116 , Células HEK293 , Humanos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Ligação Proteica , Processamento de Proteína Pós-Traducional , Transcrição Gênica , Proteínas Supressoras de Tumor , beta Catenina/metabolismo
12.
J Cachexia Sarcopenia Muscle ; 15(1): 255-269, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38062876

RESUMO

BACKGROUND: Skeletal muscle atrophy, particularly ageing-related muscular atrophy such as sarcopenia, is a significant health concern. Despite its prevalence, the underlying mechanisms remain poorly understood, and specific approved medications are currently unavailable. Deleted in breast cancer 1 (DBC1) is a well-known regulator of senescence, metabolism or apoptosis. Recent reports suggest that DBC1 may also potentially regulate muscle function, as mice lacking DBC1 exhibit weakness and limpness. However, the function of DBC1 in skeletal muscle and its associated molecular mechanisms remain unknown, thus prompting the focus of this study. METHODS: Tibialis anterior (TA) muscle-specific DBC1 knockdown C57BL/6J male mice were generated through a single injection of 2.00 E + 11 vg of adeno-associated virus 9 delivering single-guide RNA for DBC1. Grip strength and endurance were assessed 2 months later, followed by skeletal muscle harvest. Muscle atrophy model was generated by cast immobilization of the mouse hindlimb for 2 weeks. Molecular markers of atrophy were probed in muscles upon termination. Cardiotoxin (CTX) was injected in TA muscles of DBC1 knockdown mice, and muscle regeneration was assessed by immunohistochemistry, quantitative PCR and western blotting. DBC1 knockdown C2C12 cells and myotubes were investigated using immunofluorescence staining, Seahorse, immunohistology, fluorescence-activated cell sorting and RNA-sequencing analyses. RESULTS: DBC1 knockdown in skeletal muscle of young mice led to signatures of muscle atrophy, including a 28% reduction in muscle grip force (P = 0.023), a 54.4% reduction in running distance (P = 0.002), a 14.3% reduction in muscle mass (P = 0.007) and significantly smaller myofibre cross-sectional areas (P < 0.0001). DBC1 levels decrease in age-related or limb immobilization-induced atrophic mouse muscles and overexpress DBC1-attenuated atrophic phenotypes in these mice. Muscle regeneration was hampered in mice with CTX-induced muscle injury by DBC1 knockdown, as evidenced by reductions in myofibre cross-sectional areas of regenerating myofibres with centralized nuclei (P < 0.0001), percentages of MyoG+ nuclei (P < 0.0001) and fusion index (P < 0.0001). DBC1 transcriptionally regulated mouse double minute 2 (MDM2), which mediated ubiquitination and degradation of forkhead box O3 (FOXO3). Increased FOXO3 proteins hampered myogenesis in DBC1 knockdown satellite cells by compromising around 50% of mitochondrial functions (P < 0.001) and exacerbated atrophy in DBC1 knockdown myofibres by activating the ubiquitin-proteasome and autophagy-lysosome pathways. CONCLUSIONS: DBC1 is essential in maintaining skeletal muscle integrity by protecting against myofibres wasting and enhancing muscle regeneration via FOXO3. This research highlights the significance of DBC1 for healthy skeletal muscle function and its connection to muscular atrophy.


Assuntos
Músculo Esquelético , RNA Guia de Sistemas CRISPR-Cas , Animais , Masculino , Camundongos , Caquexia/patologia , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , Músculo Esquelético/patologia , Atrofia Muscular/patologia
13.
Front Genet ; 15: 1363417, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38841722

RESUMO

Introduction: Obesity is a multifactorial disease associated with the development of many comorbidities. This disease is associated with several metabolic alterations; however, it has been shown that some individuals with obesity do not exhibit metabolic syndrome. Adipose tissue neutralizes the detrimental effects of circulating fatty acids, ectopic deposition, and inflammation, among others, through its esterification into neutral lipids that are stored in the adipocyte. However, when the adipocyte is overloaded, i.e., its expansion capacity is exceeded, this protection is lost, resulting in fatty acid toxicity with ectopic fat accumulation in peripheral tissues and inflammation. In this line, this study aimed to investigate whether polymorphisms in genes that control adipose tissue fat storage capacity are potential biomarkers for severe obesity susceptibility and also metabolic complications. Methods: This study enrolled 305 individuals with severe obesity (cases, BMI≥35 kg/m2) and 196 individuals with normal weight (controls, 18.5≤BMI≤24.9 kg/m2). Demographic, anthropometric, biochemical, and blood pressure variables were collected from the participants. Plasma levels of leptin, resistin, MCP1, and PAI1 were measured by Bio-Plex 200 Multiplexing Analyzer System. Genomic DNA was extracted and variants in DBC1 (rs17060940), SIRT1 (rs7895833 and rs1467568), UCP2 (rs660339), PPARG (rs1801282) and ADRB2 (rs1042713 and rs1042714) genes were genotyped by PCR allelic discrimination using TaqMan® assays. Results: Our findings indicated that SIRT1 rs7895833 polymorphism was a risk factor for severe obesity development in the overdominant model. SIRT1 rs1467568 and UCP2 rs660339 were associated with anthropometric traits. SIRT1 rs1467568 G allele was related to lower medians of body adipose index and hip circumference, while the UCP2 rs660339 AA genotype was associate with increased body mass index. Additionally, DBC1 rs17060940 influenced glycated hemoglobin. Regarding metabolic alterations, 27% of individuals with obesity presented balanced metabolic status in our cohort. Furthermore, SIRT1 rs1467568 AG genotype increased 2.5 times the risk of developing metabolic alterations. No statistically significant results were observed with Peroxisome Proliferator-Activated Receptor Gama and ADRB2 polymorphisms. Discussion/Conclusion: This study revealed that SIRT1 rs7895833 and rs1467568 are potential biomarkers for severe obesity susceptibility and the development of unbalanced metabolic status in obesity, respectively. UCP2 rs660339 and DBC1 rs17060940 also showed a significant role in obesity related-traits.

14.
Br J Pharmacol ; 181(17): 3039-3063, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38679474

RESUMO

BACKGROUND AND PURPOSE: Amyloid-ß (Aß) peptide is one of the more important pathological markers in Alzheimer's disease (AD). The development of AD impairs autophagy, which results in an imbalanced clearance of Aß. Our previous research demonstrated that AdipoRon, an agonist of adiponectin receptors, decreased the deposition of Aß and enhanced cognitive function in AD. However, the exact mechanisms by which AdipoRon affects Aß clearance remain unclear. EXPERIMENTAL APPROACH: We studied how AdipoRon affects autophagy in HT22 cells and APP/PS1 transgenic mice. We also investigated the signalling pathway involved and used pharmacological inhibitors to examine the role of autophagy in this process. KEY RESULTS: AdipoRon promotes Aß clearance by activating neuronal autophagy in the APP/PS1 transgenic mice. Interestingly, we found that AdipoRon induces the nuclear translocation of GAPDH, where it interacts with the SIRT1/DBC1 complex. This interaction then leads to the release of DBC1 and the activation of SIRT1, which in turn activates autophagy. Importantly, we found that inhibiting either GAPDH or SIRT1 to suppress the activity of SIRT1 counteracts the elevated autophagy and decreased Aß deposition caused by AdipoRon. This suggests that SIRT1 plays a critical role in the effect of AdipoRon on autophagic induction in AD. CONCLUSION AND IMPLICATIONS: AdipoRon promotes the clearance of Aß by enhancing autophagy through the AdipoR1/AMPK-dependent nuclear translocation of GAPDH and subsequent activation of SIRT1. This novel molecular pathway sheds light on the modulation of autophagy in AD and may lead to the development of new therapeutic strategies targeting this pathway.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Autofagia , Camundongos Transgênicos , Sirtuína 1 , Sirtuína 1/metabolismo , Sirtuína 1/antagonistas & inibidores , Doença de Alzheimer/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Autofagia/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Camundongos , Piperidinas/farmacologia , Humanos , Linhagem Celular , Camundongos Endogâmicos C57BL , Receptores de Adiponectina/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Masculino
15.
Protein Sci ; 33(4): e4938, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38533551

RESUMO

Regulation of SIRT1 activity is vital to energy homeostasis and plays important roles in many diseases. We previously showed that insulin triggers the epigenetic regulator DBC1 to prime SIRT1 for repression by the multifunctional trafficking protein PACS-2. Here, we show that liver DBC1/PACS-2 regulates the diurnal inhibition of SIRT1, which is critically important for insulin-dependent switch in fuel metabolism from fat to glucose oxidation. We present the x-ray structure of the DBC1 S1-like domain that binds SIRT1 and an NMR characterization of how the SIRT1 N-terminal region engages DBC1. This interaction is inhibited by acetylation of K112 of DBC1 and stimulated by the insulin-dependent phosphorylation of human SIRT1 at S162 and S172, catalyzed sequentially by CK2 and GSK3, resulting in the PACS-2-dependent inhibition of nuclear SIRT1 enzymatic activity and translocation of the deacetylase in the cytoplasm. Finally, we discuss how defects in the DBC1/PACS-2-controlled SIRT1 inhibitory pathway are associated with disease, including obesity and non-alcoholic fatty liver disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Sirtuína 1 , Humanos , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Processamento de Proteína Pós-Traducional , Insulina/metabolismo
16.
Aging (Albany NY) ; 15(17): 8812-8832, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37683133

RESUMO

Deleted in breast cancer 1 (DBC1) was initially identified from a homozygously deleted region in human chromosome 8p21. It has been well established that DBC1 plays a dual role during cancer development. Depending on the physiological context, it can promote or inhibit tumorigenesis. Whether it plays a role in lens pathogenesis remains elusive. In the present study, we demonstrated that DBC1 is highly expressed in lens epithelial cells from different vertebrates and in retina pigment epithelial cells as well. Moreover, DBC1 is SUMOylated through SUMO1 conjugation at K591 residue in human and mouse lens epithelial cells. The SUMOylated DBC1 is localized in the nucleus and plays an essential role in promoting stress-induced apoptosis. Silence of DBC1 attenuates oxidative stress-induced apoptosis. In contrast, overexpression of DBC1 enhances oxidative stress-induced apoptosis, and this process depends on p53. Mechanistically, DBC1 interacts with p53 to regulate its phosphorylation status at multiple sites and the SUMOylation of DBC1 enhances its interaction with p53. Together, our results identify that DBC1 is an important regulator mediating stress-induced apoptosis in lens, and thus participates in control of lens cataractogenesis.


Assuntos
Apoptose , Proteína Supressora de Tumor p53 , Animais , Humanos , Camundongos , Apoptose/genética , Carcinogênese , Transformação Celular Neoplásica , Células Epiteliais , Proteína SUMO-1/genética , Proteína Supressora de Tumor p53/genética
17.
Cells ; 12(1)2022 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-36611939

RESUMO

DNA damage is the major cause of senescence and apoptosis; however, the manner by which DNA-damaged cells become senescent remains unclear. We demonstrate that DNA damage leads to a greater level of senescence rather than apoptosis in DBC1-deficient cells. In addition, we show that BLM becomes degraded during DNA damage, which induces p21 expression and senescence. DBC1 binds to and shields BLM from degradation, thus suppressing senescence. ML216 promotes DBC1-BLM interaction, which aids in the preservation of BLM following DNA damage and suppresses senescence. ML216 enhances pulmonary function by lowering the levels of senescence and fibrosis in both aged mice and a mouse model of bleomycin-induced idiopathic pulmonary fibrosis. Our data reveal a unique mechanism preventing DNA-damaged cells from becoming senescent, which may be regulated by the use of ML216 as a potential treatment for senescence-related diseases.


Assuntos
Bleomicina , Fibrose Pulmonar Idiopática , Animais , Camundongos , Bleomicina/farmacologia , DNA , Dano ao DNA , DNA Helicases , Fibrose Pulmonar Idiopática/genética , Pulmão
18.
Elife ; 112022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35913115

RESUMO

DBC1 has been characterized as a key regulator of physiological and pathophysiological activities, such as DNA damage, senescence, and tumorigenesis. However, the mechanism by which the functional stability of DBC1 is regulated has yet to be elucidated. Here, we report that the ubiquitination-mediated degradation of DBC1 is regulated by the E3 ubiquitin ligase SIAH2 and deubiquitinase OTUD5 under hypoxic stress. Mechanistically, hypoxia promoted DBC1 to interact with SIAH2 but not OTUD5, resulting in the ubiquitination and subsequent degradation of DBC1 through the ubiquitin-proteasome pathway. SIAH2 knockout inhibited tumor cell proliferation and migration, which could be rescued by double knockout of SIAH2/CCAR2. Human tissue microarray analysis further revealed that the SIAH2/DBC1 axis was responsible for tumor progression under hypoxic stress. These findings define a key role of the hypoxia-mediated SIAH2-DBC1 pathway in the progression of human breast cancer and provide novel insights into the metastatic mechanism of breast cancer.


Assuntos
Neoplasias da Mama , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Hipóxia/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
19.
Mol Cell Biol ; 42(10): e0015122, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36036574

RESUMO

Although ELL-associated factors 1 and 2 (EAF1/2) have been shown to enhance RNA polymerase II-mediated transcription in vitro, their functional roles in vivo are poorly known. In this report, we show functions of these proteins in regulating ELL stability through their competitive binding with HDAC3 at the N terminus of ELL. Reduced HDAC3 binding to ELL causes increased acetylation leading to reduced ubiquitylation-mediated degradation. Similar functional roles played by DBC1 in regulating ELL stability further prompted in-depth analyses that demonstrated presence of negative feedback loop mechanisms between DBC1 and EAF1/2 in maintaining overall ELL level. Mechanistically, increased DBC1 reduces EAF1/2 level through increased ubiquitylation involving E3 ubiquitin ligase TRIM28, whereas increased EAF1/2 reduces DBC1 level through reduced transcription. Physiologically, after a few passages, ELL levels in either DBC1 or EAF1 knockdown cells are restored through enhanced expression of EAF1 and DBC1, respectively. Interestingly, for maintenance of ELL level, mammalian cells prefer the EAF1-dependent pathway during exposure to genotoxic stress, and the DBC1-dependent pathway during exposure to growth factors. Thus, we describe coordinated functions of multiple factors, including EAF1/2, HDAC3, DBC1, and TRIM28 in regulating ELL protein level for optimal target gene expression in a context-dependent manner within mammalian cells.


Assuntos
RNA Polimerase II , Fatores de Elongação da Transcrição , Animais , Fatores de Elongação da Transcrição/metabolismo , Retroalimentação , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/genética , Mamíferos/metabolismo
20.
J Leukoc Biol ; 109(2): 449-454, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32337788

RESUMO

DBC1 (deleted in breast cancer 1) is a human nuclear protein that modulates the activities of various proteins. Most of the research on DBC1 has focused on metabolism and epigenetics because it is a crucial endogenic inhibitor of deacetylase Sirtuin1 (SIRT1). In this review, we have discussed and summarized the new advances in DBC1 research, mostly focusing on its structure, regulatory function, and significance in cancer and autoimmune diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doenças Autoimunes/metabolismo , Neoplasias/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Animais , Apoptose , Humanos , Linfócitos/metabolismo , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA