Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.387
Filtrar
1.
Nature ; 620(7976): 1047-1053, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37459895

RESUMO

Zygotic genome activation (ZGA) activates the quiescent genome to enable the maternal-to-zygotic transition1,2. However, the identity of transcription factors that underlie mammalian ZGA in vivo remains elusive. Here we show that OBOX, a PRD-like homeobox domain transcription factor family (OBOX1-OBOX8)3-5, are key regulators of mouse ZGA. Mice deficient for maternally transcribed Obox1/2/5/7 and zygotically expressed Obox3/4 had a two-cell to four-cell arrest, accompanied by impaired ZGA. The Obox knockout defects could be rescued by restoring either maternal and zygotic OBOX, which suggests that maternal and zygotic OBOX redundantly support embryonic development. Chromatin-binding analysis showed that Obox knockout preferentially affected OBOX-binding targets. Mechanistically, OBOX facilitated the 'preconfiguration' of RNA polymerase II, as the polymerase relocated from the initial one-cell binding targets to ZGA gene promoters and distal enhancers. Impaired polymerase II preconfiguration in Obox mutants was accompanied by defective ZGA and chromatin accessibility transition, as well as aberrant activation of one-cell polymerase II targets. Finally, ectopic expression of OBOX activated ZGA genes and MERVL repeats in mouse embryonic stem cells. These data thus demonstrate that OBOX regulates mouse ZGA and early embryogenesis.


Assuntos
Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Proteínas de Homeodomínio , Fatores de Transcrição , Zigoto , Animais , Camundongos , Cromatina/genética , Cromatina/metabolismo , Desenvolvimento Embrionário/genética , Genoma/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Zigoto/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , RNA Polimerase II/metabolismo , Regiões Promotoras Genéticas/genética , Elementos Facilitadores Genéticos/genética , Mutação , Células-Tronco Embrionárias Murinas/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-35289322

RESUMO

BACKGROUND: SMARCA2 (SWI/SNF Related, Matrix Associated, Actin Dependent Regulator of Chromatin, Subfamily A, Member 2) is an important ATPase catalytic subunit in the switch-sucrose nonfermenting (SWI/SNF) complex. However, its relationship with the pathological features of NSCLC and its prognosis remain unclear. METHODS: We retrospectively reviewed 2390 patients with surgically resected NSCLC, constructed tissue microarrays (TMAs) and performed immunohistochemical assays. We analyzed the correlation of SAMRCA2 with clinicopathological features and evaluated its prognostic value. RESULTS: Among 2390 NSCLC cases, the negative expression ratios of SAMRCA2, SMARCA4, ARID1A, ARID1B and INI1 were 9.3%, 1.8%, 1.2%, 0.4% and 0%, respectively. In NSCLC, male sex, T3 and T4 stage, moderate and poor differentiation, tumor ≥ 2 cm, Ki67 ≥ 15%, SOX-2 negative expression, middle lobe lesion and adenocarcinoma were relative risk factors affecting SMARCA2-negative expression. In lung adenocarcinomas, high-grade nuclei, histological morphology of acinar and papillary, solid and micropapillary and TTF-1-negative expression were relative risk factors affecting SMARCA2-negative expression. Kaplan-Meier survival analysis showed that the OS was shorter in the SMARCA2-negative group. Multivariate survival analysis revealed that SMARCA2-negative expression was an independent factor correlated with a poor prognosis in NSCLC. CONCLUSION: In conclusion, SMARCA2-negative expression is an independent predictor of a poor outcome of NSCLC and is a potential target for NSCLC treatment.


Assuntos
Adenosina Trifosfatases , Carcinoma Pulmonar de Células não Pequenas/genética , Fatores de Transcrição , Adenosina Trifosfatases/metabolismo , Humanos , Masculino , Estudos Retrospectivos , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
3.
Cell Rep ; 38(7): 110384, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35172142

RESUMO

Impaired production of thymic regulatory T cells (Tregs) is implicated in the development of Aire-dependent autoimmunity. Because Tregs require agonistic T cell receptor stimuli by self-antigens to develop, reduced expression of self-antigens from medullary thymic epithelial cells (mTECs) has been considered to play a major role in the reduced Treg production in Aire deficiency. Here, we show that mTECs abnormally express co-inhibitory receptor CTLA-4 if Aire is non-functional. Upon binding with CD80/CD86 ligands expressed on thymic dendritic cells (DCs), the ectopically expressed CTLA-4 from Aire-deficient mTECs removes the CD80/CD86 ligands from the DCs. This attenuates the ability of DCs to provide co-stimulatory signals and to present self-antigens transferred from mTECs, both of which are required for Treg production. Accordingly, impaired production of Tregs and organ-specific autoimmunity in Aire-deficient mice are rescued by the depletion of CTLA-4 expression from mTECs. Our studies illuminate the significance of mTEC-DC interaction coordinated by Aire for the establishment of thymic tolerance.


Assuntos
Autoimunidade , Antígeno CTLA-4/metabolismo , Timo/citologia , Animais , Antígenos CD/metabolismo , Autoantígenos/metabolismo , Células Epiteliais/metabolismo , Ligantes , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Células Estromais/metabolismo , Linfócitos T Reguladores/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo
4.
Development ; 149(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35179180

RESUMO

Specification of primordial germ cells requires a proportion of the cells in the posterior of the epiblast to reacquire pluripotency. A new paper in Development describes how OVOL2 is involved in regulating the balance between mesodermal fate and germ cell fate during gastrulation. We caught up with the first author, Yuki Naitou, and corresponding author, Katsuhiko Hayashi (Osaka University), to find out more about the paper and their future research.


Assuntos
Células Germinativas/metabolismo , Pesquisadores/psicologia , Fatores de Transcrição/metabolismo , Animais , Autoria , Transição Epitelial-Mesenquimal , Gastrulação , Células Germinativas/citologia , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Humanos , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
5.
Front Immunol ; 13: 829239, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35185921

RESUMO

Introduction: Hyper IgE syndromes (HIES) are a group of rare primary immunodeficiency characterized by high levels of serum IgE, cold abscesses, pulmonary infections, and eczema. ZNF341 deficiency was described in 2018 in 11 patients clinically diagnosed previously with HIES. Eight of those patients, all offspring of consanguineous couples, are from three families who live in a Muslim village in Israel which has approximately 15,000 residents. Objective: Our study aimed to evaluate the prevalence of ZNF341 mutation in the population of the village. Methods: Three hundred DNA samples of females were included in the study. The samples belong to females that were referred to the Meir Medical Center for prenatal genetic testing before pregnancy, during 2017-2019: 200 samples were from the village, and 100 samples of Muslim females were from other villages.All samples were tested by Sanger sequencing for the ZNF341 mutation (c.904C>T, NM_001282933.1). Results: Heterozygous nonsense mutation in ZNF341 was found in ten samples (5%) of the study group compared to zero in the control group (p<0.01). Conclusion: The carrier frequency of the mutation in ZNF341 in the studied village population is 1:20. This high frequency is probably due to founder mutation and consanguineous marriages.


Assuntos
Síndrome de Job/epidemiologia , Síndrome de Job/genética , Fatores de Transcrição/genética , Portador Sadio , Códon sem Sentido , Eczema , Feminino , Humanos , Imunoglobulina E/imunologia , Islamismo , Israel/epidemiologia , Síndrome de Job/imunologia , População , Fatores de Transcrição/deficiência
6.
Nat Commun ; 13(1): 149, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013221

RESUMO

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Assuntos
Caquexia/genética , Fibrose Endomiocárdica/genética , Insuficiência Cardíaca/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Fatores de Transcrição/genética , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/genética , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Caquexia/metabolismo , Caquexia/fisiopatologia , Caquexia/prevenção & controle , Estudos de Casos e Controles , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/genética , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Modelos Animais de Doenças , Fibrose Endomiocárdica/metabolismo , Fibrose Endomiocárdica/fisiopatologia , Fibrose Endomiocárdica/prevenção & controle , Feminino , Regulação da Expressão Gênica , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Testes de Função Cardíaca , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/agonistas , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/deficiência , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatologia , Atrofia Muscular/prevenção & controle , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fatores de Transcrição/agonistas , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/deficiência
7.
Nature ; 602(7895): 129-134, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35082446

RESUMO

Differentiation proceeds along a continuum of increasingly fate-restricted intermediates, referred to as canalization1,2. Canalization is essential for stabilizing cell fate, but the mechanisms that underlie robust canalization are unclear. Here we show that the BRG1/BRM-associated factor (BAF) chromatin-remodelling complex ATPase gene Brm safeguards cell identity during directed cardiogenesis of mouse embryonic stem cells. Despite the establishment of a well-differentiated precardiac mesoderm, Brm-/- cells predominantly became neural precursors, violating germ layer assignment. Trajectory inference showed a sudden acquisition of a non-mesodermal identity in Brm-/- cells. Mechanistically, the loss of Brm prevented de novo accessibility of primed cardiac enhancers while increasing the expression of neurogenic factor POU3F1, preventing the binding of the neural suppressor REST and shifting the composition of BRG1 complexes. The identity switch caused by the Brm mutation was overcome by increasing BMP4 levels during mesoderm induction. Mathematical modelling supports these observations and demonstrates that Brm deletion affects cell fate trajectory by modifying saddle-node bifurcations2. In the mouse embryo, Brm deletion exacerbated mesoderm-deleted Brg1-mutant phenotypes, severely compromising cardiogenesis, and reveals an in vivo role for Brm. Our results show that Brm is a compensable safeguard of the fidelity of mesoderm chromatin states, and support a model in which developmental canalization is not a rigid irreversible path, but a highly plastic trajectory.


Assuntos
Diferenciação Celular , Linhagem da Célula , Mesoderma/citologia , Mesoderma/metabolismo , Miócitos Cardíacos/citologia , Fatores de Transcrição/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Cromatina/genética , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Embrião de Mamíferos , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Miocárdio/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fator 6 de Transcrição de Octâmero/metabolismo , Fenótipo , Proteínas Repressoras/metabolismo , Células-Tronco/citologia , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
8.
Development ; 149(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35029669

RESUMO

In mammals, primordial germ cells (PGCs), the origin of the germ line, are specified from the epiblast at the posterior region where gastrulation simultaneously occurs, yet the functional relationship between PGC specification and gastrulation remains unclear. Here, we show that OVOL2, a transcription factor conserved across the animal kingdom, balances these major developmental processes by repressing the epithelial-to-mesenchymal transition (EMT) that drives gastrulation and the upregulation of genes associated with PGC specification. Ovol2a, a splice variant encoding a repressor domain, directly regulates EMT-related genes and, consequently, induces re-acquisition of potential pluripotency during PGC specification, whereas Ovol2b, another splice variant missing the repressor domain, directly upregulates genes associated with PGC specification. Taken together, these results elucidate the molecular mechanism underlying allocation of the germ line among epiblast cells differentiating into somatic cells through gastrulation. This article has an associated 'The people behind the papers' interview.


Assuntos
Desenvolvimento Embrionário/genética , Gastrulação/genética , Células Germinativas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Células Germinativas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Regulação para Cima , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
9.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166348, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35032595

RESUMO

ARPKD is a genetically inherited kidney disease that manifests by bilateral enlargement of cystic kidneys and liver fibrosis. It shows a range of severity, with 30% of individuals dying early on and the majority having good prognosis if they survive the first year of life. The reasons for this variability remain unclear. Two genes have been shown to cause ARPKD when mutated, PKHD1, mutations in which lead to most of ARPKD cases and DZIP1L, which is associated with moderate ARPKD. This mini review will explore the genetics of ARPKD and discuss potential genetic modifiers and phenocopies that could affect diagnosis.


Assuntos
Rim Policístico Autossômico Recessivo/patologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Humanos , Rim Policístico Autossômico Recessivo/genética , Polimorfismo Genético , Prognóstico , Receptores de Superfície Celular/genética , Transdução de Sinais/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
10.
Exp Cell Res ; 410(2): 112969, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34883111

RESUMO

MEL1 (MDS1/EVI1-like gene 1/PRDM16), a zinc finger protein, is located near the chromosomal breakpoint at 1p36 in human acute myeloid leukemia (AML) cells with the t (1; 3) (p36; q21) translocation. Mel1/Prdm16 is not only a causative gene of leukemia, but also has multiple regulatory functions, such as the regulation of fat metabolism. To investigate the function of Mel1/Prdm16, we generated Mel1/Prdm16-deficient mice, but homozygous deficiency (Mel1/Prdm16-/-) was embryonic lethal at E 11.5. Heterozygous mice showed abnormal cartilage and bone formation in the postnatal skull and long bones, suggesting that Mel1/Prdm16 expression plays an important role in bone development. In osteoblast and chondrocyte cell lines, Mel1/Prdm16 promotes the differentiation of chondrocytes and regulates the differentiation of osteoblasts. Transient repression of the master regulator Runx2 is required for chondrocyte differentiation at an early stage of differentiation. However, in Mel1/Prdm16-suppressed ATDC5 cells, the initial suppression of Runx2 was lacking and its expression was upregulated at the beginning of differentiation, suggesting that chondrogenic differentiation is suppressed in Mel1/Prdm16+/- mesenchymal progenitor cells because Runx2 expression is upregulated during the early stage of differentiation. Thus, the Mel1/Prdm16 gene may be involved in the early repression of Runx2 expression during osteochondral differentiation and promote chondrogenic differentiation.


Assuntos
Osso e Ossos/anatomia & histologia , Osso e Ossos/citologia , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Proteína Morfogenética Óssea 2/metabolismo , Cartilagem/patologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese , Transdução de Sinais , Fatores de Transcrição/deficiência
12.
Cells ; 10(12)2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34943801

RESUMO

Chronic hypoxia increases the resistance of pulmonary arteries by stimulating their contraction and augmenting their coverage by smooth muscle cells (SMCs). While these responses require adjustment of the vascular SMC transcriptome, regulatory elements are not well defined in this context. Here, we explored the functional role of the transcription factor nuclear factor of activated T-cells 5 (NFAT5/TonEBP) in the hypoxic lung. Regulatory functions of NFAT5 were investigated in cultured artery SMCs and lungs from control (Nfat5fl/fl) and SMC-specific Nfat5-deficient (Nfat5(SMC)-/-) mice. Exposure to hypoxia promoted the expression of genes associated with metabolism and mitochondrial oxidative phosphorylation (OXPHOS) in Nfat5(SMC)-/- versus Nfat5fl/fl lungs. In vitro, hypoxia-exposed Nfat5-deficient pulmonary artery SMCs elevated the level of OXPHOS-related transcripts, mitochondrial respiration, and production of reactive oxygen species (ROS). Right ventricular functions were impaired while pulmonary right ventricular systolic pressure (RVSP) was amplified in hypoxia-exposed Nfat5(SMC)-/- versus Nfat5fl/fl mice. Scavenging of mitochondrial ROS normalized the raise in RVSP. Our findings suggest a critical role for NFAT5 as a suppressor of OXPHOS-associated gene expression, mitochondrial respiration, and ROS production in pulmonary artery SMCs that is vital to limit ROS-dependent arterial resistance in a hypoxic environment.


Assuntos
Hipóxia/patologia , Pulmão/patologia , Mitocôndrias/metabolismo , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/patologia , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo , Resistência Vascular , Animais , Pressão Sanguínea , Eletrocardiografia , Regulação da Expressão Gênica , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/fisiopatologia , Metaboloma , Camundongos , Miócitos de Músculo Liso/patologia , Fosforilação Oxidativa , Consumo de Oxigênio , Transporte Proteico , Sístole , Fatores de Transcrição/deficiência , Resistência Vascular/genética
13.
Pathol Oncol Res ; 27: 1610003, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970085

RESUMO

Small cell carcinoma of hypercalcemic type (SCCOHT) is a rare gynaecological neoplasm, originating mostly in the ovaries. Cervical origin of this very aggressive malignancy with unknown histogenesis is an extremely rare condition, without published management recommendations. Alterations in SMARCA4 gene are supposed to play the major role in SCCOHT oncogenesis and their identification is crucial for the diagnosis. Adequate genetic counselling of the patients and their families seems to be of great importance. Optimal management and treatment approaches are not known yet but may extremely influence the prognosis of young female patients that suffer from this very resistant disease. Nowadays, a translational research seems to be the key for the further diagnostic and treatment strategies of SCCOHT. The purpose of the case report is to provide practical information and useful recommendations on the diagnosis, management, and treatment of SMARCA4-deficient carcinoma of the uterine cervix resembling SCCOHT.


Assuntos
Carcinoma de Células Pequenas/metabolismo , DNA Helicases/deficiência , Hipercalcemia/metabolismo , Proteínas Nucleares/deficiência , Fatores de Transcrição/deficiência , Neoplasias do Colo do Útero/metabolismo , Adolescente , Biomarcadores Tumorais/deficiência , Biomarcadores Tumorais/genética , Carcinoma de Células Pequenas/diagnóstico , Carcinoma de Células Pequenas/genética , Carcinoma de Células Pequenas/terapia , DNA Helicases/genética , Evolução Fatal , Feminino , Humanos , Hipercalcemia/diagnóstico , Hipercalcemia/genética , Hipercalcemia/terapia , Mutação , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Neoplasias do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/terapia
14.
Cell Rep ; 37(5): 109943, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731603

RESUMO

The ARID1A subunit of SWI/SNF chromatin remodeling complexes is a potent tumor suppressor. Here, a degron is applied to detect rapid loss of chromatin accessibility at thousands of loci where ARID1A acts to generate accessible minidomains of nucleosomes. Loss of ARID1A also results in the redistribution of the coactivator EP300. Co-incident EP300 dissociation and lost chromatin accessibility at enhancer elements are highly enriched adjacent to rapidly downregulated genes. In contrast, sites of gained EP300 occupancy are linked to genes that are transcriptionally upregulated. These chromatin changes are associated with a small number of genes that are differentially expressed in the first hours following loss of ARID1A. Indirect or adaptive changes dominate the transcriptome following growth for days after loss of ARID1A and result in strong engagement with cancer pathways. The identification of this hierarchy suggests sites for intervention in ARID1A-driven diseases.


Assuntos
Proteínas de Ligação a DNA/deficiência , Células-Tronco Embrionárias Murinas/metabolismo , Nucleossomos/metabolismo , Lesões Pré-Cancerosas/metabolismo , Fatores de Transcrição/deficiência , Transcrição Gênica , Ativação Transcricional , Animais , Sítios de Ligação , Linhagem Celular , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/genética , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Nucleossomos/genética , Lesões Pré-Cancerosas/genética , Proteólise , Fatores de Tempo , Fatores de Transcrição/genética
15.
Sci Rep ; 11(1): 21835, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34750411

RESUMO

Natriuretic peptides exert multiple effects by binding to natriuretic peptide receptors (NPRs). Osteocrin (OSTN) binds with high affinity to NPR-C, a clearance receptor for natriuretic peptides, and inhibits degradation of natriuretic peptides and consequently enhances guanylyl cyclase-A (GC-A/NPR1) signaling. However, the roles of OSTN in the kidney have not been well clarified. Adriamycin (ADR) nephropathy in wild-type mice showed albuminuria, glomerular basement membrane changes, increased podocyte injuries, infiltration of macrophages, and p38 mitogen-activated protein kinase (MAPK) activation. All these phenotypes were improved in OSTN- transgenic (Tg) mice and NPR3 knockout (KO) mice, with no further improvement in OSTN-Tg/NPR3 KO double mutant mice, indicating that OSTN works through NPR3. On the contrary, OSTN KO mice increased urinary albumin levels, and pharmacological blockade of p38 MAPK in OSTN KO mice ameliorated ADR nephropathy. In vitro, combination treatment with ANP and OSTN, or FR167653, p38 MAPK inhibitor, reduced Ccl2 and Des mRNA expression in murine podocytes (MPC5). OSTN increased intracellular cyclic guanosine monophosphate (cGMP) in MPC5 through GC-A. We have elucidated that circulating OSTN improves ADR nephropathy by enhancing GC-A signaling and consequently suppressing p38 MAPK activation. These results suggest that OSTN could be a promising therapeutic agent for podocyte injury.


Assuntos
Nefropatias/metabolismo , Proteínas Musculares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Nefropatias/induzido quimicamente , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Podócitos/patologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Cells ; 10(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34831029

RESUMO

Background: Although several approaches have revealed much about individual factors that regulate pancreatic development, we have yet to fully understand their complicated interplay during pancreas morphogenesis. Gfi1 is transcription factor specifically expressed in pancreatic acinar cells, whose role in pancreas cells fate identity and specification is still elusive. Methods: In order to gain further insight into the function of this factor in the pancreas, we generated animals deficient for Gfi1 specifically in the pancreas. Gfi1 conditional knockout animals were phenotypically characterized by immunohistochemistry, RT-qPCR, and RNA scope. To assess the role of Gfi1 in the pathogenesis of diabetes, we challenged Gfi1-deficient mice with two models of induced hyperglycemia: long-term high-fat/high-sugar feeding and streptozotocin injections. Results: Interestingly, mutant mice did not show any obvious deleterious phenotype. However, in depth analyses demonstrated a significant decrease in pancreatic amylase expression, leading to a diminution in intestinal carbohydrates processing and thus glucose absorption. In fact, Gfi1-deficient mice were found resistant to diet-induced hyperglycemia, appearing normoglycemic even after long-term high-fat/high-sugar diet. Another feature observed in mutant acinar cells was the misexpression of ghrelin, a hormone previously suggested to exhibit anti-apoptotic effects on ß-cells in vitro. Impressively, Gfi1 mutant mice were found to be resistant to the cytotoxic and diabetogenic effects of high-dose streptozotocin administrations, displaying a negligible loss of ß-cells and an imperturbable normoglycemia. Conclusions: Together, these results demonstrate that Gfi1 could turn to be extremely valuable for the development of new therapies and could thus open new research avenues in the context of diabetes research.


Assuntos
Proteínas de Ligação a DNA/deficiência , Diabetes Mellitus/metabolismo , Diabetes Mellitus/prevenção & controle , Fatores de Transcrição/deficiência , Células Acinares/citologia , Células Acinares/metabolismo , Amilases/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Grelina/metabolismo , Proteínas de Homeodomínio/metabolismo , Hiperglicemia/complicações , Hiperglicemia/genética , Integrases/metabolismo , Camundongos Transgênicos , Mutação/genética , Pâncreas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Cell Rep ; 37(1): 109769, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34610319

RESUMO

The ATP-dependent chromatin remodeling factor CHD1 is essential for the assembly of variant histone H3.3 into paternal chromatin during sperm chromatin remodeling in fertilized eggs. It remains unclear, however, if CHD1 has a similar role in normal diploid cells. Using a specifically tailored quantitative mass spectrometry approach, we show that Chd1 disruption results in reduced H3.3 levels in heads of Chd1 mutant flies. Chd1 deletion perturbs brain chromatin structure in a similar way as H3.3 deletion and leads to global de-repression of transcription. The physiological consequences are reduced food intake, metabolic alterations, and shortened lifespan. Notably, brain-specific CHD1 expression rescues these phenotypes. We further demonstrate a strong genetic interaction between Chd1 and H3.3 chaperone Hira. Thus, our findings establish CHD1 as a factor required for the assembly of H3.3-containing chromatin in adult cells and suggest a crucial role for CHD1 in the brain as a regulator of organismal health and longevity.


Assuntos
Encéfalo/metabolismo , Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/metabolismo , Histonas/metabolismo , Metaboloma/fisiologia , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/química , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Comportamento Alimentar , Feminino , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/análise , Longevidade , Masculino , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo
18.
Immunohorizons ; 5(10): 802-817, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663594

RESUMO

ARID3a is a DNA-binding protein important for normal hematopoiesis in mice and for in vitro lymphocyte development in human cultures. ARID3a knockout mice die in utero with defects in both early hematopoietic stem cell populations and erythropoiesis. Recent transcriptome analyses in human erythropoietic systems revealed increases in ARID3a transcripts implicating potential roles for ARID3a in human erythrocyte development. However, ARID3a transcript levels do not faithfully reflect protein levels in many cells, and the functions and requirements for ARID3a protein in those systems have not been explored. We used the erythroleukemic cell line K562 as a model to elucidate functions of ARID3a protein in early human erythropoiesis. ARID3a knockdown of hemin-stimulated K562 cells resulted in lack of fetal globin production and modifications in gene expression. Temporal RNA sequencing data link ARID3a expression with the important erythroid regulators Gata1, Gata2, and Klf1 Ablation of ARID3a using CRISPR-Cas9 further demonstrated it is required to maintain chromatin structures associated with erythropoietic differentiation potential. These data demonstrate that the ARID3a protein is required for early erythropoietic events and provide evidence for the requirement of ARID3a functions for proper maintenance of appropriate chromatin structures.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/deficiência , Eritropoese/genética , Fatores de Transcrição/deficiência , Sistemas CRISPR-Cas/genética , Cromatina/genética , Sequenciamento de Cromatina por Imunoprecipitação , Proteínas de Ligação a DNA/genética , Globulinas Fetais/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Células K562 , RNA-Seq , Fatores de Transcrição/genética
19.
Cell Death Dis ; 12(11): 990, 2021 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-34689165

RESUMO

ARID1A, encoding a subunit of SWI/SNF chromatin remodeling complex, is widely recognized as a tumor suppressor gene in multiple tumor types including liver cancer. Previous studies have demonstrated that ARID1A deficiency can cause liver cancer metastasis, possibly due to the altered chromatin organization, however the underlying mechanisms remain poorly understood. To address the effect of Arid1a deficiency on chromatin organization, we generated chromatin interaction matrices, and exploited the conformation changes upon Arid1a depletion in hepatocytes. Our results demonstrated that Arid1a deficiency induced A/B compartment switching, topologically associated domain (TAD) remodeling, and decrease of chromatin loops. Further mechanism studies revealed that ATPase BRG1 of SWI/SNF complex could physically interact with RAD21, a structural subunit of chromatin architectural element cohesin; whereas ARID1A deficiency significantly diminished the coupled BRG1-RAD21. Interestingly, the tumor-associated genes within the switched compartments were differentially expressed depending upon Arid1a depletion or not. As a consequence of ARID1A deficiency-induced conformational alteration, the dysregulation of some genes such as PMP22 and GSC, promoted the invasion capacity of liver cancer cells. This study provides an insight into liver cancer tumorigenesis and progression related to ARID1A mutations.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Cromatina/metabolismo , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/deficiência , Neoplasias Hepáticas/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Metástase Neoplásica , Transfecção
20.
BMB Rep ; 54(9): 470-475, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34488932

RESUMO

Low-dose metronomic chemotherapy has been introduced as a less toxic and effective strategy to inhibit tumor angiogenesis, but its anti-angiogenic mechanism on endothelial progenitor cells (EPCs) has not been fully elucidated. Here, we investigated the functional role of regulated in development and DNA damage response 1 (REDD1), an endogenous inhibitor of mTORC1, in low-dose doxorubicin (DOX)-mediated dysregulation of EPC functions. DOX treatment induced REDD1 expression in bone marrow mononuclear cells (BMMNCs) and subsequently reduced mTORC1-dependent translation of endothelial growth factor (VEGF) receptor (Vegfr)-2 mRNA, but not that of the mRNA transcripts for Vegfr-1, epidermal growth factor receptor, and insulin-like growth factor-1 receptor. This selective event was a risk factor for the inhibition of BMMNC differentiation into EPCs and their angiogenic responses to VEGF-A, but was not observed in Redd1-deficient BMMNCs. Low-dose metronomic DOX treatment reduced the mobilization of circulating EPCs in B16 melanoma-bearing wild-type but not Redd1-deficient mice. However, REDD1 overexpression inhibited the differentiation and mobilization of EPCs in both wild-type and Redd1-deficient mice. These data suggest that REDD1 is crucial for metronomic DOX-mediated EPC dysfunction through the translational repression of Vegfr-2 transcript, providing REDD1 as a potential therapeutic target for the inhibition of tumor angiogenesis and tumor progression. [BMB Reports 2021; 54(9): 470-475].


Assuntos
Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Doxorrubicina/farmacologia , Fatores de Transcrição/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Neovascularização Patológica , Óxido Nítrico/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...