Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Am J Physiol Renal Physiol ; 324(6): F581-F589, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37141146

RESUMO

Chronic kidney disease (CKD) is a major health problem. Kidney fibrosis is a hallmark and final common pathway of CKD. The Hippo/yes-associated protein (YAP) pathway regulates organ size, inflammation, and tumorigenesis. Our previous study demonstrated tubular YAP activation by tubule-specific double knockout of mammalian STE20-like protein kinase 1/2 (Mst1/2) induced CKD in mice, but the underlying mechanisms remain to be fully elucidated. Activator protein (AP)-1 activation was found to promote tubular atrophy and tubulointerstitial fibrosis. Therefore, we studied whether YAP regulates AP-1 expression in the kidney. We found that expression of various AP-1 components was induced in kidneys subjected to unilateral ureteric obstruction and in Mst1/2 double knockout kidneys, and these inductions were blocked by deletion of Yap in tubular cells, with Fosl1 being most affected compared with other AP-1 genes. Inhibition of Yap also most highly suppressed Fosl1 expression among AP-1 genes in HK-2 and IMCD3 renal tubular cells. YAP bound to the Fosl1 promoter and promoted Fosl1 promoter-luciferase activity. Our results suggest that YAP controls AP-1 expression and that Fosl1 is the primary target of YAP in renal tubular cells.NEW & NOTEWORTHY Yes-associated protein (YAP) activation leads to tubular injury, renal inflammation, and fibrosis, but the underlying mechanisms are not fully understood. We now provide genetic evidence that YAP promotes activator protein-1 expression and that Fosl1 is the primary target of YAP in renal tubular cells.


Assuntos
Insuficiência Renal Crônica , Obstrução Ureteral , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Epiteliais/metabolismo , Fibrose , Inflamação/metabolismo , Rim/metabolismo , Mamíferos/metabolismo , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Proteínas de Sinalização YAP
2.
J Biol Chem ; 298(7): 102010, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35525270

RESUMO

Follistatin (FS)-like 1 (FSTL1) is a member of the FS-SPARC (secreted protein, acidic and rich in cysteine) family of secreted and extracellular matrix proteins. The functions of FSTL1 have been studied in heart and lung injury as well as in wound healing; however, the role of FSTL1 in the kidney is largely unknown. Here, we show using single-cell RNA-Seq that Fstl1 was enriched in stromal cells in obstructed mouse kidneys. In addition, immunofluorescence demonstrated that FSTL1 expression was induced in fibroblasts during kidney fibrogenesis in mice and human patients. We demonstrate that FSTL1 overexpression increased renal fibrosis and activated the Wnt/ß-catenin signaling pathway, known to promote kidney fibrosis, but not the transforming growth factor ß (TGF-ß), Notch, Hedgehog, or Yes-associated protein (YAP) signaling pathways in obstructed mouse kidneys, whereas inhibition of FSTL1 lowered Wnt/ß-catenin signaling. Importantly, we show that FSTL1 interacted with Wnt ligands and the Frizzled (FZD) receptors but not the coreceptor lipoprotein receptor-related protein 6 (LRP6). Specifically, we found FSTL1 interacted with Wnt3a through its extracellular calcium-binding (EC) domain and von Willebrand factor type C-like (VWC) domain, and with FZD4 through its EC domain. Furthermore, we show that FSTL1 increased the association of Wnt3a with FZD4 and promoted Wnt/ß-catenin signaling and fibrogenesis. The EC domain interacting with both Wnt3a and FZD4 also enhanced Wnt3a signaling. Therefore, we conclude that FSTL1 is a novel extracellular enhancer of the Wnt/ß-catenin pathway.


Assuntos
Proteínas Relacionadas à Folistatina , Receptores Frizzled , Rim , Via de Sinalização Wnt , Animais , Proteínas Relacionadas à Folistatina/genética , Proteínas Relacionadas à Folistatina/metabolismo , Receptores Frizzled/metabolismo , Humanos , Rim/metabolismo , Rim/fisiopatologia , Ligantes , Camundongos , Proteína Wnt3A
3.
J Biol Chem ; 298(3): 101701, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35148988

RESUMO

Increasing evidences indicate that unlimited capacity for self-renewal and pluripotency, two unique properties of embryonic stem cells (ESCs), are intrinsically linked to cell cycle control. However, the precise mechanisms coordinating cell fate decisions and cell cycle regulation remain to be fully explored. Here, using CRISPR/Cas9-mediated genome editing, we show that in ESCs, deficiency of components of the cell cycle regulatory MuvB complex Lin54 or Lin52, but not Lin9 or Lin37, triggers G2/M arrest, loss of pluripotency, and spontaneous differentiation. Further dissection of these phenotypes demonstrated that this cell cycle arrest is accompanied by the gradual activation of mesoendodermal lineage-specifying genes. Strikingly, the abnormalities observed in Lin54-null ESCs were partially but significantly rescued by ectopic coexpression of genes encoding G2/M proteins Cyclin B1 and Cdk1. Thus, our study provides new insights into the mechanisms by which the MuvB complex determines cell fate through regulation of the cell cycle machinery.


Assuntos
Proteínas de Ciclo Celular , Células-Tronco Embrionárias , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular , Humanos , Camundongos , Fatores de Transcrição/metabolismo
4.
Stem Cell Reports ; 16(3): 566-581, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33606987

RESUMO

Polycomb group (PcG) proteins exist in distinct multi-protein complexes and play a central role in silencing developmental genes, yet the underlying mechanisms remain elusive. Here, we show that deficiency of retinoblastoma binding protein 4 (RBBP4), a component of the Polycomb repressive complex 2 (PRC2), in embryonic stem cells (ESCs) leads to spontaneous differentiation into mesendodermal lineages. We further show that Rbbp4 and core PRC2 share an important number of common genomic targets, encoding regulators involved in early germ layer specification. Moreover, we find that Rbbp4 is absolutely essential for genomic targeting of PRC2 to a subset of developmental genes. Interestingly, we demonstrate that Rbbp4 is necessary for sustaining the expression of Oct4 and Sox2 and that the forced co-expression of Oct4 and Sox2 fully rescues the pluripotency of Rbbp4-null ESCs. Therefore, our study indicates that Rbbp4 links maintenance of the pluripotency regulatory network with repression of mesendoderm lineages.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Complexo Repressor Polycomb 2/fisiologia , Proteína 4 de Ligação ao Retinoblastoma/fisiologia , Animais , Linhagem Celular , Autorrenovação Celular , Sequenciamento de Cromatina por Imunoprecipitação , Técnicas de Inativação de Genes , Células HEK293 , Histonas/metabolismo , Humanos , Metilação , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo
5.
J Am Soc Nephrol ; 31(5): 946-961, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32253273

RESUMO

BACKGROUND: The serine/threonine kinases MST1 and MST2 are core components of the Hippo pathway, which has been found to be critically involved in embryonic kidney development. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are the pathway's main effectors. However, the biologic functions of the Hippo/YAP pathway in adult kidneys are not well understood, and the functional role of MST1 and MST2 in the kidney has not been studied. METHODS: We used immunohistochemistry to examine expression in mouse kidneys of MST1 and MST2, homologs of Hippo in Drosophila. We generated mice with tubule-specific double knockout of Mst1 and Mst2 or triple knockout of Mst1, Mst2, and Yap. PCR array and mouse inner medullary collecting duct cells were used to identify the primary target of Mst1/Mst2 deficiency. RESULTS: MST1 and MST2 were predominantly expressed in the tubular epithelial cells of adult kidneys. Deletion of Mst1/Mst2 in renal tubules increased activity of YAP but not TAZ. The kidneys of mutant mice showed progressive inflammation, tubular and glomerular damage, fibrosis, and functional impairment; these phenotypes were largely rescued by deletion of Yap in renal tubules. TNF-α expression was induced via both YAP-dependent and YAP-independent mechanisms, and TNF-α and YAP amplified the signaling activities of each other in the tubules of kidneys with double knockout of Mst1/Mst2. CONCLUSIONS: Our findings show that tubular Mst1/Mst2 deficiency leads to CKD through both the YAP and non-YAP pathways and that tubular YAP activation induces renal fibrosis. The pathogenesis seems to involve the reciprocal stimulation of TNF-α and YAP signaling activities.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Ciclo Celular/fisiologia , Túbulos Renais/enzimologia , Proteínas Serina-Treonina Quinases/deficiência , Insuficiência Renal Crônica/enzimologia , Animais , Células Cultivadas , Fibrose , Regulação da Expressão Gênica , Via de Sinalização Hippo , Marcação In Situ das Extremidades Cortadas , Rim/embriologia , Rim/enzimologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Serina-Treonina Quinase 3 , Transdução de Sinais , Transativadores/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Proteínas de Sinalização YAP
6.
Cell Death Differ ; 27(10): 2797-2809, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32332916

RESUMO

Although the roles of the Hippo pathway in organogenesis and tumorigenesis have been well studied in multiple organs, its role in sperm maturation and male fertility has not been investigated. The initial segment (IS) of the epididymis plays a critical role in sperm maturation. IS differentiation is governed by ERK1/2, but the mechanisms of ERK1/2 activation in IS are not fully understood. Here we show that double knockout (dKO) of mammalian sterile 20-like kinases 1 and 2 (Mst1 and Mst2), homologs of Hippo in Drosophila, in the epididymal epithelium led to male infertility in mice. Sperm in the cauda epididymides of mutant mice were immotile with flagellar angulation and severely disorganized structures. Loss of Mst1/2 activated YAP and increased proliferation and cell death in all the segments of epididymis. The mutant mice showed substantially suppressed MEK/ERK signaling in the IS and failed IS differentiation. Deletion of Yap restored the reduced MEK/ERK signaling, and partially rescued the defective IS differentiation and fertility in Mst1/2 dKO mice. Our results demonstrate that YAP inhibits the MEK/ERK pathway in IS epithelial cells, and MST1/2 control IS differentiation and fertility at least partially by repressing YAP. Taken together, the Hippo pathway is essential for sperm maturation and male fertility.


Assuntos
Epididimo , Células Epiteliais , Infertilidade Masculina/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Diferenciação Celular , Epididimo/citologia , Epididimo/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Serina-Treonina Quinase 3
7.
J Orthop Surg Res ; 14(1): 59, 2019 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-30786900

RESUMO

BACKGROUND: Proinflammatory cytokines, which can upregulate the expression of matrix-degrading enzymes in chondrocytes, play important roles in the development of osteoarthritis. BET family proteins, acting as the "readers" of acetylated modifications on histones, have been linked to transcriptional regulation. And a BET protein inhibitor, I-BET151, has been shown to inhibit the induction of matrix-degrading enzymes by proinflammatory cytokines in chondrocytes. Our objective is to clarify the role and mechanism of BET proteins on matrix-degrading enzyme gene expression by using a human chondrosarcoma cell line (SW1353). METHODS: We pretreated SW1353 cells with I-BET151 prior to treatment with IL-1ß or TNF-α and then checked the expression of four matrix-degrading enzyme genes (MMP1, MMP3, MMP13, and ADAMTS4). We performed knockdown of BET protein family members (BRD2, BRD3, and BRD4) with corresponding siRNAs in SW1353 cells prior to treatment with IL-1ß or TNF-α and checked the expression of the matrix-degrading enzyme genes. We evaluated Brd-mediated transcriptional regulation on the matrix-degrading enzyme genes by ChIP assay. RESULTS: We confirmed that I-BET151 could suppress the IL-1ß- or TNF-α-induced expression of MMP1, MMP3, MMP13, and ADAMTS4 in SW1353 cells. Brd3 and Brd4 were required for the IL-1ß- or TNF-α-induced expression of matrix-degrading enzyme genes in SW1353 cells. We revealed that inducible acetylation of H4k5/8/12 and the recruitment of Brd3, Brd4, and p-TEFb to chromatin were involved in IL-1ß- or TNF-α-induced transcription. CONCLUSIONS: Our findings suggested that Brd3 and Brd4 were essential for the IL-1ß- or TNF-α-induced transcription of matrix-degrading enzyme genes, and recruitment of Brd3 and Brd4 to chromatin of these genes played the main role in this process.


Assuntos
Cromatina/metabolismo , Citocinas/metabolismo , Regulação Enzimológica da Expressão Gênica , Mediadores da Inflamação/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Citocinas/genética , Humanos , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética
8.
Cell Death Differ ; 26(11): 2194-2207, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30760872

RESUMO

Lethal (3) malignant brain tumor like 2 (L3MBTL2) is a member of the MBT-domain proteins, which are involved in transcriptional repression and implicated in chromatin compaction. Our previous study has shown that L3MBTL2 is highly expressed in the testis, but its role in spermatogenesis remains unclear. In the present study, we found that L3MBTL2 was most highly expressed in pachytene spermatocytes within the testis. Germ cell-specific ablation of L3mbtl2 in the testis led to increased abnormal spermatozoa, progressive decrease of sperm counts and premature testicular failure in mice. RNA-sequencing analysis on L3mbtl2 deficient testes confirmed that L3MBTL2 was a transcriptional repressor but failed to reveal any significant changes in spermatogenesis-associated genes. Interestingly, L3mbtl2 deficiency resulted in increased γH2AX deposition in the leptotene spermatocytes, subsequent inappropriate retention of γH2AX on autosomes, and defective crossing-over and synapsis during the pachytene stage of meiosis I, and more germ cell apoptosis and degeneration in aging mice. L3MBTL2 interacted with the histone ubiquitin ligase RNF8. Inhibition of L3MBTL2 reduced nuclear RNF8 and ubH2A levels in GC2 cells. L3mbtl2 deficiency led to decreases in the levels of the RNF8 and ubH2A pathway and in histone acetylation in elongating spermatids, and in protamine 1 deposition and chromatin condensation in sperm. These results suggest that L3MBTL2 plays important roles in chromatin remodeling during meiosis and spermiogenesis.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Cromatina/metabolismo , Proteínas Nucleares/genética , Espermatócitos/metabolismo , Espermatogênese/genética , Fatores de Transcrição/genética , Acetilação , Animais , Apoptose/genética , Montagem e Desmontagem da Cromatina/fisiologia , Histonas/metabolismo , Masculino , Prófase Meiótica I/fisiologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/metabolismo , Estágio Paquíteno/fisiologia , Proteínas do Grupo Polycomb/metabolismo , Contagem de Espermatozoides , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
9.
J Orthop Surg Res ; 13(1): 118, 2018 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-29784003

RESUMO

BACKGROUND: Proinflammatory cytokines, which can upregulate the expression of matrix-degrading enzymes in chondrocytes, play important roles in the development of osteoarthritis. And a BET protein inhibitor, I-BET151, has been shown to exert an anti-inflammatory effect by repressing the BET protein-mediated expression of inflammatory genes. Our objective is to investigate the effect of I-BET151 on a surgical mouse model of osteoarthritis (OA) and human chondrocytes. METHODS: We first treated a surgical mouse model of OA with I-BET151 once per day and evaluated the knee joints at 6 and 8 weeks after treatment. We then pretreated the human chondrocytes with I-BET151 prior to treatment with IL-1ß or TNF-α and checked the expression and activity of the matrix-degrading enzyme genes. We also checked the expression of ACAN, COL2A1, and SOX9. RESULTS: We demonstrated that I-BET151 could prevent articular cartilage damage in the surgical mouse model of OA at an earlier time after treatment, but not at a later time after treatment. I-BET151 could robustly suppress the IL-1ß- and TNF-α-induced expression and activity of several matrix-degrading enzymes in human chondrocytes. I-BET151 could also suppress the expression of ACAN, COL2A1, and SOX9. CONCLUSIONS: Our findings suggested that inhibiting BET proteins could exert a repression effect on both of chondrocyte anabolism and catabolism, and the effect of BET protein inhibitor on surgical mouse model of OA needs further evaluation.


Assuntos
Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Articulação do Joelho/metabolismo , Idoso , Animais , Cartilagem Articular/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Humanos , Articulação do Joelho/efeitos dos fármacos , Articulação do Joelho/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Pessoa de Meia-Idade , Osteoartrite do Joelho/tratamento farmacológico , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia
10.
Exp Ther Med ; 15(4): 3623-3632, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29545892

RESUMO

The ovariectomized (OVX) mouse model has been widely accepted to be suitable for the study of postmenopausal osteoporosis. However, whether C57BL/6J mice, a commonly used genetic background mouse strain, is an appropriate model for postmenopausal osteoporosis remains controversial. The present study investigated the effect of the OVX model on alterations in bone density and microarchitecture in C57BL/6J female mice of different ages. C57BL/6J mice were divided into 8-, 12- and 16-week-old groups (OVX8, OVX12 and OVX16) from the beginning of OVX. At 8 weeks post-surgery, the mice were anesthetized and micro-computed tomography was used to analyze the bone density and microarchitecture. The results revealed that OVX-induced loss of cancellous bone was greatest in OVX8, moderate in OVX12, and only a weak bone loss was observed in the OVX16 group when compared with the SHAM16 control group. In addition, the effect of genetic backgrounds in response to the OVX model were examined. Several other strains of mice, including inbred (BALB/c) and outbred (ICR and Kunming), were used in the present study, all of which were subjected to OVX at 8 weeks of age. The present findings revealed that the highest rate of bone loss was detected in C57BL/6J female mice. In addition, treatment with estrogen (17ß-estradiol, 30 µg/kg five times per week) led to a significant increase in bone density in C57BL/6J mice compared with the other strains of mice. Therefore, these results may provide novel insights into the age- and strain-associated effect of OVX on regulating turnover of bone in female mice. The present findings also suggest 8-week-old C57BL/6J mice as an animal model for postmenopausal osteoporosis and preclinical testing of potential therapies for this disease.

11.
Kidney Int ; 93(4): 855-870, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29276099

RESUMO

DNA damage contributes to renal tubular cell death during kidney injury, but how DNA damage in tubular cells is regulated is not fully understood. Lethal (3) malignant brain tumor-like 2 (L3MBTL2), a novel polycomb group protein, has been implicated in regulating chromatin architecture. However, the biological functions of L3MBTL2 are largely undefined. Here we found that L3MBTL2 was expressed in the nuclei of renal tubular epithelial cells in mice. Ablation of L3mbtl2 in renal tubular cells resulted in increases in nuclear DNA damage, p53 activation, apoptosis, tubular injury and kidney dysfunction after cisplatin treatment or unilateral ureteral obstruction. In vitro, inhibition of L3MBTL2 sequentially promoted histone γH2AX expression, p53 activation and apoptosis in cisplatin-treated mouse proximal tubular TKPTS cells. Inhibition of p53 activity attenuated the apoptosis induced by L3mbtl2 deficiency after cisplatin treatment both in vivo and in vitro. Intriguingly, unlike other polycomb proteins, L3MBTL2 was not recruited to DNA damage sites, but instead increased nuclear chromatin density and reduced initial DNA damage load. Thus, L3MBTL2 plays a protective role in kidney injury, in part by inhibiting the DNA damage-p53-apoptosis pathway.


Assuntos
Injúria Renal Aguda/metabolismo , Apoptose , Dano ao DNA , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas Nucleares/metabolismo , Insuficiência Renal Crônica/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Células Cultivadas , Montagem e Desmontagem da Cromatina , Cisplatino , Modelos Animais de Doenças , Células Epiteliais/patologia , Histonas/metabolismo , Túbulos Renais Proximais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Transdução de Sinais , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Obstrução Ureteral/complicações
12.
Int J Biol Sci ; 11(5): 604-17, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25892967

RESUMO

DNA methylation regulates many cellular processes, including embryonic development, transcription, chromatin structure, X-chromosome inactivation, genomic imprinting and chromosome stability. DNA methyltransferases establish and maintain the presence of 5-methylcytosine (5mC), and ten-eleven translocation cytosine dioxygenases (TETs) oxidise 5mC to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC), which can be removed by base excision repair (BER) proteins. Multiple forms of DNA methylation are recognised by methyl-CpG binding proteins (MeCPs), which play vital roles in chromatin-based transcriptional regulation, DNA repair and replication. Accordingly, defects in DNA methylation and its mediators may cause silencing of tumour suppressor genes and misregulation of multiple cell cycles, DNA repair and chromosome stability genes, and hence contribute to genome instability in various human diseases, including cancer. Thus, understanding functional genetic mutations and aberrant expression of these DNA methylation mediators is critical to deciphering the crosstalk between concurrent genetic and epigenetic alterations in specific cancer types and to the development of new therapeutic strategies.


Assuntos
Carcinogênese , Metilação de DNA/fisiologia , Epigenômica , Humanos , Neoplasias/metabolismo
13.
J Exp Med ; 211(5): 909-27, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24711581

RESUMO

The zinc finger transcriptional repressor Gfi-1b is essential for erythroid and megakaryocytic development in the embryo. Its roles in the maintenance of bone marrow erythropoiesis and thrombopoiesis have not been defined. We investigated Gfi-1b's adult functions using a loxP-flanked Gfi-1b allele in combination with a novel doxycycline-inducible Cre transgene that efficiently mediates recombination in the bone marrow. We reveal strict, lineage-intrinsic requirements for continuous adult Gfi-1b expression at two distinct critical stages of erythropoiesis and megakaryopoiesis. Induced disruption of Gfi-1b was lethal within 3 wk with severely reduced hemoglobin levels and platelet counts. The erythroid lineage was arrested early in bipotential progenitors, which did not give rise to mature erythroid cells in vitro or in vivo. Yet Gfi-1b(-/-) progenitors had initiated the erythroid program as they expressed many lineage-restricted genes, including Klf1/Eklf and Erythropoietin receptor. In contrast, the megakaryocytic lineage developed beyond the progenitor stage in Gfi-1b's absence and was arrested at the promegakaryocyte stage, after nuclear polyploidization, but before cytoplasmic maturation. Genome-wide analyses revealed that Gfi-1b directly regulates a wide spectrum of megakaryocytic and erythroid genes, predominantly repressing their expression. Together our study establishes Gfi-1b as a master transcriptional repressor of adult erythropoiesis and thrombopoiesis.


Assuntos
Plaquetas/fisiologia , Medula Óssea/fisiologia , Eritrócitos/fisiologia , Regulação da Expressão Gênica/genética , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Primers do DNA/genética , Células-Tronco Embrionárias/metabolismo , Citometria de Fluxo , Vetores Genéticos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Microscopia de Fluorescência , Mutagênese , Proteínas Proto-Oncogênicas/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/genética
14.
Cell Stem Cell ; 11(3): 319-32, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22770845

RESUMO

L3mbtl2 has been implicated in transcriptional repression and chromatin compaction but its biological function has not been defined. Here we show that disruption of L3mbtl2 results in embryonic lethality with failure of gastrulation. This correlates with compromised proliferation and abnormal differentiation of L3mbtl2(-/-) embryonic stem (ES) cells. L3mbtl2 regulates genes by recruiting a Polycomb Repressive Complex1 (PRC1)-related complex, resembling the previously described E2F6-complex, and including G9A, Hdac1, and Ring1b. The presence of L3mbtl2 at target genes is associated with H3K9 dimethylation, low histone acetylation, and H2AK119 ubiquitination, but the latter is neither dependent on L3mbtl2 nor sufficient for repression. Genome-wide studies revealed that the L3mbtl2-dependent complex predominantly regulates genes not bound by canonical PRC1 and PRC2. However, some developmental regulators are repressed by the combined activity of all three complexes. Together, we have uncovered a highly selective, essential role for an atypical PRC1-family complex in ES cells and early development.


Assuntos
Desenvolvimento Embrionário , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Montagem e Desmontagem da Cromatina/genética , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Genoma/genética , Camundongos , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/química , Células-Tronco Pluripotentes/citologia , Complexo Repressor Polycomb 2 , Ligação Proteica/genética , Estrutura Terciária de Proteína , Proteínas Repressoras/metabolismo , Fatores de Transcrição/química , Transcrição Gênica , Proteínas Supressoras de Tumor/química , Dedos de Zinco
15.
J Biol Chem ; 285(36): 27767-75, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20592034

RESUMO

L3MBTL1, a paralogue of Drosophila tumor suppressor lethal(3)malignant brain tumor (l(3)mbt), binds histones in a methylation state-dependent manner and contributes to higher order chromatin structure and transcriptional repression. It is the founding member of a family of MBT domain-containing proteins that has three members in Drosophila and nine in mice and humans. Knockdown experiments in cell lines suggested that L3MBTL1 has non-redundant roles in the suppression of oncogene expression. We generated a mutant mouse strain that lacks exons 13-20 of L3mbtl1. Markedly reduced levels of a mutant mRNA with an out-of-frame fusion of exons 12 and 21 were expressed, but a mutant protein was undetectable by Western blot analysis. L3MBTL1(-/-) mice developed and reproduced normally. The highest expression of L3MBTL1 was detected in the brain, but its disruption did not affect brain development, spontaneous movement, and motor coordination. Despite previous implications of L3mbtl1 in the biology of hematopoietic transcriptional regulators, lack of L3MBTL1 did not result in deficiencies in lymphopoiesis or hematopoiesis. In contrast with its demonstrated biochemical activities, embryonic stem (ES) cells lacking L3MBTL1 displayed no abnormalities in H4 lysine 20 (H4K20) mono-, di-, or trimethylation; had normal global chromatin density as assessed by micrococcal nuclease digests; and expressed normal levels of c-myc. Embryonic fibroblasts lacking L3MBTL1 displayed unaltered cell cycle arrest and down-regulation of cyclin E expression after irradiation. In cohorts of mice followed for more than 2 years, lack of L3MBTL1 did not alter normal lifespan or survival with or without sublethal irradiation.


Assuntos
Cromatina/metabolismo , Crescimento e Desenvolvimento/genética , Neoplasias/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Ciclina E/metabolismo , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/efeitos da radiação , Técnicas de Inativação de Genes , Células HeLa , Histonas/química , Histonas/metabolismo , Humanos , Longevidade/genética , Longevidade/efeitos da radiação , Linfopoese/genética , Lisina/metabolismo , Masculino , Metilação , Camundongos , Dados de Sequência Molecular , Neoplasias/genética , Proteínas Nucleares/química , Proteínas Nucleares/deficiência , Proteínas Repressoras , Taxa de Sobrevida , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/deficiência
16.
Cell Stem Cell ; 5(1): 43-53, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-19570513

RESUMO

The initial steps in the pathogenesis of acute leukemia remain incompletely understood. The TEL-AML1 gene fusion, the hallmark translocation in Childhood Acute Lymphoblastic Leukemia and the first hit, occurs years before the clinical disease, most often in utero. We have generated mice in which TEL-AML1 expression is driven from the endogenous promoter and can be targeted to specific populations. TEL-AML1 renders mice prone to malignancy after chemical mutagenesis when expressed in hematopoietic stem cells (HSCs), but not in early lymphoid progenitors. We reveal that TEL-AML1 markedly increases the number of HSCs and predominantly maintains them in the quiescent (G(0)) stage of the cell cycle. TEL-AML1(+) HSCs retain self-renewal properties and contribute to hematopoiesis, but fail to out-compete normal HSCs. Our work shows that stem cells are susceptible to subversion by weak oncogenes that can subtly alter their molecular program to provide a latent reservoir for the accumulation of further mutations.


Assuntos
Células da Medula Óssea/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/genética , Proteínas de Fusão Oncogênica/genética , Oncogenes , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Fatores de Ligação ao Core/metabolismo , Desenvolvimento Embrionário , Marcação de Genes , Hepatócitos/metabolismo , Humanos , Interleucina-7/metabolismo , Leucemia Mieloide Aguda/metabolismo , Camundongos , Modelos Animais , Fusão Oncogênica , Proteínas de Fusão Oncogênica/metabolismo
17.
Immunity ; 26(4): 461-75, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17398123

RESUMO

Despite constant contact with the large population of commensal bacteria, the colonic mucosa is normally hyporesponsive to these potentially proinflammatory signals. Here we report that the single immunoglobulin IL-1 receptor-related molecule (SIGIRR), a negative regulator for Toll-IL-1R signaling, plays a critical role in gut homeostasis, intestinal inflammation, and colitis-associated tumorigenesis by maintaining the microbial tolerance of the colonic epithelium. SIGIRR-deficient (Sigirr(-/-)) colonic epithelial cells displayed commensal bacteria-dependent homeostatic defects, as shown by constitutive upregulation of inflammatory genes, increased inflammatory responses to dextran sulfate sodium (DSS) challenge, and increased Azoxymethane (AOM)+DSS-induced colitis-associated tumorigenesis. Gut epithelium-specific expression of the SIGIRR transgene in the SIGIRR-deficient background reduced the cell survival of the SIGIRR-deficient colon epithelium, abrogated the hypersensitivity of the Sigirr(-/-) mice to DSS-induced colitis, and reduced AOM+DSS-induced tumorigenesis. Taken together, our results indicate that epithelium-derived SIGIRR is critical in controlling the homeostasis and innate immune responses of the colon to enteric microflora.


Assuntos
Transformação Celular Neoplásica/imunologia , Colite/imunologia , Colo/imunologia , Neoplasias do Colo/imunologia , Imunidade Inata/genética , Receptores de Interleucina-1/fisiologia , Animais , Bactérias/imunologia , Proliferação de Células , Transformação Celular Neoplásica/genética , Quimiocinas/genética , Colite/genética , Colite/patologia , Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação da Expressão Gênica , Homeostase/genética , Homeostase/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Receptores de Interleucina-1/genética , Fator de Transcrição STAT3/metabolismo
18.
J Biol Chem ; 280(26): 25233-41, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15866876

RESUMO

The Toll-interleukin-1 receptor (TIR) domain-containing orphan receptor SIGIRR (single immunoglobulin interleukin-1 receptor-related protein) acts as a negative regulator of interleukin (IL)-1 and lipopolysaccharide (LPS) signaling. Endogenous SIGIRR transiently interacted with IL-1 receptor and the receptor-proximal signaling components (MyD88, IRAK, and tumor necrosis factor receptor-associated factor 6) upon IL-1 stimulation, indicating that SIGIRR interacts with the IL-1 receptor complex in a ligand-dependent manner. Similar interaction was also observed between SIGIRR and Toll-like receptor 4 receptor complex upon LPS stimulation. To identify the domains of SIGIRR required for its interaction with the Toll-like receptor 4 and IL-1 receptor complexes, several SIGIRR deletion mutants were generated, including DeltaN (lacking the extracellular immunoglobulin (Ig) domain with deletion of amino acids 1-119), DeltaC (lacking the C-terminal domain with deletion of amino acids 313-410), and DeltaTIR (lacking the TIR domain with deletion of amino acids 161-313). Whereas both the extracellular Ig domain and the intracellular TIR domains are important for SIGIRR to inhibit IL-1 signaling, only the TIR domain is necessary for SIGIRR to inhibit LPS signaling. The extracellular Ig domain exerts its inhibitory role in IL-1 signaling by interfering with the heterodimerization of IL-1 receptor and IL-1RAcP, whereas the intracellular TIR domain inhibits both IL-1 and LPS signaling by attenuating the recruitment of receptor-proximal signaling components to the receptor. These results indicate that SIGIRR inhibits IL-1 and LPS signaling pathways through differential mechanisms.


Assuntos
Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Antígenos de Diferenciação/metabolismo , Ligação Competitiva , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , DNA/química , DNA/metabolismo , Dimerização , Deleção de Genes , Humanos , Imunoprecipitação , Interleucina-1/metabolismo , Quinases Associadas a Receptores de Interleucina-1 , Lipopolissacarídeos/química , Lipopolissacarídeos/metabolismo , Luciferases/metabolismo , Mutação , Fator 88 de Diferenciação Mieloide , NF-kappa B/metabolismo , Oligonucleotídeos/química , Ligação Proteica , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , Receptores Imunológicos/metabolismo , Receptores de Interleucina-1/genética , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo , Fatores de Tempo , Receptor 4 Toll-Like , Receptores Toll-Like , Transfecção
19.
J Mol Med (Berl) ; 83(4): 258-66, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15662540

RESUMO

Toll-like receptors (TLRs) belong to the Toll-interleukin 1 receptor superfamily, which is defined by a common intracellular Toll-IL-1 receptor (TIR) domain. A group of TIR domain containing adaptors (MyD88, TIRAP, TRIF and TRAM), are differentially recruited to the Toll-IL-1 receptors, contributing to the specificity of signaling. The IL-1 mediated signaling pathway serves as a "prototype" for other family members. Genetic and biochemical studies reveal that IL-1R uses adaptor molecule MyD88 to mediate a very complex pathway, involving a cascade of kinases organized by multiple adapter molecules into signaling complexes, leading to activation of the transcription factor NFkappaB. Several Toll-like receptors utilize variations of the "prototype" pathway by employing different adaptor molecules. Double-stranded RNA triggered, TLR3-mediated signaling is independent of MyD88, IRAK4, and IRAK. The adapter molecule TRIF is utilized by TLR3 to mediate the activation of NFkappaB and IRF3. LPS-induced, TLR4-mediated signaling employs multiple TIR-domain containing adaptors, MyD88/TIRAP to mediate NFkappaB activation, TRIF/TRAM for IRF3 activation. Recent studies have also begun to unravel how these pathways are negatively regulated. SIGIRR (also known as TIR8), a member of TIR superfamily that does not activate the transcription factors NFkappaB and IRF3, instead negatively modulates responses. Cells from SIGIRR-null mice show enhanced activation in response to either IL-1 or certain Toll ligands. In addition to SIGIRR, several other negative regulators have been shown to inhibit the TIR signaling, including ST2, IRAKM, MyD88s, SOCS1, and Triad3A. The coordinated positive and negative regulation of the TIR signaling ensures the appropriate modulation of the innate and inflammatory responses.


Assuntos
Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Humanos , Quinases Associadas a Receptores de Interleucina-1 , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fator 88 de Diferenciação Mieloide , Proteínas Quinases/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Proteínas Repressoras/metabolismo , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina , Receptor 3 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Fatores de Transcrição/genética
20.
Immunity ; 21(4): 575-87, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15485634

RESUMO

TNF receptor (TNFR) superfamily members, CD40, and BAFFR play critical roles in B cell survival and differentiation. Genetic deficiency in a novel adaptor molecule, Act1, for CD40 and BAFF results in a dramatic increase in peripheral B cells, which culminates in lymphadenopathy and splenomegaly, hypergammaglobulinemia, and autoantibodies. While the B cell-specific Act1 knockout mice displayed a similar phenotype with less severity, the pathology of the Act1-deficient mice was mostly blocked in CD40-Act1 and BAFF-Act1 double knockout mice. CD40- and BAFF-mediated survival is significantly increased in Act1-deficent B cells, with stronger IkappaB phosphorylation, processing of NF-kappaB2 (p100/p52), and activation of JNK, ERK, and p38 pathways, indicating that Act1 negatively regulates CD40- and BAFF-mediated signaling events. These findings demonstrate that Act1 plays an important role in the homeostasis of B cells by attenuating CD40 and BAFFR signaling.


Assuntos
Linfócitos B/imunologia , Antígenos CD40/imunologia , Proteínas de Transporte/metabolismo , Proteínas de Membrana/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Autoanticorpos/sangue , Fator Ativador de Células B , Receptor do Fator Ativador de Células B , Linfócitos B/metabolismo , Southern Blotting , Western Blotting , Antígenos CD40/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Sobrevivência Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Hipergamaglobulinemia/etiologia , Proteínas I-kappa B/imunologia , Proteínas I-kappa B/metabolismo , Imuno-Histoquímica , Imunoprecipitação , Proteínas Quinases JNK Ativadas por Mitógeno/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , MAP Quinase Quinase 4 , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Quinases de Proteína Quinase Ativadas por Mitógeno/imunologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA