Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(7)2023 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-37047469

RESUMO

Many crucial epigenetic changes occur during early skeletal development and throughout life due to aging, disease and are heavily influenced by an individual's lifestyle. Epigenetics is the study of heritable changes in gene expression as the result of changes in the environment without any mutation in the underlying DNA sequence. The epigenetic profiles of cells are dynamic and mediated by different mechanisms, including histone modifications, non-coding RNA-associated gene silencing and DNA methylation. Given the underlining role of dysfunctional mesenchymal tissues in common age-related skeletal diseases such as osteoporosis and osteoarthritis, investigations into skeletal stem cells or mesenchymal stem cells (MSC) and their functional deregulation during aging has been of great interest and how this is mediated by an evolving epigenetic landscape. The present review describes the recent findings in epigenetic changes of MSCs that effect growth and cell fate determination in the context of aging, diet, exercise and bone-related diseases.


Assuntos
Metilação de DNA , Células-Tronco Mesenquimais , Epigênese Genética , Diferenciação Celular/genética , Células-Tronco Mesenquimais/metabolismo
2.
Stem Cells Dev ; 32(13-14): 398-409, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37078151

RESUMO

The five flat bones of developing cranial plates are bounded by fibrous sutures, which remain open during development to accommodate for the growing brain. Kdm6A is a demethylase that removes the epigenetic repressive mark, trimethylated lysine 27 on histone 3 (H3K27me3), from the promoters of osteogenic genes, and has previously been reported to promote osteogenesis in cranial bone cells. This study generated a mesenchyme-specific deletion of a histone demethylase, Kdm6a, to assess the effects of Kdm6a loss, in cranial plate development and suture fusion. The results showed that the loss of Kdm6a in Prx1+ cranial cells caused increased anterior width and length in the calvaria of both male and female mice. However, the posterior length was further decreased in female mice. Moreover, loss of Kdm6a resulted in suppression of late suture development and calvarial frontal bone formation predominantly in female mice. In vitro assessment of calvaria cultures isolated from female Kdm6a knockout mice found significantly suppressed calvarial osteogenic differentiation potential, associated with decreased gene expression levels of Runx2 and Alkaline Phosphatase and increased levels of the suppressive mark, H3K27me3, on the respective gene promoters. Conversely, cultured calvaria bone cultures isolated from male Kdm6a knockout mice exhibited an increased osteogenic differentiation potential. Interestingly, the milder effects on cranial suture development in Kdm6a knockout male mice, were associated with an overcompensation of the Kdm6a Y-homolog, Kdm6c, and increased expression levels of Kdm6b in calvarial bone cultures. Taken together, these data demonstrate a role for Kdm6a during calvarial development and patterning, predominantly in female mice, and highlight the potential role of Kdm6 family members in patients with unexplained craniofacial deformities.


Assuntos
Suturas Cranianas , Osso Frontal , Animais , Feminino , Masculino , Camundongos , Suturas Cranianas/metabolismo , Osso Frontal/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Histonas/metabolismo , Camundongos Knockout , Osteogênese/genética , Fatores Sexuais
3.
Leukemia ; 36(11): 2678-2689, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36038666

RESUMO

Therapy-related myeloid neoplasm (tMN) is considered a direct consequence of DNA damage in hematopoietic stem cells. Despite increasing recognition that altered stroma can also drive leukemogenesis, the functional biology of the tMN microenvironment remains unknown. We performed multiomic (transcriptome, DNA damage response, cytokine secretome and functional profiling) characterization of bone marrow stromal cells from tMN patients. Critically, we also compared (i) patients with myeloid neoplasm and another cancer but without cytotoxic exposure, (ii) typical primary myeloid neoplasm, and (iii) age-matched controls to decipher the microenvironmental changes induced by cytotoxics vs. neoplasia. Strikingly, tMN exhibited a profoundly senescent phenotype with induction of CDKN1A and ß-Galactosidase, defective phenotype, and proliferation. Moreover, tMN stroma showed delayed DNA repair and defective adipogenesis. Despite their dormant state, tMN stromal cells were metabolically highly active with a switch toward glycolysis and secreted multiple pro-inflammatory cytokines indicative of a senescent-secretory phenotype that inhibited adipogenesis. Critically, senolytics not only eliminated dormant cells, but also restored adipogenesis. Finally, sequential patient sampling showed senescence phenotypes are induced within months of cytotoxic exposure, well prior to the onset of secondary cancer. Our data underscores a role of senescence in the pathogenesis of tMN and provide a valuable resource for future therapeutics.


Assuntos
Antineoplásicos , Células-Tronco Mesenquimais , Neoplasias , Humanos , Senescência Celular/genética , Secretoma , Células-Tronco Mesenquimais/metabolismo , Antineoplásicos/farmacologia , Citocinas/metabolismo , Microambiente Tumoral
4.
Stem Cell Res Ther ; 11(1): 529, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33298158

RESUMO

BACKGROUND: During development, excessive osteogenic differentiation of mesenchymal progenitor cells (MPC) within the cranial sutures can lead to premature suture fusion or craniosynostosis, leading to craniofacial and cognitive issues. Saethre-Chotzen syndrome (SCS) is a common form of craniosynostosis, caused by TWIST-1 gene mutations. Currently, the only treatment option for craniosynostosis involves multiple invasive cranial surgeries, which can lead to serious complications. METHODS: The present study utilized Twist-1 haploinsufficient (Twist-1del/+) mice as SCS mouse model to investigate the inhibition of Kdm6a and Kdm6b activity using the pharmacological inhibitor, GSK-J4, on calvarial cell osteogenic potential. RESULTS: This study showed that the histone methyltransferase EZH2, an osteogenesis inhibitor, is downregulated in calvarial cells derived from Twist-1del/+ mice, whereas the counter histone demethylases, Kdm6a and Kdm6b, known promoters of osteogenesis, were upregulated. In vitro studies confirmed that siRNA-mediated inhibition of Kdm6a and Kdm6b expression suppressed osteogenic differentiation of Twist-1del/+ calvarial cells. Moreover, pharmacological targeting of Kdm6a and Kdm6b activity, with the inhibitor, GSK-J4, caused a dose-dependent suppression of osteogenic differentiation by Twist-1del/+ calvarial cells in vitro and reduced mineralized bone formation in Twist-1del/+ calvarial explant cultures. Chromatin immunoprecipitation and Western blot analyses found that GSK-J4 treatment elevated the levels of the Kdm6a and Kdm6b epigenetic target, the repressive mark of tri-methylated lysine 27 on histone 3, on osteogenic genes leading to repression of Runx2 and Alkaline Phosphatase expression. Pre-clinical in vivo studies showed that local administration of GSK-J4 to the calvaria of Twist-1del/+ mice prevented premature suture fusion and kept the sutures open up to postnatal day 20. CONCLUSION: The inhibition of Kdm6a and Kdm6b activity by GSK-J4 could be used as a potential non-invasive therapeutic strategy for preventing craniosynostosis in children with SCS. Pharmacological targeting of Kdm6a/b activity can alleviate craniosynostosis in Saethre-Chotzen syndrome. Aberrant osteogenesis by Twist-1 mutant cranial suture mesenchymal progenitor cells occurs via deregulation of epigenetic modifiers Ezh2 and Kdm6a/Kdm6b. Suppression of Kdm6a- and Kdm6b-mediated osteogenesis with GSK-J4 inhibitor can prevent prefusion of cranial sutures.


Assuntos
Acrocefalossindactilia , Acrocefalossindactilia/genética , Acrocefalossindactilia/terapia , Animais , Histona Desmetilases , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Terapia de Alvo Molecular , Proteínas Nucleares/genética , Osteogênese , Proteína 1 Relacionada a Twist/genética
5.
Curr Osteoporos Rep ; 18(5): 597-605, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32794139

RESUMO

PURPOSE OF REVIEW: Although many signalling pathways have been discovered to be essential in mesenchymal stem/stromal (MSC) differentiation, it has become increasingly clear in recent years that epigenetic regulation of gene transcription is a vital component of lineage determination, encompassing diet, lifestyle and parental influences on bone, fat and cartilage development. RECENT FINDINGS: This review discusses how specific enzymes that modify histone methylation and acetylation or DNA methylation orchestrate the differentiation programs in lineage determination of MSC and the epigenetic changes that facilitate development of bone related diseases such as osteoporosis. The review also describes how environmental factors such as mechanical loading influence the epigenetic signatures of MSC, and how the use of chemical agents or small peptides can regulate epigenetic drift in MSC populations during ageing and disease. Epigenetic regulation of MSC lineage commitment is controlled through changes in enzyme activity, which modifies DNA and histone residues leading to alterations in chromatin structure. The co-ordinated epigenetic regulation of transcriptional activation and repression act to mediate skeletal tissue homeostasis, where deregulation of this process can lead to bone loss during ageing or osteoporosis.


Assuntos
Envelhecimento/genética , Diferenciação Celular/genética , Epigênese Genética/genética , Células-Tronco Mesenquimais/citologia , Osteoporose/genética , Acetilação , Linhagem da Célula , Metilação de DNA , Repressão Epigenética/genética , Regulação da Expressão Gênica/genética , Código das Histonas , Humanos , Metilação , Ativação Transcricional/genética , Suporte de Carga
6.
Bone ; 137: 115440, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32445894

RESUMO

There is mounting evidence in the literature that mesenchymal stromal/stem cell (MSC) like populations derived from different tissues, undergo epigenetic changes during aging, leading to compromised connective tissue integrity and function. This body of work has linked the biological aging of MSC to changes in their epigenetic signatures affecting growth, lifespan, self-renewal and multi-potential, due to deregulation of processes such as cellular senescence, oxidative stress, DNA damage, telomere shortening and DNA damage. This review addresses recent findings examining DNA methylation, histone modifications and miRNA changes in aging MSC populations. Moreover, we explore how epigenetic factors alter cellular pathways and associated biological networks, contributing to the MSC aging phenotype. Finally we discuss the crucial areas requiring a greater understanding of these processes, in order to piece together a global picture of the changing epigenetic landscape in MSC during aging.


Assuntos
Células-Tronco Mesenquimais , Senescência Celular/genética , Epigênese Genética , Epigenômica
7.
Aging Dis ; 10(1): 174-189, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30705777

RESUMO

In this review we explore the importance of epigenetics as a contributing factor for aging adult stem cells. We summarize the latest findings of epigenetic factors deregulated as adult stem cells age and the consequence on stem cell self-renewal and differentiation, with a focus on adult stem cells in the bone marrow. With the latest whole genome bisulphite sequencing and chromatin immunoprecipitations we are able to decipher an emerging pattern common for adult stem cells in the bone marrow niche and how this might correlate to epigenetic enzymes deregulated during aging. We begin by briefly discussing the initial observations in yeast, drosophila and Caenorhabditis elegans (C. elegans) that led to the breakthrough research that identified the role of epigenetic changes associated with lifespan and aging. We then focus on adult stem cells, specifically in the bone marrow, which lends strong support for the deregulation of DNA methyltransferases, histone deacetylases, acetylates, methyltransferases and demethylases in aging stem cells, and how their corresponding epigenetic modifications influence gene expression and the aging phenotype. Given the reversible nature of epigenetic modifications we envisage "epi" targeted therapy as a means to reprogram aged stem cells into their younger counterparts.

8.
Epigenetics Chromatin ; 12(1): 3, 2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30606231

RESUMO

BACKGROUND: The 5 hydroxymethylation (5hmC) mark and TET DNA dioxygenases play a pivotal role in embryonic stem cell differentiation and animal development. However, very little is known about TET enzymes in lineage determination of human bone marrow-derived mesenchymal stem/stromal cells (BMSC). We examined the function of all three TET DNA dioxygenases, responsible for DNA hydroxymethylation, in human BMSC cell osteogenic and adipogenic differentiation. RESULTS: We used siRNA knockdown and retroviral mediated enforced expression of TET molecules and discovered TET1 to be a repressor of both osteogenesis and adipogenesis. TET1 was found to recruit the co-repressor proteins, SIN3A and the histone lysine methyltransferase, EZH2 to osteogenic genes. Conversely, TET2 was found to be a promoter of both osteogenesis and adipogenesis. The data showed that TET2 was directly responsible for 5hmC levels on osteogenic and adipogenic lineage-associated genes, whereas TET1 also played a role in this process. Interestingly, TET3 showed no functional effect in BMSC osteo-/adipogenic differentiation. Finally, in a mouse model of ovariectomy-induced osteoporosis, the numbers of clonogenic BMSC were dramatically diminished corresponding to lower trabecular bone volume and reduced levels of TET1, TET2 and 5hmC. CONCLUSION: The present study has discovered an epigenetic mechanism mediated through changes in DNA hydroxymethylation status regulating the activation of key genes involved in the lineage determination of skeletal stem cells, which may have implications in BMSC function during normal bone regulation. Targeting TET molecules or their downstream targets may offer new therapeutic strategies to help prevent bone loss and repair following trauma or disease.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Mesenquimais/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Adipogenia , Animais , Células Cultivadas , Metilação de DNA , Proteínas de Ligação a DNA/genética , Dioxigenases , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Oxigenases de Função Mista/genética , Osteogênese , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/metabolismo , Complexo Correpressor Histona Desacetilase e Sin3
9.
FASEB J ; 31(3): 1011-1027, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27934660

RESUMO

In this study, we examined the functional importance of EZH2 during skeletal development and homeostasis using the conditional deletion of Ezh2 (Ezh2fl/fl ) in early mesenchyme with the use of a Prrx-1-cre driver mouse (Ezh2+/+). Heterozygous (Ezh2+/-) newborn and 4-wk-old mice exhibited increased skeletal size, growth plate size, and weight when compared to the wild-type control (Ezh2+/+), whereas homozygous deletion of Ezh2 (Ezh2-/-) resulted in skeletal deformities and reduced skeletal size, growth plate size, and weight in newborn and 4-wk-old mice. Ezh2-/- mice exhibited enhanced trabecular patterning. Osteogenic cortical and trabecular bone formation was enhanced in Ezh2+/- and Ezh2-/- animals. Ezh2+/- and Ezh2-/- mice displayed thinner cortical bone and decreased mechanical strength compared to the wild-type control. Differences in cortical bone thickness were attributed to an increased number of osteoclasts, corresponding with elevated levels of the bone turnover markers cross-linked C-telopeptide-1 and tartrate-resistant acid phosphatase, detected within serum. Moreover, Ezh2+/- mice displayed increased osteoclastogenic potential coinciding with an upregulation of Rankl and M-csf expression by mesenchymal stem cells (MSCs). MSCs isolated from Ezh2+/- mice also exhibited increased trilineage potential compared with wild-type bone marrow stromal/stem cells (BMSCs). Gene expression studies confirmed the upregulation of known Ezh2 target genes in Ezh2-/- bone tissue, many of which are involved in Wnt/BMP signaling as promoters of osteogenesis and inhibitors of adipogenesis. In summary, EZH2 appears to be an important orchestrator of skeletal development, postnatal bone remodelling and BMSC fate determination in vitro and in vivo-Hemming, S., Cakouros, D., Codrington, J., Vandyke, K., Arthur, A., Zannettino, A., Gronthos, S. EZH2 deletion in early mesenchyme compromises postnatal bone microarchitecture and structural integrity and accelerates remodeling.


Assuntos
Remodelação Óssea , Osso Esponjoso/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Mesoderma/metabolismo , Osteogênese , Animais , Osso Esponjoso/citologia , Osso Esponjoso/crescimento & desenvolvimento , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Deleção de Genes , Heterozigoto , Homozigoto , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/citologia , Osteoclastos/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Via de Sinalização Wnt
10.
Stem Cells Dev ; 25(12): 909-21, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27168161

RESUMO

Histone three lysine 27 (H3K27) methyltransferase enhancer of zeste homolog 2 (EZH2) is a critical epigenetic modifier, which regulates gene transcription through the trimethylation of the H3K27 residue leading to chromatin compaction and gene repression. EZH2 has previously been identified to regulate human bone marrow-derived mesenchymal stem cells (MSC) lineage specification. MSC lineage specification is regulated by the presence of EZH2 and its H3K27me3 modification or the removal of the H3K27 modification by lysine demethylases 6A and 6B (KDM6A and KDM6B). This study used a bioinformatics approach to identify novel genes regulated by EZH2 during MSC osteogenic differentiation. In this study, we identified the EZH2 targets, ZBTB16, MX1, and FHL1, which were expressed at low levels in MSC. EZH2 and H3K27me3 were found to be present along the transcription start site of their respective promoters. During osteogenesis, these genes become actively expressed coinciding with the disappearance of EZH2 and H3K27me3 on the transcription start site of these genes and the enrichment of the active H3K4me3 modification. Overexpression of EZH2 downregulated the transcript levels of ZBTB16, MX1, and FHL1 during osteogenesis. Small interfering RNA targeting of MX1 and FHL1 was associated with a downregulation of the key osteogenic transcription factor, RUNX2, and its downstream targets osteopontin and osteocalcin. These findings highlight that EZH2 not only acts through the direct regulation of signaling modules and lineage-specific transcription factors but also targets many novel genes important for mediating MSC osteogenic differentiation.


Assuntos
Diferenciação Celular , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Adulto , Diferenciação Celular/genética , Imunoprecipitação da Cromatina , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Lisina/metabolismo , Metilação , Proteínas Musculares/metabolismo , Proteínas de Resistência a Myxovirus/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/metabolismo , Regulação para Cima/genética
11.
Stem Cells ; 34(2): 504-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718114

RESUMO

Twist-1 encodes a basic helix-loop-helix transcription factor, known to contribute to mesodermal and skeletal tissue development. We have reported previously that Twist-1 maintains multipotent human bone marrow-derived mesenchymal stem/stromal cells (BMSC) in an immature state, enhances their life-span, and influences cell fate determination. In this study, human BMSC engineered to express high levels of Twist-1 were found to express elevated levels of the chemokine, CXCL12. Analysis of the CXCL12 proximal promoter using chromatin immunoprecipitation analysis identified several E-box DNA sites bound by Twist-1. Functional studies using a luciferase reporter construct showed that Twist-1 increased CXCL12 promoter activity in a dose dependent manner. Notably, Twist-1 over-expressing BMSC exhibited an enhanced capacity to maintain human CD34 + hematopoietic stem cells (HSC) in long-term culture-initiating cell (LTC-IC) assays. Moreover, the observed increase in HSC maintenance by Twist-1 over-expressing BMSC was blocked in the presence of the CXCL12 inhibitor, AMD3100. Supportive studies, using Twist-1 deficient heterozygous mice demonstrated a significant decrease in the frequency of stromal progenitors and increased numbers of osteoblasts within the bone. These observations correlated to a decreased incidence in the number of clonogenic stromal progenitors (colony forming unit-fibroblasts) and lower levels of CXCL12 in Twist-1 mutant mice. Furthermore, Twist-1 deficient murine stromal feeder layers, exhibited a significant decrease in CXCL12 levels and lower numbers of hematopoietic colonies in LTC-IC assays, compared with wild type controls. These findings demonstrate that Twist-1, which maintains BMSC at an immature state, endows them with an increased capacity for supporting hematopoiesis via direct activation of CXCL12 gene expression.


Assuntos
Células da Medula Óssea/metabolismo , Quimiocina CXCL12/biossíntese , Regulação da Expressão Gênica , Hematopoese , Células-Tronco Mesenquimais/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Células da Medula Óssea/citologia , Quimiocina CXCL12/genética , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Proteínas Nucleares/genética , Proteína 1 Relacionada a Twist/genética
12.
Stem Cells Dev ; 24(11): 1297-308, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25579220

RESUMO

Basic helix-loop-helix (bHLH) transcription factors are pivotal regulators of cellular differentiation and development. The bHLH factor, Twist-1 has previously been found to control bone marrow stromal/stem cells (BMSC) self-renewal, life span, and differentiation, however not much is known about its mechanism of action. In this study, we have discovered a novel Twist-1 regulated bHLH gene, Hes4, expressed in humans, but not in mice. Its closest homologue in both humans and mice is Hes1. Overexpression and knockdown studies demonstrated that Hes4 promotes osteogenesis resulting in an increase in Runx2, osteocalcin, osteopontin, and bone sialoprotein expression. Conversely, Hes4 was found to inhibit adipogenesis accompanied by a decrease in PPARγ2, adiponectin, and adipsin expression. In vitro studies indicate that Hes4 employs a mechanism to counteract the negative function of Twist-1 on osteogenesis by binding to Twist-1 and inhibiting the ability of Twist-1 to bind and inhibit Runx2. In vivo chromatin immunoprecipitation and in vitro reporter assays illustrated that Runx2 recruitment to the osterix promoter, was found to be enhanced in the presence of Hes4 and inhibited in the presence of Twist-1. Therefore, Hes4 antagonizes the function of Twist-1 to regulate lineage commitment of BMSC. These studies highlight the potential differences in molecular mechanisms that regulate BMSC osteogenic differentiation between human and mouse.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem da Célula , Células-Tronco Mesenquimais/metabolismo , Proteínas Nucleares/metabolismo , Osteoblastos/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Adipogenia , Adiponectina/genética , Adiponectina/metabolismo , Sequência de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Fator D do Complemento/genética , Fator D do Complemento/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Dados de Sequência Molecular , Proteínas Nucleares/genética , Osteoblastos/citologia , Osteocalcina/genética , Osteocalcina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Especificidade da Espécie , Proteína 1 Relacionada a Twist/genética
14.
Stem Cells ; 32(3): 802-15, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24123378

RESUMO

The methyltransferase, Enhancer of Zeste homology 2 (EZH2), trimethylates histone 3 lysine 27 (H3K27me3) on chromatin and this repressive mark is removed by lysine demethylase 6A (KDM6A). Loss of these epigenetic modifiers results in developmental defects. We demonstrate that Ezh2 and Kdm6a transcript levels change during differentiation of multipotential human bone marrow-derived mesenchymal stem cells (MSC). Enforced expression of Ezh2 in MSC promoted adipogenic in vitro and inhibited osteogenic differentiation potential in vitro and in vivo, whereas Kdm6a inhibited adipogenesis in vitro and promoted osteogenic differentiation in vitro and in vivo. Inhibition of EZH2 activity and knockdown of Ezh2 gene expression in human MSC resulted in decreased adipogenesis and increased osteogenesis. Conversely, knockdown of Kdm6a gene expression in MSC leads to increased adipogenesis and decreased osteogenesis. Both Ezh2 and Kdm6a were shown to affect expression of master regulatory genes involved in adipogenesis and osteogenesis and H3K27me3 on the promoters of master regulatory genes. These findings demonstrate an important epigenetic switch centered on H3K27me3 which dictates MSC lineage determination.


Assuntos
Linhagem da Célula/genética , Epigênese Genética , Histona Desmetilases/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/genética , Adolescente , Adulto , Animais , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Histona Desmetilases/genética , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Camundongos , Camundongos SCID , Proteínas Nucleares/genética , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , Complexo Repressor Polycomb 2/genética , Regiões Promotoras Genéticas/genética , Transcrição Gênica , Adulto Jovem
15.
Nat Commun ; 4: 2916, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24336022

RESUMO

Correct spatial and temporal induction of numerous cell type-specific genes during development requires regulated removal of the repressive histone H3 lysine 27 trimethylation (H3K27me3) modification. Here we show that the H3K27me3 demethylase dUTX is required for hormone-mediated transcriptional regulation of apoptosis and autophagy genes during ecdysone-regulated programmed cell death of Drosophila salivary glands. We demonstrate that dUTX binds to the nuclear hormone receptor complex Ecdysone Receptor/Ultraspiracle, and is recruited to the promoters of key apoptosis and autophagy genes. Salivary gland cell death is delayed in dUTX mutants, with reduced caspase activity and autophagy that coincides with decreased apoptosis and autophagy gene transcripts. We further show that salivary gland degradation requires dUTX catalytic activity. Our findings provide evidence for an unanticipated role for UTX demethylase activity in regulating hormone-dependent cell death and demonstrate how a single transcriptional regulator can modulate a specific complex functional outcome during animal development.


Assuntos
Autofagia/genética , Proteínas de Drosophila/metabolismo , Ecdisona/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Glândulas Salivares/citologia , Animais , Apoptose/genética , Caspases/genética , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila/citologia , Drosophila/genética , Proteínas de Drosophila/genética , Ecdisona/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Histonas/metabolismo , Larva , Metilação , Mutação , Oxirredutases N-Desmetilantes/genética , Regiões Promotoras Genéticas , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Glândulas Salivares/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Mol Cell Biol ; 32(8): 1433-41, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22290439

RESUMO

The main impairment to tissue maintenance during aging is the reduced capacity for stem cell self-renewal over time due to senescence, the irreversible block in proliferation. We have previously described that the basic helix-loop-helix (bHLH) transcription factor Twist-1 can greatly enhance the life span of bone marrow-derived mesenchymal stem/stromal cells (MSCs). In the present study, we show that Twist-1 potently suppresses senescence and the Ink4A/Arf locus with a dramatic decrease in the expression of p16 and to some extent a decrease in p14. Furthermore, the polycomb group protein and histone methyltransferase Ezh2, which suppresses the Ink4A/Arf locus, was found to be induced by Twist-1, resulting in an increase in H3K27me3 along the Ink4A/Arf locus, repressing transcription of both p16/p14 and senescence of human MSCs. Furthermore, Twist-1 inhibits the expression of the bHLH transcription factor E47, which is normally expressed in senescent MSCs and induces transcription of the p16 promoter. Reduced Twist-1 wild-type expression and function in bone cells derived from Saethre-Chotzen patients also revealed an increase in senescence. These studies for the first time link Twist-1 to histone methylation of the Ink4A/Arf locus by controlling the expression of histone methyltransferases as well as the expression of other bHLH factors.


Assuntos
Senescência Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Mesenquimais , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Adulto , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética , Regulação da Expressão Gênica , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Metilação , Proteínas Nucleares/genética , Complexo Repressor Polycomb 2 , Fatores de Transcrição/genética , Proteína 1 Relacionada a Twist/genética
17.
Proc Natl Acad Sci U S A ; 106(13): 5336-41, 2009 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-19279217

RESUMO

Apoptosis is mediated by the caspase family of proteases that act as effectors of cell death by cleaving many cellular substrates. Caspase-2 is one of the most evolutionarily conserved caspases, yet its physiological function has remained enigmatic because caspase-2-deficient mice develop normally and are viable. We report here that the caspase-2(-/-) mouse embryonic fibroblasts (MEFs) show increased proliferation. When transformed with E1A and Ras oncogenes, caspase-2(-/-) MEFs grew significantly faster than caspase-2(+/+) MEFs and formed more aggressive and accelerated tumors in nude mice. To assess whether the loss of caspase-2 predisposes animals to tumor development, we used the mouse Emu-Myc lymphoma model. Our findings suggest that loss of even a single allele of caspase-2 resulted in accelerated tumorigenesis, and this was further enhanced in caspase-2(-/-) mice. The caspase-2(-/-) cells showed resistance to apoptosis induced by chemotherapeutic drugs and DNA damage. Furthermore, caspase-2(-/-) MEFs had a defective apoptotic response to cell-cycle checkpoint regulation and showed abnormal cycling following gamma-irradiation. These data show that loss of caspase-2 results in an increased ability of cells to acquire a transformed phenotype and become malignant, indicating that caspase-2 is a tumor suppressor protein.


Assuntos
Caspase 2/deficiência , Caspase 2/fisiologia , Proteínas Supressoras de Tumor , Animais , Apoptose , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Modelos Animais de Doenças , Linfoma/etiologia , Camundongos , Camundongos Knockout , Camundongos Nus
18.
J Cell Biol ; 182(3): 481-95, 2008 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-18695041

RESUMO

The sequential modifications of histones form the basis of the histone code that translates into either gene activation or repression. Nuclear receptors recruit a cohort of histone-modifying enzymes in response to ligand binding and regulate proliferation, differentiation, and cell death. In Drosophila melanogaster, the steroid hormone ecdysone binds its heterodimeric receptor ecdysone receptor/ultraspiracle to spatiotemporally regulate the transcription of several genes. In this study, we identify a novel cofactor, Drosophila lysine ketoglutarate reductase (dLKR)/saccharopine dehydrogenase (SDH), that is involved in ecdysone-mediated transcription. dLKR/SDH binds histones H3 and H4 and suppresses ecdysone-mediated transcription of cell death genes by inhibiting histone H3R17me2 mediated by the Drosophila arginine methyl transferase CARMER. Our data suggest that the dynamic recruitment of dLKR/SDH to ecdysone-regulated gene promoters controls the timing of hormone-induced gene expression. In the absence of dLKR/SDH, histone methylation occurs prematurely, resulting in enhanced gene activation. Consistent with these observations, the loss of dLKR/SDH in Drosophila enhances hormone-regulated gene expression, affecting the developmental timing of gene activation.


Assuntos
Arginina/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Ecdisona/farmacologia , Histonas/metabolismo , Sacaropina Desidrogenases/metabolismo , Transcrição Gênica/efeitos dos fármacos , Animais , Sítios de Ligação , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes de Insetos , Cinética , Metilação/efeitos dos fármacos , Modelos Genéticos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Interferência de RNA , Receptores de Esteroides/metabolismo , Ativação Transcricional
19.
J Bone Miner Res ; 22(6): 897-906, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17352650

RESUMO

UNLABELLED: Human BMSSCs lose telomerase activity in vitro, which leads to chromosomal instability and cellular senescence. We observed an inverse expression pattern between the osteogenic master regulatory gene, CBFA1, and the stem cell-associated gene, hTERT. We showed that Cbfa1 acts as a partial repressor of TERT, which may facilitate cellular differentiation. INTRODUCTION: The absence of telomerase activity by cultured human bone marrow stromal stem cells (BMSSCs) causes critical shortening of chromosomal telomeres, leading eventually to cellular senescence. Ex vivo expansion of BMSSCs correlates to an increase in osteogenic lineage associated markers such as alkaline phosphatase, bone sialoprotein, and osteocalcin that are regulated by the master regulatory transcription factor, Cbfa1 (Runx2). This study examined whether Cbfa1 was capable of regulating the promoter of the early stem cell-associated gene, telomerase reverse transcriptase (TERT). MATERIALS AND METHODS: Human BMSSCs were isolated by fluorescence-activated cell sorting. Telomerase activity was determined using the telometric repeat amplification protocol. CBFA1 and TERT gene expression was assessed by real-time PCR. The functional capacity of Cbfa1 to bind to the hTERT promoter was performed using a modified electrophoretic mobility shift assay (EMSA). Chromatin immunoprecipitation (ChIP) analysis was used to examine Cbfa1 binding to the hTERT promoter in vivo. Functional analysis of CBFA-1 wildtype and mutant DNA binding sites on TERT promoter fragments was assessed using the promoterless green fluorescence protein (GFP) reporter vector, pEGFP-1, after transfection into HOS cells. RESULTS: This study showed an inverse expression pattern between the osteogenic master regulatory gene, CBFA1, and the stem cell-associated gene, hTERT. The data showed that BMSSCs undergo osteogenic commitment after the loss of hTERT expression, with concomitant elevated levels of CBFA1 transcripts. In addition, two unique Cbfa1 DNA binding sites were identified on the hTERT proximal promoter by EMSA supershift assay. Mutated forms of the putative Cbfa1 binding sites, created by site-directed mutagenesis, were able to abolish this interaction. ChIP analysis showed that Cbfa1 interacted directly with the hTERT promoter in vivo. Functional studies using GFP reporter constructs, driven by 2- and 3-kbp hTERT proximal promoter fragments, showed significantly lower levels of transcriptional activity compared with corresponding constructs with mutated Cbfa1 binding site Oligo 2. CONCLUSIONS: These studies suggest that Cbfa1 may act as a repressor of early stem cell markers such as hTERT as one possible mechanism for facilitating cellular differentiation.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Telomerase/genética , Adulto , Fosfatase Alcalina/metabolismo , Antígenos de Superfície/metabolismo , Becaplermina , Sítios de Ligação , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Imunoprecipitação da Cromatina , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Meios de Cultura Livres de Soro/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Mutação , Oligodesoxirribonucleotídeos/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-sis , Telomerase/metabolismo , Transfecção , Fator de Crescimento Transformador beta1/farmacologia
20.
J Biol Chem ; 280(12): 11981-6, 2005 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-15657059

RESUMO

The Drosophila steroid hormone ecdysone mediates cell death during metamorphosis by regulating the transcription of a number of cell death genes. The apical caspase DRONC is known to be transcriptionally regulated by ecdysone during development. Here we demonstrate that ecdysone also regulates the transcription of DRICE, a major effector caspase and a downstream target for DRONC in the fly. Using RNA interference in an ecdysone-responsive Drosophila cell line, we show that drice up-regulation is essential for apoptosis induced by ecdysone. We also show that drice expression is specifically controlled by the ecdysone-regulated transcription factor BR-C. Combined with previous observations, our results indicate that transcriptional regulation of the components of the core apoptotic machinery plays a key role in hormone-regulated programmed cell death during Drosophila development.


Assuntos
Apoptose , Caspases/genética , Proteínas de Drosophila/genética , Ecdisona/farmacologia , Animais , Células Cultivadas , Drosophila , Proteínas de Drosophila/fisiologia , Ativação Enzimática , RNA Mensageiro/análise , Glândulas Salivares/metabolismo , Fatores de Transcrição/fisiologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...