Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.634
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Mol Biol Cell ; 35(2): ar21, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38088875

RESUMO

In vertebrates, two distinct condensin complexes, condensin I and condensin II, cooperate to drive mitotic chromosome assembly. It remains largely unknown how the two complexes differentially contribute to this process at a mechanistic level. We have previously dissected the role of individual subunits of condensin II by introducing recombinant complexes into Xenopus egg extracts. Here we extend these efforts by introducing a modified functional assay using extracts depleted of topoisomerase IIα (topo IIα), which allows us to further elucidate the functional similarities and differences between condensin I and condensin II. The intrinsically disordered C-terminal region of the CAP-D3 subunit (the D3 C-tail) is a major target of Cdk1 phosphorylation, and phosphorylation-deficient mutations in this region impair condensin II functions. We also identify a unique helical structure in CAP-D3 (the D3 HEAT docker) that is predicted to directly interact with CAP-G2. Deletion of the D3 HEAT docker, along with the D3 C-tail, enhances the ability of condensin II to assemble mitotic chromosomes. Taken together, we propose a self-suppression mechanism unique to condensin II that is released by mitotic phosphorylation. Evolutionary implications of our findings are also discussed.


Assuntos
Cromossomos , Proteínas de Ligação a DNA , Animais , Proteínas de Ligação a DNA/fisiologia , Complexos Multiproteicos/genética , Adenosina Trifosfatases/genética , Mitose
2.
Mol Cancer ; 21(1): 89, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35354467

RESUMO

BACKGROUND: Frequent truncation mutations of the histone lysine N-methyltransferase KMT2C have been detected by whole exome sequencing studies in various cancers, including malignancies of the prostate. However, the biological consequences of these alterations in prostate cancer have not yet been elucidated. METHODS: To investigate the functional effects of these mutations, we deleted the C-terminal catalytic core motif of Kmt2c specifically in mouse prostate epithelium. We analysed the effect of Kmt2c SET domain deletion in a Pten-deficient PCa mouse model in vivo and of truncation mutations of KMT2C in a large number of prostate cancer patients. RESULTS: We show here for the first time that impaired KMT2C methyltransferase activity drives proliferation and PIN formation and, when combined with loss of the tumour suppressor PTEN, triggers loss of senescence, metastatic dissemination and dramatically reduces life expectancy. In Kmt2c-mutated tumours we show enrichment of proliferative MYC gene signatures and loss of expression of the cell cycle repressor p16INK4A. In addition, we observe a striking reduction in disease-free survival of patients with KMT2C-mutated prostate cancer. CONCLUSIONS: We identified truncating events of KMT2C as drivers of proliferation and PIN formation. Loss of PTEN and KMT2C in prostate cancer results in loss of senescence, metastatic dissemination and reduced life expectancy. Our data demonstrate the prognostic significance of KMT2C mutation status in prostate cancer patients. Inhibition of the MYC signalling axis may be a viable treatment option for patients with KMT2C truncations and therefore poor prognosis.


Assuntos
Metiltransferases , Neoplasias da Próstata , Animais , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/fisiologia , Humanos , Masculino , Metiltransferases/genética , Camundongos , Mutação , Neoplasias da Próstata/metabolismo , Sequenciamento do Exoma
3.
FASEB J ; 36(4): e22231, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35230719

RESUMO

The dysfunction of osteogenesis is a key character in the pathogenesis of osteoporosis, but the network of signaling mechanisms in controlling the differentiation of osteoblast remain unclear. Thrap3 has been proved participating in various biological process, especially in the differentiation of stem cells. Here, we demonstrate that Thrap3 could promote osteogenesis through the inhibition of the degradation of Runx2, which is a key molecular structure in early osteoblast differentiation. Furthermore, we found that the osteogenesis enhancing capacity of Thrap3 was caused by physically binding with Sox9, inhibiting the transcriptional activity of Sox9, and then decreasing the decomposition-promoted effect of Sox9 on Runx2. Our data shows that Thrap3 promotes osteoblast differentiation through the Thrap3-Sox9-Runx2 axis. What we found may help for further clarifying the molecular mechanism of osteogenic differentiation and give a new potential therapeutic target for osteoporosis.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Proteínas de Ligação a DNA/fisiologia , Osteogênese/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/antagonistas & inibidores , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Osteoblastos/citologia , Fatores de Transcrição SOX9/fisiologia
4.
Sci Rep ; 12(1): 2306, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35145145

RESUMO

Myocardin-related transcription factor A (MRTF-A), a coactivator of serum response factor (SRF), regulates the expression of many cytoskeletal genes in response to cytoplasmic and nuclear actin dynamics. Here we describe a novel mechanism to regulate MRTF-A activity within the nucleus by showing that lamina-associated polypeptide 2α (Lap2α), the nucleoplasmic isoform of Lap2, is a direct binding partner of MRTF-A, and required for the efficient expression of MRTF-A/SRF target genes. Mechanistically, Lap2α is not required for MRTF-A nuclear localization, unlike most other MRTF-A regulators, but is required for efficient recruitment of MRTF-A to its target genes. This regulatory step takes place prior to MRTF-A chromatin binding, because Lap2α neither interacts with, nor specifically influences active histone marks on MRTF-A/SRF target genes. Phenotypically, Lap2α is required for serum-induced cell migration, and deregulated MRTF-A activity may also contribute to muscle and proliferation phenotypes associated with loss of Lap2α. Our studies therefore add another regulatory layer to the control of MRTF-A-SRF-mediated gene expression, and broaden the role of Lap2α in transcriptional regulation.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica/genética , Proteínas de Membrana/fisiologia , Transativadores/genética , Transativadores/metabolismo , Actinas/metabolismo , Animais , Movimento Celular/genética , Cromatina , Citoplasma/metabolismo , Citoesqueleto/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Células NIH 3T3 , Ligação Proteica/genética , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transativadores/fisiologia , Transcrição Gênica/genética
5.
Cell Mol Life Sci ; 79(2): 112, 2022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-35099607

RESUMO

T cell activation initiates protective adaptive immunity, but counterbalancing mechanisms are critical to prevent overshooting responses and to maintain immune homeostasis. The CARD11-BCL10-MALT1 (CBM) complex bridges T cell receptor engagement to NF-κB signaling and MALT1 protease activation. Here, we show that ABIN-1 is modulating the suppressive function of A20 in T cells. Using quantitative mass spectrometry, we identified ABIN-1 as an interactor of the CBM signalosome in activated T cells. A20 and ABIN-1 counteract inducible activation of human primary CD4 and Jurkat T cells. While A20 overexpression is able to silence CBM complex-triggered NF-κB and MALT1 protease activation independent of ABIN-1, the negative regulatory function of ABIN-1 depends on A20. The suppressive function of A20 in T cells relies on ubiquitin binding through the C-terminal zinc finger (ZnF)4/7 motifs, but does not involve the deubiquitinating activity of the OTU domain. Our mechanistic studies reveal that the A20/ABIN-1 module is recruited to the CBM complex via A20 ZnF4/7 and that proteasomal degradation of A20 and ABIN-1 releases the CBM complex from the negative impact of both regulators. Ubiquitin binding to A20 ZnF4/7 promotes destructive K48-polyubiquitination to itself and to ABIN-1. Further, after prolonged T cell stimulation, ABIN-1 antagonizes MALT1-catalyzed cleavage of re-synthesized A20 and thereby diminishes sustained CBM complex signaling. Taken together, interdependent post-translational mechanisms are tightly controlling expression and activity of the A20/ABIN-1 silencing module and the cooperative action of both negative regulators is critical to balance CBM complex signaling and T cell activation.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Linfócitos T/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/fisiologia , Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Células Cultivadas , Guanilato Ciclase/metabolismo , Células HEK293 , Humanos , Células Jurkat , Ativação Linfocitária/genética , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Complexos Multiproteicos/metabolismo , NF-kappa B/metabolismo , Ligação Proteica , Interferência de RNA/imunologia , Transdução de Sinais/fisiologia , Linfócitos T/imunologia
6.
PLoS Genet ; 18(1): e1009928, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35100262

RESUMO

Intermediate neural progenitors (INPs) boost the number and diversity of neurons generated from neural stem cells (NSCs) by undergoing transient proliferation. In the developing Drosophila brains, INPs are generated from type II neuroblasts (NBs). In order to maintain type II NB identity and their capability to produce INPs, the proneural protein Asense (Ase) needs to be silenced by the Ets transcription factor pointed P1 (PntP1), a master regulator of type II NB development. However, the molecular mechanisms underlying the PntP1-mediated suppression of Ase is still unclear. In this study, we utilized genetic and molecular approaches to determine the transcriptional property of PntP1 and identify the direct downstream effector of PntP1 and the cis-DNA elements that mediate the suppression of ase. Our results demonstrate that PntP1 directly activates the expression of the transcriptional repressor, Tailless (Tll), by binding to seven Ets-binding sites, and Tll in turn suppresses the expression of Ase in type II NBs by binding to two hexameric core half-site motifs. We further show that Tll provides positive feedback to maintain the expression of PntP1 and the identity of type II NBs. Thus, our study identifies a novel direct target of PntP1 and reveals mechanistic details of the specification and maintenance of the type II NB identity by PntP1.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/genética , Fatores de Transcrição/fisiologia , Animais , Sítios de Ligação , Proteínas de Ligação a DNA/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Elementos Facilitadores Genéticos , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Transgenes
7.
Pathol Res Pract ; 229: 153734, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35030351

RESUMO

Clear cell renal cell carcinoma (ccRCC) is recognized as one of the most lethal malignancies among the urological system, with constantly increasing mortality. While the molecular mechanisms underlying ccRCC progression are still poorly understood, the molecular and functional role of lncRNA in multiple diseases has been well demonstrated. In this study, we hypothesized that lncRNA MEG8 might participate in ccRCC development. At first, we found that MEG8 expression was increased in ccRCC tumor tissues and cells. Next, we demonstrated that MEG8 knockdown suppressed cell viability, migration, and invasion in vitro and inhibited tumor growth in vivo. Subsequently, we utilized bioinformatics analysis, ChIP, and luciferase assays, and we found that PLAG1 could transcriptionally regulate MEG8 in ccRCC cells. Furthermore, MEG8 promoted G3BP1 expression to aggravate ccRCC tumorigenic properties through sponging miR-495-3p. Our study identified a novel PLAG1/MEG8/miR-495-3p/G3BP1 network in ccRCC development, which might be a promising direction for developing new diagnoses or therapeutic agents for ccRCC.


Assuntos
Carcinoma de Células Renais/genética , DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Neoplasias Renais/genética , MicroRNAs/fisiologia , Proteínas de Ligação a Poli-ADP-Ribose/fisiologia , RNA Helicases/fisiologia , Proteínas com Motivo de Reconhecimento de RNA/fisiologia , RNA Longo não Codificante , Humanos , Células Tumorais Cultivadas
8.
Cell Rep ; 38(2): 110211, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021076

RESUMO

Oncogene-induced senescence (OIS) is a form of stable cell-cycle arrest arising in response to oncogenic stimulation. OIS must be bypassed for transformation, but the mechanisms of OIS establishment and bypass remain poorly understood, especially at the post-transcriptional level. Here, we show that the RNA-binding protein UNR/CSDE1 enables OIS in primary mouse keratinocytes. Depletion of CSDE1 leads to senescence bypass, cell immortalization, and tumor formation, indicating that CSDE1 behaves as a tumor suppressor. Unbiased high-throughput analyses uncovered that CSDE1 promotes OIS by two independent molecular mechanisms: enhancement of the stability of senescence-associated secretory phenotype (SASP) factor mRNAs and repression of Ybx1 mRNA translation. Importantly, depletion of YBX1 from immortal keratinocytes rescues senescence and uncouples proliferation arrest from the SASP, revealing multilayered mechanisms exerted by CSDE1 to coordinate senescence. Our data highlight the relevance of post-transcriptional control in the regulation of senescence.


Assuntos
Senescência Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular , Proliferação de Células/fisiologia , Senescência Celular/genética , Proteínas de Ligação a DNA/fisiologia , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Oncogenes/genética , Cultura Primária de Células , Processamento Pós-Transcricional do RNA/fisiologia , Proteínas de Ligação a RNA/fisiologia , Fenótipo Secretor Associado à Senescência/genética , Fenótipo Secretor Associado à Senescência/fisiologia , Transdução de Sinais/fisiologia , Proteína 1 de Ligação a Y-Box/metabolismo
9.
Clin Transl Oncol ; 24(1): 84-92, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34181232

RESUMO

PURPOSE: To investigate the effect of microRNA-543 (miR-543) on the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of triple-negative breast cancer (TNBC) cells, and the associated mechanism. METHODS: Human breast cancer cells (MDA-MB-231, HCC1937, and MCF-7, ZR-75-1) and normal human breast epithelial cell line (MCF10A) were transfected with miR-543 mimics or inhibitor using lipofectamine 2000. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting were used to determine the mRNA and protein expression levels of miR-543, actin-like protein 6A (ACTL6A), vimentin, Snail, and E-cadherin in breast cancer cells/tissue. Cell counting kit-8 (CCK-8), wound-healing, and Transwell assays were used to measure the effect of miR-543 on TNBC cell proliferation, invasion, and migration. Overall survival was determined using data from Gene Expression Omnibus (GEO) and Cancer Genome Atlas (TCGA) databases. Bioinformatics analysis and luciferase reporter gene assay were used to determine the regulatory effect of miR-543 on ACTL6A. RESULTS: The level of expression of miR-543 was significantly lower in breast cancer cells/tissue than in normal human breast epithelial cell/tissue (p < 0.05). MicroRNA-543 expression level was significantly reduced in TNBC cells/tissue, relative to the other breast cancer cells/normal breast tissue (p < 0.05). MicroRNA-543 significantly suppressed tumor growth and the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of TNBC cells, in mouse xenograft model (p < 0.05). CONCLUSIONS: miR-543 influences the biological behavior of TNBC cells by directly targeting ACTL6A gene. miR-543 could serve as a novel diagnostic and therapeutic target for TNBC.


Assuntos
Actinas/fisiologia , Movimento Celular , Proliferação de Células , Proteínas Cromossômicas não Histona/fisiologia , Proteínas de Ligação a DNA/fisiologia , Regulação para Baixo , Transição Epitelial-Mesenquimal , MicroRNAs/fisiologia , Neoplasias de Mama Triplo Negativas/patologia , Animais , Humanos , Camundongos , Invasividade Neoplásica , Células Tumorais Cultivadas
10.
Gut ; 71(3): 467-478, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33785559

RESUMO

BACKGROUND: Gastric adenocarcinoma (GAC) is a lethal disease with limited therapeutic options. Genetic alterations in chromatin remodelling gene AT-rich interactive domain 1A (ARID1A) and mTOR pathway activation occur frequently in GAC. Targeting the mechanistic target of rapamycin (mTOR) pathway in unselected patients has failed to show survival benefit. A deeper understanding of GAC might identify a subset that can benefit from mTOR inhibition. METHODS: Genomic alterations in ARID1A were analysed in GAC. Mouse gastric epithelial cells from CK19-Cre-Arid1Afl/fl and wild-type mice were used to determine the activation of oncogenic genes due to loss of Arid1A. Functional studies were performed to determine the significance of loss of ARID1A and the sensitivity of ARID1A-deficient cancer cells to mTOR inhibition in GAC. RESULTS: More than 30% of GAC cases had alterations (mutations or deletions) of ARID1A and ARID1A expression was negatively associated with phosphorylation of S6 and SOX9 in GAC tissues and patient-derived xenografts (PDXs). Activation of mTOR signalling (increased pS6) and SOX9 nuclear expression were strongly increased in Arid1A-/- mouse gastric tissues which could be curtailed by RAD001, an mTOR inhibitor. Knockdown of ARID1A in GAC cell lines increased pS6 and nuclear SOX9 and increased sensitivity to an mTOR inhibitor which was further amplified by its combination with fluorouracil both in vitro and in vivo in PDXs. CONCLUSIONS: The loss of ARID1A activates pS6 and SOX9 in GAC, which can be effectively targeted by an mTOR inhibitor. Therefore, our studies suggest a new therapeutic strategy of clinically targeting the mTOR pathway in patients with GAC with ARID1A deficiency.


Assuntos
Adenocarcinoma/etiologia , Proteínas de Ligação a DNA/fisiologia , Fatores de Transcrição SOX9/fisiologia , Transdução de Sinais/fisiologia , Neoplasias Gástricas/etiologia , Serina-Treonina Quinases TOR/fisiologia , Fatores de Transcrição/fisiologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células , Camundongos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
11.
Cell Rep ; 37(11): 110124, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34910919

RESUMO

Regulatory T (Treg) cells play crucial roles in suppressing deleterious immune response. Here, we investigate how Treg cells are mechanistically induced in vitro (iTreg) and stabilized via transcriptional regulation of Treg lineage-specifying factor Foxp3. We find that acetylation of histone tails at the Foxp3 promoter is required for inducing Foxp3 transcription. Upon induction, histone acetylation signals via bromodomain-containing proteins, particularly targets of inhibitor JQ1, and sustains Foxp3 transcription via a global or trans effect. Subsequently, Tet-mediated DNA demethylation of Foxp3 cis-regulatory elements, mainly enhancer CNS2, increases chromatin accessibility and protein binding, stabilizing Foxp3 transcription and obviating the need for the histone acetylation signal. These processes transform stochastic iTreg induction into a stable cell fate, with the former sensitive and the latter resistant to genetic and environmental perturbations. Thus, sequential histone acetylation and DNA demethylation in Foxp3 induction and maintenance reflect stepwise mechanical switches governing iTreg cell lineage specification.


Assuntos
Desmetilação do DNA , Proteínas de Ligação a DNA/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Histonas/química , Proteínas Proto-Oncogênicas/fisiologia , Linfócitos T Reguladores/imunologia , Acetilação , Animais , Diferenciação Celular , Metilação de DNA , Feminino , Fatores de Transcrição Forkhead/genética , Histonas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico
12.
Biomed Res Int ; 2021: 5282894, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34778452

RESUMO

This study is aimed at exploring the potential role of GSDMC in kidney renal clear cell carcinoma (KIRC). We analyzed the expression of GSDMC in 33 types of cancers in TCGA database. The results showed that the expression of GSDMC was upregulated in most cancers. We found a significant association between high expression of GSDMC and shortened patient overall survival, progression-free survival, and disease-specific survival. In vitro experiments have shown that the expression of GSDMC was significantly elevated in KIRC cell lines. Moreover, decreased expression of GSDMC was significantly associated with decreased cell proliferation. In summary, we believe that this study provides valuable data supporting future clinical treatment.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Proteínas de Ligação a DNA/genética , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/fisiologia , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , China , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Bases de Dados Genéticas , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Rim/patologia , Neoplasias Renais/genética , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Transdução de Sinais/genética , Ativação Transcricional/genética , Transcriptoma/genética
13.
Cell Rep ; 37(8): 110036, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818550

RESUMO

Balance between the hematopoietic stem cell (HSC) duality to either possess self-renewal capacity or differentiate into multipotency progenitors (MPPs) is crucial for maintaining homeostasis of the hematopoietic stem/progenitor cell (HSPC) compartment. To retain the HSC self-renewal activity, KIT, a receptor tyrosine kinase, in HSCs is activated by its cognate ligand KITLG originating from niche cells. Here, we show that AT-rich interaction domain 4B (ARID4B) interferes with KITLG/KIT signaling, consequently allowing HSC differentiation. Conditional Arid4b knockout in mouse hematopoietic cells blocks fetal HSC differentiation, preventing hematopoiesis. Mechanistically, ARID4B-deficient HSCs self-express KITLG and overexpress KIT. As to downstream pathways of KITLG/KIT signaling, inhibition of Src family kinases rescues the HSC differentiation defect elicited by ARID4B loss. In summary, the intrinsic ARID4B-KITLG/KIT-Src axis is an HSPC regulatory program that enables the differentiation state, while KIT stimulation by KITLG from niche cells preserves the HSPC undifferentiated pool.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Comunicação Autócrina , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Autorrenovação Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Feminino , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais/fisiologia , Fator de Células-Tronco/metabolismo , Nicho de Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Quinases da Família src/metabolismo
14.
Cell Rep ; 37(8): 110034, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34818558

RESUMO

Endogenous metabolites, environmental agents, and therapeutic drugs promote formation of covalent DNA-protein crosslinks (DPCs). Persistent DPCs compromise genome integrity and are eliminated by multiple repair pathways. Aberrant Top1-DNA crosslinks, or Top1ccs, are processed by Tdp1 and Wss1 functioning in parallel pathways in Saccharomyces cerevisiae. It remains obscure how cells choose between diverse mechanisms of DPC repair. Here, we show that several SUMO biogenesis factors (Ulp1, Siz2, Slx5, and Slx8) control repair of Top1cc or an analogous DPC lesion. Genetic analysis reveals that SUMO promotes Top1cc processing in the absence of Tdp1 but has an inhibitory role if cells additionally lack Wss1. In the tdp1Δ wss1Δ mutant, the E3 SUMO ligase Siz2 stimulates sumoylation in the vicinity of the DPC, but not SUMO conjugation to Top1. This Siz2-dependent sumoylation inhibits alternative DPC repair mechanisms, including Ddi1. Our findings suggest that SUMO tunes available repair pathways to facilitate faithful DPC repair.


Assuntos
Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Cisteína Endopeptidases/metabolismo , DNA/metabolismo , Reparo do DNA/genética , DNA Topoisomerases Tipo I/metabolismo , Proteínas de Ligação a DNA/genética , Diester Fosfórico Hidrolases/metabolismo , Proteína SUMO-1/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação/genética , Sumoilação/fisiologia , Ubiquitina-Proteína Ligases/metabolismo
15.
Biochemistry ; 60(45): 3385-3397, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34723504

RESUMO

Condensins play a key role in higher order chromosome organization. In budding yeast Saccharomyces cerevisiae, a condensin complex consists of five subunits: two conserved structural maintenance of chromosome subunits, Smc2 and Smc4, a kleisin Brn1, and two HEAT repeat subunits, Ycg1, which possesses a DNA binding activity, and Ycs4, which can transiently associate with Smc4 and thereby disrupt its association with the Smc2 head. We characterized here DNA binding activity of the non-SMC subunits using an agnostic, model-independent approach. To this end, we mapped the DNA interface of the complex using sulfo-NHS biotin labeling. Besides the known site on Ycg1, we found a patch of lysines at the C-terminal domain of Ycs4 that were protected from biotinylation in the presence of DNA. Point mutations at the predicted protein-DNA interface reduced both Ycs4 binding to DNA and the DNA stimulated ATPase activity of the reconstituted condensin, whereas overproduction of the mutant Ycs4 was detrimental for yeast viability. Notably, the DNA binding site on Ycs4 partially overlapped with its interface with SMC4, revealing an intricate interplay between DNA binding, engagement of the Smc2-Smc4 heads, and ATP hydrolysis and suggesting a mechanism for ATP-modulated loading and translocation of condensins on DNA.


Assuntos
Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/fisiologia , Proteínas de Ligação a DNA/metabolismo , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/fisiologia , Adenosina Trifosfatases/genética , Sítios de Ligação/genética , Biotinilação , Comunicação Celular , Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona , DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Complexos Multiproteicos/genética , Proteínas Nucleares , Fagocitose , Mutação Puntual/genética , Domínios Proteicos/fisiologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
J BUON ; 26(5): 1925-1930, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34761601

RESUMO

PURPOSE: RNF6 is verified to promote the malignant growth of colorectal cancer (CRC) and its level is linked to prognosis in CRC patients. Radioresistance is a key factor influencing prognosis in CRC. This study aimed to uncover the potential regulation of ring finger protein 6 (RNF6) in CRC radioresistance. METHODS: RNF6 levels in radioresistant and non-radioresistant CRC patients were detected. In vitro and in vivo regulatory effects of RNF6 on radioresistant CRC cell lines and nude mice bearing radioresistant CRC were examined, respectively. The involvement of Wnt pathway in CRC radioresistance was explored by Western blot. RESULTS: RNF6 was highly expressed in radioresistant CRC species than that of non-radioresistant ones. Identically, RNF6 was upregulated in radioresistant CRC cells compared to parental cells. SW1116 cells overexpressing RNF6 were more tolerant to radiotherapy, and similar results were obtained in nude mice bearing radioresistant CRC with overexpression of RNF6. Moreover, the Wnt pathway was activated during RNF6-induced radioresistance improvement in CRC. CONCLUSIONS: RNF6 enhances radioresistance of CRC through activating the Wnt pathway.


Assuntos
Neoplasias Colorretais/radioterapia , Proteínas de Ligação a DNA/fisiologia , Tolerância a Radiação/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus
17.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34702735

RESUMO

Cell competition is a homeostatic process that eliminates by apoptosis unfit or undesirable cells from animal tissues, including tumor cells that appear during the life of the organism. In Drosophila there is evidence that many types of oncogenic cells are eliminated by cell competition. One exception is cells mutant for polyhomeotic (ph), a member of the Polycomb family of genes; most of the isolated mutant ph clones survive and develop tumorous overgrowths in imaginal discs. To characterize the tumorigenic effect of the lack of ph, we first studied the growth of different regions of the wing disc deficient in ph activity and found that the effect is restricted to the proximal appendage. Moreover, we found that ph-deficient tissue is partially refractory to apoptosis. Second, we analyzed the behavior of clones lacking ph function and found that many suffer cell competition but are not completely eliminated. Unexpectedly, we found that nonmutant cells also undergo cell competition when surrounded by ph-deficient cells, indicating that within the same tissue cell competition may operate in opposite directions. We suggest two reasons for the incompleteness of cell competition in ph mutant cells: 1) These cells are partially refractory to apoptosis, and 2) the loss of ph function alters the identity of imaginal cells and subsequently their cell affinities. It compromises the winner/loser interaction, a prerequisite for cell competition.


Assuntos
Carcinogênese , Competição entre as Células , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/fisiologia , Discos Imaginais/crescimento & desenvolvimento , Complexo Repressor Polycomb 1/fisiologia , Animais , Apoptose , Drosophila , Sistema de Sinalização das MAP Quinases
18.
J Clin Invest ; 131(22)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34609966

RESUMO

Ferroptosis, an iron-dependent nonapoptotic cell death, is a highly regulated tumor suppressing process. However, functions and mechanisms of RNA-binding proteins in regulation of evasion of ferroptosis during lung cancer progression are still largely unknown. Here, we report that the RNA-binding protein RBMS1 participates in lung cancer development via mediating ferroptosis evasion. Through an shRNA-mediated systematic screen, we discovered that RBMS1 is a key ferroptosis regulator. Clinically, RBMS1 was elevated in lung cancer and its high expression was associated with reduced patient survival. Conversely, depletion of RBMS1 inhibited lung cancer progression both in vivo and in vitro. Mechanistically, RBMS1 interacted with the translation initiation factor eIF3d directly to bridge the 3'- and 5'-UTR of SLC7A11. RBMS1 ablation inhibited the translation of SLC7A11, reduced SLC7A11-mediated cystine uptake, and promoted ferroptosis. In a drug screen that targeted RBMS1, we further uncovered that nortriptyline hydrochloride decreased the level of RBMS1, thereby promoting ferroptosis. Importantly, RBMS1 depletion or inhibition by nortriptyline hydrochloride sensitized radioresistant lung cancer cells to radiotherapy. Our findings established RBMS1 as a translational regulator of ferroptosis and a prognostic factor with therapeutic potential and clinical value.


Assuntos
Sistema y+ de Transporte de Aminoácidos/genética , Proteínas de Ligação a DNA/fisiologia , Neoplasias Pulmonares/patologia , Biossíntese de Proteínas , Proteínas de Ligação a RNA/fisiologia , Animais , Linhagem Celular Tumoral , Ferroptose , Células HEK293 , Humanos , Neoplasias Pulmonares/radioterapia , Camundongos , Proteínas Proto-Oncogênicas c-ets/fisiologia , Tolerância a Radiação , Fatores de Transcrição/fisiologia
19.
Biochemistry ; 60(43): 3236-3252, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34665609

RESUMO

The summarized amalgam of internal relaxation modulations and external forces like pH, temperature, and solvent conditions determine the protein structure, stability, and function. In a free-energy landscape, although conformers are arranged in vertical hierarchy, there exist several adjacent parallel sets with conformers occupying equivalent energy cleft. Such conformational states are pre-requisites for the functioning of proteins that have oscillating environmental conditions. As these conformational changes have utterly small re-arrangements, nuclear magnetic resonance (NMR) spectroscopy is unique in elucidating the structure-dynamics-stability-function relationships for such conformations. Helicobacter pylori survives and causes gastric cancer at extremely low pH also. However, least is known as to how the genome of the pathogen is protected from reactive oxygen species (ROS) scavenging in the gut at low pH under acidic stress. In the current study, biophysical characteristics of H. pylori DNA binding protein (Hup) have been elucidated at pH 2 using a combination of circular dichroism, fluorescence, NMR spectroscopy, and molecular dynamics simulations. Interestingly, the protein was found to have conserved structural features, differential backbone dynamics, enhanced stability, and DNA binding ability at low pH as well. In summary, the study suggests the partaking of Hup protein even at low pH in DNA protection for maintaining the genome integrity.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Helicobacter pylori/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Proteínas de Transporte/metabolismo , Dicroísmo Circular/métodos , DNA/química , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Entropia , Fluorescência , Helicobacter pylori/patogenicidade , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética/métodos , Conformação Molecular , Simulação de Dinâmica Molecular , Espécies Reativas de Oxigênio/metabolismo , Solventes/química , Temperatura
20.
Viruses ; 13(10)2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34696437

RESUMO

The 2016 Zika virus (ZIKV) epidemic illustrates the impact of flaviviruses as emerging human pathogens. For unknown reasons, ZIKV replicates more efficiently in neural progenitor cells (NPCs) than in postmitotic neurons. Here, we identified host factors used by ZIKV using the NCI-60 library of cell lines and COMPARE analysis, and cross-analyzed this library with two other libraries of host factors with importance for ZIKV infection. We identified BAF45b, a subunit of the BAF (Brg1/Brm-associated factors) protein complexes that regulate differentiation of NPCs to post-mitotic neurons. ZIKV (and other flaviviruses) infected HAP1 cells deficient in expression of BAF45b and other BAF subunits less efficiently than wildtype (WT) HAP1 cells. We concluded that subunits of the BAF complex are important for infection of ZIKV and other flavivirus. Given their function in cell and tissue differentiation, such regulators may be important determinants of tropism and pathogenesis of arthropod-borne flaviviruses.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Infecção por Zika virus/metabolismo , Zika virus/metabolismo , Aedes/virologia , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Flavivirus , Haploidia , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Replicação Viral/fisiologia , Zika virus/patogenicidade , Infecção por Zika virus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA