Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Cell Physiol ; 233(9): 7320-7332, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29663378

RESUMO

Saethre-Chotzen syndrome (SCS), associated with TWIST-1 mutations, is characterized by premature fusion of cranial sutures. TWIST-1 haploinsufficiency, leads to alterations in suture mesenchyme cellular gene expression patterns, resulting in aberrant osteogenesis and craniosynostosis. We analyzed the expression of the TWIST-1 target, Tyrosine kinase receptor c-ros-oncogene 1 (C-ROS-1) in TWIST-1 haploinsufficient calvarial cells derived from SCS patients and calvaria of Twist-1del/+ mutant mice and found it to be highly expressed when compared to TWIST-1 wild-type controls. Knock-down of C-ROS-1 expression in TWIST-1 haploinsufficient calvarial cells derived from SCS patients was associated with decreased capacity for osteogenic differentiation in vitro. Furthermore, treatment of human SCS calvarial cells with the tyrosine kinase chemical inhibitor, Crizotinib, resulted in reduced C-ROS-1 activity and the osteogenic potential of human SCS calvarial cells with minor effects on cell viability or proliferation. Cultured human SCS calvarial cells treated with Crizotinib exhibited a dose-dependent decrease in alkaline phosphatase activity and mineral deposition, with an associated decrease in expression levels of Runt-related transcription factor 2 and OSTEOPONTIN, with reduced PI3K/Akt signalling in vitro. Furthermore, Crizotinib treatment resulted in reduced BMP-2 mediated bone formation potential of whole Twist-1del/+ mutant mouse calvaria organotypic cultures. Collectively, these results suggest that C-ROS-1 promotes osteogenic differentiation of TWIST-1 haploinsufficient calvarial osteogenic progenitor cells. Furthermore, the aberrant osteogenic potential of these cells is inhibited by the reduction of C-ROS-1. Therefore, targeting C-ROS-1 with a pharmacological agent, such as Crizotinib, may serve as a novel therapeutic strategy to alleviate craniosynostosis associated with aberrant TWIST-1 function.


Assuntos
Acrocefalossindactilia/genética , Acrocefalossindactilia/patologia , Haploinsuficiência/genética , Osteogênese , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Crânio/patologia , Proteína 1 Relacionada a Twist/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Suturas Cranianas/patologia , Crizotinibe/farmacologia , Heterozigoto , Humanos , Camundongos , Mutação/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
2.
J Cell Physiol ; 232(2): 402-416, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27206577

RESUMO

Neural crest cells (NCC) hold great promise for tissue engineering, however the inability to easily obtain large numbers of NCC is a major factor limiting their use in studies of regenerative medicine. Induced pluripotent stem cells (iPSC) are emerging as a novel candidate that could provide an unlimited source of NCC. In the present study, we examined the potential of neural crest tissue-derived periodontal ligament (PDL) iPSC to differentiate into neural crest-like cells (NCLC) relative to iPSC generated from a non-neural crest derived tissue, foreskin fibroblasts (FF). We detected high HNK1 expression during the differentiation of PDL and FF iPSC into NCLC as a marker for enriching for a population of cells with NCC characteristics. We isolated PDL iPSC- and FF iPSC-derived NCLC, which highly expressed HNK1. A high proportion of the HNK1-positive cell populations generated, expressed the MSC markers, whilst very few cells expressed the pluripotency markers or the hematopoietic markers. The PDL and FF HNK1-positive populations gave rise to smooth muscle, neural, glial, osteoblastic and adipocytic like cells and exhibited higher expression of smooth muscle, neural, and glial cell-associated markers than the PDL and FF HNK1-negative populations. Interestingly, the HNK1-positive cells derived from the PDL-iPSC exhibited a greater ability to differentiate into smooth muscle, neural, glial cells and adipocytes, than the HNK1-positive cells derived from the FF-iPSC. Our work suggests that HNK1-enriched NCLC from neural crest tissue-derived iPSC more closely resemble the phenotypic and functional hallmarks of NCC compared to the HNK1-low population and non-neural crest iPSC-derived NCLC. J. Cell. Physiol. 232: 402-416, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Crista Neural/citologia , Ligamento Periodontal/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Membrana Celular/metabolismo , Forma Celular , Células Cultivadas , Fibroblastos/citologia , Humanos , Masculino , Mesoderma/citologia , Camundongos
3.
Proc Natl Acad Sci U S A ; 111(20): 7337-42, 2014 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-24808138

RESUMO

In vertebrate embryos, cardiac progenitor cells (CPCs) undergo long-range migration after emerging from the primitive streak during gastrulation. Together with other mesoderm progenitors, they migrate laterally and then toward the ventral midline, where they form the heart. Signals controlling the migration of different progenitor cell populations during gastrulation are poorly understood. Several pathways are involved in the epithelial-to-mesenchymal transition and ingression of mesoderm cells through the primitive streak, including fibroblast growth factors and wingless-type family members (Wnt). Here we focus on early CPC migration and use live video microscopy in chicken embryos to demonstrate a role for bone morphogenetic protein (BMP)/SMA and MAD related (Smad) signaling. We identify an interaction of BMP and Wnt/glycogen synthase kinase 3 beta (GSK3ß) pathways via the differential phosphorylation of Smad1. Increased BMP2 activity altered migration trajectories of prospective cardiac cells and resulted in their lateral displacement and ectopic differentiation, as they failed to reach the ventral midline. Constitutively active BMP receptors or constitutively active Smad1 mimicked this phenotype, suggesting a cell autonomous response. Expression of GSK3ß, which promotes the turnover of active Smad1, rescued the BMP-induced migration phenotype. Conversely, expression of GSK3ß-resistant Smad1 resulted in aberrant CPC migration trajectories. De-repression of GSK3ß by dominant negative Wnt3a restored normal migration patterns in the presence of high BMP activity. The data indicate the convergence of BMP and Wnt pathways on Smad1 during the early migration of prospective cardiac cells. Overall, we reveal molecular mechanisms that contribute to the emerging paradigm of signaling pathway integration in embryo development.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Miocárdio/citologia , Miocárdio/metabolismo , Proteína Smad1/fisiologia , Células-Tronco/citologia , Proteína Wnt3A/metabolismo , Animais , Padronização Corporal , Diferenciação Celular , Movimento Celular , Embrião de Galinha , Genes Dominantes , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Coração/embriologia , Mesoderma/metabolismo , Fenótipo , Linha Primitiva/metabolismo , Transdução de Sinais
4.
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
5.
Stem Cells ; 28(9): 1457-64, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20578184

RESUMO

Gonadal development in vertebrates depends on the early determination of primordial germ cells (PGCs) and their correct migration to the sites where the gonads develop. Several genes have been implicated in PGC specification and migration in vertebrates. Additionally, some of the genes associated with pluripotency, such as Oct4 and Nanog, are expressed in PGCs and gonads, suggesting a role for these genes in maintaining pluripotency of the germ lineage, which may be considered the only cell type that perpetually maintains stemness properties. Here, we report that medaka Nanog (Ol-Nanog) is expressed in the developing PGCs. Depletion of Ol-Nanog protein causes aberrant migration of PGCs and inhibits expression of Cxcr4b in PGCs, where it normally serves as the receptor of Sdf1a to guide PGC migration. Moreover, chromatin immunoprecipitation analysis demonstrates that Ol-Nanog protein binds to the promoter region of Cxcr4b, suggesting a direct regulation of Cxcr4b by Ol-Nanog. Simultaneous overexpression of Cxcr4b mRNA and depletion of Ol-Nanog protein in PGCs rescues the migration defective phenotype induced by a loss of Ol-Nanog, whereas overexpression of Sdf1a, the ligand for Cxcr4b, does not restore proper PGC migration. These results indicate that Ol-Nanog mediates PGC migration by regulating Cxcr4b expression.


Assuntos
Movimento Celular , Proteínas de Peixes/metabolismo , Células Germinativas/metabolismo , Proteínas de Homeodomínio/metabolismo , Receptores CXCR4/metabolismo , Regiões 3' não Traduzidas , Animais , Sítios de Ligação , Movimento Celular/genética , Quimiocina CXCL12/metabolismo , Imunoprecipitação da Cromatina , Proteínas de Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Genótipo , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Hibridização In Situ , Fases de Leitura Aberta , Oryzias/embriologia , Fenótipo , Regiões Promotoras Genéticas , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Dev Dyn ; 239(1): 297-310, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20014102

RESUMO

Differentiation of neural retinal precursor (NRP) cells in vertebrates follows an established order of cell-fate determination associated with exit from the cell cycle. Wnt signaling regulates cell cycle in colon carcinoma cells and has been implicated in different aspects of retinal development in various species. To better understand the biological roles of Wnt in the developing retina, we have used a transgenic and pharmacological approach to manipulate the Wnt signaling pathway during retinal development in medaka embryos. With the use of both approaches, we observed that during the early phase of retinal development Wnt signaling regulated cell cycle progression, proliferation, apoptosis, and differentiation of NRP cells. However, during later phases of retinal development, proliferation and apoptosis were not affected by manipulation of Wnt signaling. Instead, Wnt regulated Vsx1 expression, but not the expression of other retinal cell markers tested. Thus, the response of NRP cells to Wnt signaling is stage-dependent.


Assuntos
Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Retina/embriologia , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Fatores Etários , Animais , Apoptose/fisiologia , Primers do DNA/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Oryzias , Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Dev Dyn ; 239(2): 672-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20034054

RESUMO

Oct4 is a crucial transcription factor for controlling pluripotency in embryonic stem cells and the epiblast of mouse embryos. We have characterized the expression pattern of medaka (Oryzias latipes) Ol-Oct4 during embryonic development and in the adult gonads. Genomic analysis showed that Ol-Oct4 is the ortholog of zebrafish spg/pou2. However, their expression patterns are not the same, suggesting that Oct4 may play different roles in zebrafish and medaka. Using specific antibodies for the Ol-Oct4 protein, we showed that Ol-Oct4 is also expressed in primordial germ cells, in the spermatogonia (male germ stem cells), and during different stages of oocyte development. These results suggest that Ol-Oct4 plays a post-embryonic role in the maturing gonads and gametes. The Ol-Oct4 mRNA and protein expression patterns are similar to those of mammalian Oct4 and introduce medaka fish as a valid model for the functional and evolutionary study of pluripotency genes in vivo.


Assuntos
Desenvolvimento Embrionário , Fator 3 de Transcrição de Octâmero/metabolismo , Oryzias/embriologia , Ovário/metabolismo , Testículo/metabolismo , Sequência de Aminoácidos , Animais , Evolução Biológica , Encéfalo/metabolismo , Feminino , Células Germinativas/metabolismo , Masculino , Modelos Animais , Dados de Sequência Molecular , Oryzias/metabolismo , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico
8.
Stem Cells ; 27(9): 2081-91, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19544407

RESUMO

Nanog is involved in controlling pluripotency and differentiation of stem cells in vitro. However, its function in vivo has been studied only in mouse embryos and various reports suggest that Nanog may not be required for the regulation of differentiation. To better understand endogenous Nanog function, more animal models should be introduced to complement the murine model. Here, we have identified the homolog of the mammalian Nanog gene in teleost fish and describe the endogenous expression of Ol-Nanog mRNA and protein during medaka (Oryzias latipes) embryonic development and in the adult gonads. Using medaka fish as a vertebrate model to study Nanog function, we demonstrate that Ol-Nanog is necessary for S-phase transition and proliferation in the developing embryo. Moreover, inhibition or overexpression of Ol-Nanog does not affect gene expression of various pluripotency and differentiation markers, suggesting that this transcription factor may not play a direct role in embryonic germ layer differentiation. STEM CELLS 2009;27:2081-2091.


Assuntos
Proteínas de Peixes/fisiologia , Proteínas de Homeodomínio/fisiologia , Oryzias/embriologia , Oryzias/metabolismo , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proliferação de Células , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Gônadas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Oryzias/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/genética
9.
Sci Rep ; 10(1): 11345, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647304

RESUMO

Previous studies of global binding patterns identified the epigenetic factor, EZH2, as a regulator of the homeodomain-only protein homeobox (HOPX) gene expression during bone marrow stromal cell (BMSC) differentiation, suggesting a potential role for HOPX in regulating BMSC lineage specification. In the present study, we confirmed that EZH2 direct binds to the HOPX promoter region, during normal growth and osteogenic differentiation but not under adipogenic inductive conditions. HOPX gene knockdown and overexpression studies demonstrated that HOPX is a promoter of BMSC proliferation and an inhibitor of adipogenesis. However, functional studies failed to observe any affect by HOPX on BMSC osteogenic differentiation. RNA-seq analysis of HOPX overexpressing BMSC during adipogenesis, found HOPX function to be acting through suppression of adipogenic pathways associated genes such as ADIPOQ, FABP4, PLIN1 and PLIN4. These findings suggest that HOPX gene target pathways are critical factors in the regulation of fat metabolism.


Assuntos
Adipogenia , Proteínas de Homeodomínio/fisiologia , Células-Tronco Mesenquimais/citologia , Osteogênese , Proteínas Supressoras de Tumor/fisiologia , Adolescente , Adulto , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Feminino , Humanos , Masculino , Adulto Jovem
10.
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
11.
Stem Cells Dev ; 29(13): 823-834, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32268840

RESUMO

Multipotent bone marrow-derived mesenchymal stem/stromal cells (BMSCs) exhibit a finite life span after ex vivo expansion leading to cellular senescence. Many factors can contribute to this. Recently, our group has identified for the first time expression of the chemokine-like factor superfamily 8 (CMTM8) gene in cultured human BMSCs. In this study, we examine the role of CMTM8 in BMSC proliferation, migration, and differentiation. Functional studies using siRNA-mediated knockdown of CMTM8 in human BMSCs resulted in decreased capacity to undergo proliferation and migration and an increased capacity for osteogenic differentiation in vitro. Furthermore, reduced CMTM8 levels led to a decrease in the epidermal growth factor receptor (EGFR) signaling pathway during BMSC proliferation and migration, respectively. Supportive studies using retroviral mediated enforced expression of CMTM8 in BMSC resulted in an increased capacity for proliferation and migration but a decreased osteogenic differentiation potential. Collectively, these data suggest that CMTM8 promotes BMSC proliferation and BMSC migration through the EGFR/ERK1/2 pathway. This study provides insight into novel regulatory mechanisms of human BMSC growth and cell fate determination.


Assuntos
Diferenciação Celular , Quimiocinas/metabolismo , Proteínas com Domínio MARVEL/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Proliferação de Células , Células Cultivadas , Quimiocinas/genética , Receptores ErbB/metabolismo , Humanos , Proteínas com Domínio MARVEL/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Osteoblastos/metabolismo , Transdução de Sinais
12.
Stem Cells Dev ; 27(23): 1621-1633, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30229694

RESUMO

Key transcription factors, which activate or repress master gene regulators and signaling pathways, tightly regulate self-renewal and cell lineage differentiation of bone marrow-derived stromal cells (BMSC). Among these factors is the basic helix-loop-helix transcription factor Twist-related protein 1 (TWIST-1), which is important in BMSC self-renewal, life span, and differentiation. Another layer of gene regulation comes from microRNAs (miRNAs). miRNAs are short noncoding RNAs that interfere with translation of specific target mRNAs and thereby regulate diverse biological processes, including BMSC lineage commitment. However, little is known of how TWIST-1-regulated miRNAs control osteogenic commitment, and influence the fate of bone precursor cells. In this study, we have discovered a novel TWIST-1-regulated miRNA, miR-376c-3p. Reduced miR-376c-3p expression by a miR-376c-3p inhibitor or due to TWIST-1 haploinsufficiency promotes alkaline phosphatase (ALP) activity, mineral deposition, and expression of osteoblast-associated genes in BMSC and calvarial cells. Conversely, overexpression of miR-376c-3p using a miR-376c-3p mimic inhibited BMSC proliferation and the osteogenic potential of BMSC and TWIST-1 haploinsufficient calvarial cells. This was demonstrated by a decrease in insulin growth factor 1 receptor (IGF1R) levels, Akt signaling, ALP activity, mineral deposition, and expression of osteoblast-associated genes. Thus, miR-376c-3p reduces IGF1R/Akt signaling in BMSC and is one mechanism by which osteogenesis may be inhibited. Overall, we have identified miR-376c-3p as a TWIST-1-regulated miRNA, which plays an important role in the osteogenesis of bone precursor cells and can mediate TWIST-1 inhibition of osteogenesis. Furthermore, overexpression of miRNA-376c-3p in TWIST-1 haploinsufficient calvarial cells can decrease the aberrant osteogenesis of these cells, which contributes to increased calvarial bone volume and premature fusion of the coronal sutures.


Assuntos
Células da Medula Óssea/citologia , MicroRNAs/genética , Proteínas Nucleares/genética , Osteogênese/genética , Proteína 1 Relacionada a Twist/genética , Adolescente , Adulto , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Haploinsuficiência/genética , Humanos , Masculino , Osteoblastos/citologia , Osteoblastos/metabolismo , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Transdução de Sinais/genética , Crânio/citologia , Crânio/crescimento & desenvolvimento , Células Estromais/citologia , Células Estromais/metabolismo , Adulto Jovem
13.
Bone ; 94: 98-107, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27669657

RESUMO

The TWIST-1 gene encodes a basic helix-loop-helix (bHLH) transcription factor important in mediating skeletal and head mesodermal tissue development. Bone marrow-derived mesenchymal stem/stromal cells (BMSC), express high levels of TWIST-1, which is down regulated during ex vivo expansion. Cultured BMSC over-expressing TWIST-1 display decreased capacity for osteogenic differentiation and an enhanced capacity to undergo adipogenesis, suggesting that TWIST-1 is a mediator of lineage commitment. However, little is known regarding the mechanism(s) by which TWIST-1 mediates cell fate determination. In this study, microarray analysis was used to identify a novel downstream TWIST-1 target, tyrosine kinase receptor c-ros-oncogene 1 (C-ROS-1), which was down regulated in TWIST-1 over-expressing BMSC. Chromatin immunoprecipitation analysis showed that TWIST-1 directly bound to two E-box binding sites on the proximal C-ROS-1 promoter. Knock-down of C-ROS-1 in human BMSC and cranial bone cells resulted in a decreased capacity for osteogenic differentiation in vitro. Conversely, suppression of C-ROS-1 in BMSC resulted in an enhanced capacity to undergo adipogenesis. Furthermore, reduced C-ROS-1 levels led to activation of different components of the PI3K/AKT/mTORC1 signalling pathway during osteogenic and adipogenic differentiation. Collectively, these data suggest that C-ROS-1 is involved in BMSC fate switching between osteogenesis and adipogenesis, mediated via PI3K/AKT/mTORC1 signalling.


Assuntos
Linhagem da Célula , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Adipogenia/genética , Adulto , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Osteogênese/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Crânio/citologia
14.
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
15.
PLoS One ; 7(12): e51948, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23272192

RESUMO

Heart development depends on the spatio-temporally regulated contribution of progenitor cells from the primary, secondary and anterior heart fields. Primary heart field (PHF) cells are first recruited to form a linear heart tube; later, they contribute to the inflow myocardium of the four-chambered heart. Subsequently cells from the secondary (SHF) and anterior heart fields (AHF) are added to the heart tube and contribute to both the inflow and outflow myocardium. In amniotes, progenitors of the linear heart tube have been mapped to the anterior-middle region of the early primitive streak. After ingression, these cells are located within bilateral heart fields in the lateral plate mesoderm. On the other hand SHF/AHF field progenitors are situated anterior to the linear heart tube, however, the origin and location of these progenitors prior to the development of the heart tube remains elusive. Thus, an unresolved question in the process of cardiac development is where SHF/AHF progenitors originate from during gastrulation and whether they come from a region in the primitive streak distinct from that which generates the PHF. To determine the origin and location of SHF/AHF progenitors we used vital dye injection and tissue grafting experiments to map the location and ingression site of outflow myocardium progenitors in early primitive streak stage chicken embryos. Cells giving rise to the AHF ingressed from a rostral region of the primitive streak, termed region 'A'. During development these cells were located in the cranial paraxial mesoderm and in the pharyngeal mesoderm. Furthermore we identified region 'B', located posterior to 'A', which gave rise to progenitors that contributed to the primary heart tube and the outflow tract. Our studies identify two regions in the early primitive streak, one which generates cells of the AHF and a second from which cardiac progenitors of the PHF and SHF emerge.


Assuntos
Coração/embriologia , Linha Primitiva/embriologia , Células-Tronco/metabolismo , Animais , Região Branquial/embriologia , Movimento Celular , Embrião de Galinha , Mesoderma/metabolismo , Modelos Biológicos , Linha Primitiva/citologia
16.
Front Biosci (Landmark Ed) ; 16(7): 2416-26, 2011 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622186

RESUMO

During vertebrate embryogenesis the heart is the first functioning organ and cardiac progenitor cells (CPCs), which form the future heart, are among the first cell types to be established during gastrulation. A large number of studies indicate that cardiac development is tightly regulated by a series of molecular signaling pathways and morphological events. The cellular and molecular events that control early cardiac development are conserved among vertebrates. The favorable experimental characteristics of the chicken embryo and the ease in which cell labeling and imaging can be performed has allowed direct observation of the process of gastrulation and cell migration trajectories. This has enabled the study of the signaling proteins and molecular pathways required to specify early embryonic cells to the myocardial lineage. In this review we discuss the major morphogenetic and regulatory events that control gastrulation and migration of CPCs in the chicken embryo. We also describe the signaling mechanisms critical for early CPC specification in pre-gastrula, gastrula and early neurula stage embryos.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Coração Fetal/citologia , Coração Fetal/embriologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular , Movimento Celular , Embrião de Galinha , Transição Epitelial-Mesenquimal , Fatores de Crescimento de Fibroblastos/fisiologia , Gastrulação , Coração/embriologia , Camundongos , Linha Primitiva/citologia , Linha Primitiva/embriologia , Transdução de Sinais , Proteínas Wnt/fisiologia
17.
Int J Biol Sci ; 7(4): 410-7, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21547058

RESUMO

To understand the molecular mechanisms that regulate the biology of embryonic stem cells (ESCs) it is necessary to study how they behave in vivo in their natural environment. It is particularly important to study the roles and interactions of the different proteins involved in pluripotency and to use this knowledge for therapeutic purposes. The recent description of key pluripotency factors like Oct4 and Nanog in non-mammalian species has introduced other animal models, such as chicken, Xenopus, zebrafish and medaka, to the study of pluripotency in vivo. These animal models complement the mouse model and have provided new insights into the evolution of Oct4 and Nanog and their different functions during embryonic development. Furthermore, other pluripotency factors previously identified in teleost fish such as Klf4, STAT3, Sox2, telomerase and Tcf3 can now be studied in the context of a functional pluripotency network. The many experimental advantages of fish will fuel rapid analysis of the roles of pluripotency factors in fish embryonic development and the identification of new molecules and mechanisms governing pluripotency.


Assuntos
Oryzias/metabolismo , Células-Tronco Pluripotentes/fisiologia , Peixe-Zebra/metabolismo , Animais , Gônadas/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Humanos , Fator 4 Semelhante a Kruppel , Mamíferos/metabolismo , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/fisiologia , Oryzias/embriologia , Células-Tronco Pluripotentes/metabolismo , Peixe-Zebra/embriologia
18.
Zebrafish ; 7(2): 155-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20515319

RESUMO

Reliable animal models are required to facilitate the understanding of neurodegenerative pathways in Alzheimer's disease. Animal models can also be employed to search for disease-modifying drugs. The embryos and larvae of zebrafish are particularly advantageous for this purpose. For Alzheimer's disease, drugs that can ameliorate amyloid beta (A beta) toxicity have therapeutic and/or prophylactic potential. We attempted to generate a zebrafish model of A beta toxicity that would be viable and fertile but have a highly visible pigmentation phenotype in larvae. The larvae could then be arrayed in microtiter plates to screen compound libraries for drugs acting to reduce A beta toxicity. We used the promoter of the zebrafish mitfa (nacre) gene to drive expression of the pathological 42 amino acid species of human A beta, A beta(42), specifically in the highly visible melanophores (melanocytes) of transgenic zebrafish. However, the transgenic fish only showed an aberrant pigment phenotype in adults at the advanced age of 16 months. Nevertheless, our results show that alteration of zebrafish pigment pattern may be useful for analysis of toxic peptide action.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/toxicidade , Modelos Animais de Doenças , Peixe-Zebra , Animais , Primers do DNA/genética , Descoberta de Drogas/métodos , Técnicas de Transferência de Genes , Hibridização In Situ , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Pigmentação/efeitos dos fármacos , Pigmentação/fisiologia , Reação em Cadeia da Polimerase , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
19.
Pigment Cell Res ; 16(2): 117-26, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12622788

RESUMO

Sequence comparisons and functional analysis of the 5' upstream regions of tyrosinase genes have revealed the importance of cis-regulatory elements acting to control the spatiotemporal expression of tyrosinase in the melanocytes and retinal pigmented epithelium of developing embryos. To date there are no reports addressing the control of tyrosinase gene transcription in zebrafish, a vertebrate model organism of increasing importance. To exploit the tyrosinase gene as a marker in zebrafish we set out to clone its promoter and analyse its regulation during embryogenesis. Amplification of a zebrafish tyrosinase complementary DNA fragment by reverse transcriptase polymerase chain reaction allowed us to isolate and sequence a 1041 nt genomic DNA fragment that includes a transcription initiation site and 73 nt of the open reading frame. Bioinformatic analysis of this genomic sequence revealed five E-box motifs, including one CATGTG type E-box present in a putative initiation region. These are conserved positive regulatory elements in vertebrate tyrosinase promoters. We show that a region of 814 nt upstream from the translation start site of the zebrafish tyrosinase gene can drive expression in retinal pigmented epithelium in transiently transgenic zebrafish embryos but that its activity is not restricted to melanin-producing cells. This region is unable to drive transcription in human melanoma cell lines. Ectopic expression from this zebrafish tyrosinase promoter fragment is probably due to the absence of positive and negative cis-regulatory elements, such as a tyrosinase distal element, which is known to function as a pigment cell-specific enhancer.


Assuntos
Embrião não Mamífero/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação Enzimológica da Expressão Gênica/genética , Melaninas/biossíntese , Monofenol Mono-Oxigenase/genética , Epitélio Pigmentado Ocular/embriologia , Regiões Promotoras Genéticas/genética , Peixe-Zebra/embriologia , Animais , Sequência de Bases/genética , Elementos E-Box/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/enzimologia , Genes Reguladores/genética , Humanos , Melanócitos/enzimologia , Melanoma/enzimologia , Melanoma/genética , Dados de Sequência Molecular , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/enzimologia , Sítio de Iniciação de Transcrição/fisiologia , Células Tumorais Cultivadas/enzimologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
20.
Dev Genes Evol ; 213(1): 35-43, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12590351

RESUMO

Three homologues of the Drosophilaregion-specific homeotic gene spalt (sal) have been isolated in zebrafish, sall1a, sall1b and sall3. Phylogenetic analysis of these genes against known salDNA sequences showed zebrafish sall1aand sall1b to be orthologous to other vertebrate sal-1 genes and zebrafish sall3to be orthologous to other vertebrate sal-3 genes, except Xenopus sall3. Phylogenetic reconstruction suggests that zebrafish sall1a and sall1bresulted from a gene duplication event occurring prior to the divergence of the ray-finned and lobe-finned fish lineages. Analysis of the expression pattern of the zebrafish sal genes shows that sall1a and sall3 share expression domains with both orthologous and non-orthologous vertebrate sal genes. Both are expressed in various regions of the CNS, including in primary motor neurons. Outside of the CNS, sall1a expression is observed in the otic vesicle (ear), heart and in a discrete region of the pronephric ducts. These analyses indicate that orthologies between zebrafish sal genes and other vertebrate sal genes do not imply equivalence of expression pattern and, therefore, that biological functions are not entirely conserved. However we suggest that, like other vertebrate sal genes, zebrafish sal genes have a role in neural development. Also, expression of zebrafish sall1a in the otic vesicle, heart sac and the pronephric ducts of zebrafish embryos is possibly consistent with some of the abnormalities seen in Sall1-deficient mice and in Townes-Brocks Syndrome, a human disorder which is caused by mutations in the human spalt gene SALL1.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Filogenia , Fatores de Transcrição/genética , Proteínas de Xenopus , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Sequência de Bases , Evolução Molecular , Perfilação da Expressão Gênica , Cadeias de Markov , Dados de Sequência Molecular , Método de Monte Carlo , Alinhamento de Sequência , Homologia de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA