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










Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 41(18): e110815, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35912849

RESUMO

In vitro oogenesis is key to elucidating the mechanism of human female germ-cell development and its anomalies. Accordingly, pluripotent stem cells have been induced into primordial germ cell-like cells and into oogonia with epigenetic reprogramming, yet further reconstitutions remain a challenge. Here, we demonstrate ex vivo reconstitution of fetal oocyte development in both humans and cynomolgus monkeys (Macaca fascicularis). With an optimized culture of fetal ovary reaggregates over three months, human and monkey oogonia enter and complete the first meiotic prophase to differentiate into diplotene oocytes that form primordial follicles, the source for oogenesis in adults. The cytological and transcriptomic progressions of fetal oocyte development in vitro closely recapitulate those in vivo. A comparison of single-cell transcriptomes among humans, monkeys, and mice unravels primate-specific and conserved programs driving fetal oocyte development, the former including a distinct transcriptomic transformation upon oogonia-to-oocyte transition and the latter including two active X chromosomes with little X-chromosome upregulation. Our study provides a critical step forward for realizing human in vitro oogenesis and uncovers salient characteristics of fetal oocyte development in primates.


Assuntos
Meiose , Oogênese , Animais , Feminino , Humanos , Macaca fascicularis , Camundongos , Oócitos , Oogênese/fisiologia , Ovário
2.
Life Sci Alliance ; 5(12)2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35944930

RESUMO

Single-cell RNA sequencing (scRNA-seq) can determine gene expression in numerous individual cells simultaneously, promoting progress in the biomedical sciences. However, scRNA-seq data are high-dimensional with substantial technical noise, including dropouts. During analysis of scRNA-seq data, such noise engenders a statistical problem known as the curse of dimensionality (COD). Based on high-dimensional statistics, we herein formulate a noise reduction method, RECODE (resolution of the curse of dimensionality), for high-dimensional data with random sampling noise. We show that RECODE consistently resolves COD in relevant scRNA-seq data with unique molecular identifiers. RECODE does not involve dimension reduction and recovers expression values for all genes, including lowly expressed genes, realizing precise delineation of cell fate transitions and identification of rare cells with all gene information. Compared with representative imputation methods, RECODE employs different principles and exhibits superior overall performance in cell-clustering, expression value recovery, and single-cell-level analysis. The RECODE algorithm is parameter-free, data-driven, deterministic, and high-speed, and its applicability can be predicted based on the variance normalization performance. We propose RECODE as a powerful strategy for preprocessing noisy high-dimensional data.


Assuntos
Análise de Dados , Análise de Célula Única , Análise por Conglomerados , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Sequenciamento do Exoma
3.
EMBO J ; 41(13): e110600, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35703121

RESUMO

Germ cells are unique in engendering totipotency, yet the mechanisms underlying this capacity remain elusive. Here, we perform comprehensive and in-depth nucleome analysis of mouse germ-cell development in vitro, encompassing pluripotent precursors, primordial germ cells (PGCs) before and after epigenetic reprogramming, and spermatogonia/spermatogonial stem cells (SSCs). Although epigenetic reprogramming, including genome-wide DNA de-methylation, creates broadly open chromatin with abundant enhancer-like signatures, the augmented chromatin insulation safeguards transcriptional fidelity. These insulatory constraints are then erased en masse for spermatogonial development. Notably, despite distinguishing epigenetic programming, including global DNA re-methylation, the PGCs-to-spermatogonia/SSCs development entails further euchromatization. This accompanies substantial erasure of lamina-associated domains, generating spermatogonia/SSCs with a minimal peripheral attachment of chromatin except for pericentromeres-an architecture conserved in primates. Accordingly, faulty nucleome maturation, including persistent insulation and improper euchromatization, leads to impaired spermatogenic potential. Given that PGCs after epigenetic reprogramming serve as oogenic progenitors as well, our findings elucidate a principle for the nucleome programming that creates gametogenic progenitors in both sexes, defining a basis for nuclear totipotency.


Assuntos
Epigênese Genética , Células Germinativas , Animais , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA , Epigenômica , Feminino , Células Germinativas/metabolismo , Masculino , Mamíferos/genética , Camundongos , Espermatogônias
4.
Cell Stem Cell ; 28(12): 2167-2179.e9, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34496297

RESUMO

Mammalian male germ-cell development consists of three distinct phases: primordial germ cell (PGC) development, male germ-cell specification for spermatogonium development, and ensuing spermatogenesis. Here, we show an in vitro reconstitution of whole male germ-cell development by pluripotent stem cells (PSCs). Mouse embryonic stem cells (mESCs) are induced into PGC-like cells (mPGCLCs), which are expanded for epigenetic reprogramming. In reconstituted testes under an optimized condition, such mPGCLCs differentiate into spermatogonium-like cells with proper developmental transitions, gene expression, and cell-cycle dynamics and are expanded robustly as germline stem cell-like cells (GSCLCs) with an appropriate androgenetic epigenome. Importantly, GSCLCs show vigorous spermatogenesis, not only upon transplantation into testes in vivo but also under an in vitro culture of testis transplants, and the resultant spermatids contribute to fertile offspring. By uniting faithful recapitulations of the three phases of male germ-cell development, our study creates a paradigm for the in vitro male gametogenesis by PSCs.


Assuntos
Células-Tronco Pluripotentes , Animais , Diferenciação Celular , Epigenômica , Células Germinativas , Masculino , Camundongos , Espermatogênese , Espermatogônias
5.
Cell Stem Cell ; 28(6): 1023-1039.e13, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33831365

RESUMO

Trophoblasts are extraembryonic cells that are essential for maintaining pregnancy. Human trophoblasts arise from the morula as trophectoderm (TE), which, after implantation, differentiates into cytotrophoblasts (CTs), syncytiotrophoblasts (STs), and extravillous trophoblasts (EVTs), composing the placenta. Here we show that naïve, but not primed, human pluripotent stem cells (PSCs) recapitulate trophoblast development. Naive PSC-derived TE and CTs (nCTs) recreated human and monkey TE-to-CT transition. nCTs self-renewed as CT stem cells and had the characteristics of proliferating villous CTs and CTs in the cell column of the first trimester. Notably, although primed PSCs differentiated into trophoblast-like cells (BMP4, A83-01, and PD173074 [BAP]-treated primed PSCs [pBAPs]), pBAPs were distinct from nCTs and human placenta-derived CT stem cells, exhibiting properties consistent with the amnion. Our findings establish an authentic paradigm for human trophoblast development, demonstrating the invaluable properties of naive human PSCs. Our system provides a platform to study the molecular mechanisms underlying trophoblast development and related diseases.


Assuntos
Células-Tronco Pluripotentes , Trofoblastos , Diferenciação Celular , Feminino , Humanos , Placenta , Gravidez
6.
Life Sci Alliance ; 4(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33608411

RESUMO

The in vitro reconstitution of human germ-cell development provides a robust framework for clarifying key underlying mechanisms. Here, we explored transcription factors (TFs) that engender the germ-cell fate in their pluripotent precursors. Unexpectedly, SOX17, TFAP2C, and BLIMP1, which act under the BMP signaling and are indispensable for human primordial germ-cell-like cell (hPGCLC) specification, failed to induce hPGCLCs. In contrast, GATA3 or GATA2, immediate BMP effectors, combined with SOX17 and TFAP2C, generated hPGCLCs. GATA3/GATA2 knockouts dose-dependently impaired BMP-induced hPGCLC specification, whereas GATA3/GATA2 expression remained unaffected in SOX17, TFAP2C, or BLIMP1 knockouts. In cynomolgus monkeys, a key model for human development, GATA3, SOX17, and TFAP2C were co-expressed exclusively in early PGCs. Crucially, the TF-induced hPGCLCs acquired a hallmark of bona fide hPGCs to undergo epigenetic reprogramming and mature into oogonia/gonocytes in xenogeneic reconstituted ovaries. By uncovering a TF circuitry driving the germ line program, our study provides a paradigm for TF-based human gametogenesis.


Assuntos
Células Germinativas/metabolismo , Fatores de Transcrição SOXF/metabolismo , Fator de Transcrição AP-2/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Feminino , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Células Germinativas/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Macaca fascicularis , Camundongos , Camundongos Endogâmicos ICR , Fatores de Transcrição SOXF/genética , Transdução de Sinais/genética , Fator de Transcrição AP-2/genética , Fatores de Transcrição/metabolismo
7.
Nat Protoc ; 15(4): 1560-1583, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32231324

RESUMO

The human germ-cell lineage originates as human primordial germ cells (hPGCs). hPGCs undergo genome-wide epigenetic reprogramming and differentiate into oogonia or gonocytes, precursors for oocytes or spermatogonia, respectively. Here, we describe a protocol to differentiate human induced pluripotent stem cells (hiPSCs) into oogonia in vitro. hiPSCs are induced into incipient mesoderm-like cells (iMeLCs) using activin A and a WNT pathway agonist. iMeLCs, or, alternatively, hPSCs cultured with divergent signaling inhibitors, are induced into hPGC-like cells (hPGCLCs) in floating aggregates by cytokines including bone morphogenic protein 4. hPGCLCs are aggregated with mouse embryonic ovarian somatic cells to form xenogeneic reconstituted ovaries, which are cultured under an air-liquid interface condition for ~4 months for hPGCLCs to differentiate into oogonia and immediate precursory states for oocytes. To date, this is the only approach that generates oogonia from hPGCLCs. The protocol is suitable for investigating the mechanisms of hPGC specification and epigenetic reprogramming.


Assuntos
Diferenciação Celular/fisiologia , Técnicas Citológicas/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Oogônios/citologia , Animais , Células Cultivadas , Feminino , Células Germinativas/citologia , Humanos , Mesoderma/citologia , Camundongos
8.
Science ; 362(6412): 356-360, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30237246

RESUMO

Human in vitro gametogenesis may transform reproductive medicine. Human pluripotent stem cells (hPSCs) have been induced into primordial germ cell-like cells (hPGCLCs); however, further differentiation to a mature germ cell has not been achieved. Here, we show that hPGCLCs differentiate progressively into oogonia-like cells during a long-term in vitro culture (approximately 4 months) in xenogeneic reconstituted ovaries with mouse embryonic ovarian somatic cells. The hPGCLC-derived oogonia display hallmarks of epigenetic reprogramming-genome-wide DNA demethylation, imprint erasure, and extinguishment of aberrant DNA methylation in hPSCs-and acquire an immediate precursory state for meiotic recombination. Furthermore, the inactive X chromosome shows a progressive demethylation and reactivation, albeit partially. These findings establish the germline competence of hPSCs and provide a critical step toward human in vitro gametogenesis.


Assuntos
Técnicas de Reprogramação Celular/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Oogênese , Oogônios/citologia , Ovário/crescimento & desenvolvimento , Metilação de DNA , Epigênese Genética , Feminino , Humanos
9.
Cell Stem Cell ; 21(4): 517-532.e5, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28985527

RESUMO

Germline specification underlies human reproduction and evolution, but it has proven difficult to study in humans since it occurs shortly after blastocyst implantation. This process can be modeled with human induced pluripotent stem cells (hiPSCs) by differentiating them into primordial germ cell-like cells (hPGCLCs) through an incipient mesoderm-like cell (iMeLC) state. Here, we elucidate the key transcription factors and their interactions with important signaling pathways in driving hPGCLC differentiation from iPSCs. Germline competence of iMeLCs is dictated by the duration and dosage of WNT signaling, which induces expression of EOMES to activate SOX17, a key driver of hPGCLC specification. Upon hPGCLC induction, BMP signaling activates TFAP2C in a SOX17-independent manner. SOX17 and TFAP2C then cooperatively instate an hPGCLC transcriptional program, including BLIMP1 expression. This specification program diverges from its mouse counterpart regarding key transcription factors and their hierarchies, and it provides a foundation for further study of human germ cell development.


Assuntos
Evolução Biológica , Linhagem da Célula , Células Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Transdução de Sinais/genética , Transcrição Gênica , Animais , Linhagem da Célula/genética , Implantação do Embrião/genética , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Primatas , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Via de Sinalização Wnt/genética
10.
Philos Trans R Soc Lond B Biol Sci ; 369(1657)2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25349457

RESUMO

Mouse epiblast stem cells (EpiSCs) display temporal differences in the upregulation of Mixl1 expression during the initial steps of in vitro differentiation, which can be correlated with their propensity for endoderm differentiation. EpiSCs that upregulated Mixl1 rapidly during differentiation responded robustly to both Activin A and Nodal in generating foregut endoderm and precursors of pancreatic and hepatic tissues. By contrast, EpiSCs that delayed Mixl1 upregulation responded less effectively to Nodal and showed an overall suboptimal outcome of directed differentiation. The enhancement in endoderm potency in Mixl1-early cells may be accounted for by a rapid exit from the progenitor state and the efficient response to the induction of differentiation by Nodal. EpiSCs that readily differentiate into the endoderm cells are marked by a distinctive expression fingerprint of transforming growth factor (TGF)-ß signalling pathway genes and genes related to the endoderm lineage. Nodal appears to elicit responses that are associated with transition to a mesenchymal phenotype, whereas Activin A promotes gene expression associated with maintenance of an epithelial phenotype. We postulate that the formation of definitive endoderm (DE) in embryoid bodies follows a similar process to germ layer formation from the epiblast, requiring an initial de-epithelialization event and subsequent re-epithelialization. Our results show that priming EpiSCs with the appropriate form of TGF-ß signalling at the formative phase of endoderm differentiation impacts on the further progression into mature DE-derived lineages, and that this is influenced by the initial characteristics of the cell population. Our study also highlights that Activin A, which is commonly used as an in vitro surrogate for Nodal in differentiation protocols, does not elicit the same downstream effects as Nodal, and therefore may not effectively mimic events that take place in the mouse embryo.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Endoderma/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camadas Germinativas/embriologia , Subunidades beta de Inibinas/metabolismo , Proteína Nodal/metabolismo , Animais , Endoderma/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Camadas Germinativas/citologia , Proteínas de Homeodomínio/metabolismo , Camundongos , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
11.
Semin Cell Dev Biol ; 34: 65-75, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24954643

RESUMO

The timing of developmental events during early mouse development has been investigated in embryos that have been subject to experimental manipulation of cell number and tissue mass. These phenomenological studies revealed that the timing of preimplantation events, such as compaction, formation of blastocyst cavity and lineage allocation is correlated with the rounds of cleavage division or DNA replication of the blastomeres. Timing of postimplantation processes, such as formation of proamniotic cavity and onset of gastrulation is sensitive to cell number and probably the tissue mass, which may be measured by a mechanosensory signaling mechanism. Developmental changes in these two physical attributes are correlated with the cell proliferative activity and the growth trajectory of the whole embryo prior to the transit to organogenesis. During organogenesis, timing of morphogenesis appears to be regulated by individual devices that could be uncoupled during compensatory growth. Insights of the timing mechanism may be gleaned from the analysis of genomic activity associated with the transition through developmental milestones.


Assuntos
Blastocisto/fisiologia , Animais , Blastocisto/citologia , Blastômeros/fisiologia , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Morfogênese , Transcriptoma
12.
Cell Stem Cell ; 14(1): 107-20, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24139757

RESUMO

Mouse epiblast stem cells (EpiSCs) can be derived from a wide range of developmental stages. To characterize and compare EpiSCs with different origins, we derived a series of EpiSC lines from pregastrula stage to late-bud-stage mouse embryos. We found that the transcriptomes of these cells are hierarchically distinct from those of the embryonic stem cells, induced pluripotent stem cells (iPSCs), and epiblast/ectoderm. The EpiSCs display globally similar gene expression profiles irrespective of the original developmental stage of the source tissue. They are developmentally similar to the ectoderm of the late-gastrula-stage embryo and behave like anterior primitive streak cells when differentiated in vitro and in vivo. The EpiSC lines that we derived can also be categorized based on a correlation between gene expression signature and predisposition to differentiate into particular germ-layer derivatives. Our findings therefore highlight distinct identifying characteristics of EpiSCs and provide a foundation for further examination of EpiSC properties and potential.


Assuntos
Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Linha Primitiva/citologia , Animais , Biomarcadores/metabolismo , Western Blotting , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Gastrulação , Perfilação da Expressão Gênica , Camadas Germinativas/metabolismo , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco Pluripotentes/metabolismo , Linha Primitiva/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Dev Growth Differ ; 53(7): 843-56, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21762130

RESUMO

WNT signaling activity is involved in the regulation of many cellular functions, including proliferation, migration, cell fate specification, maintenance of pluripotency and induction of tumorigenicity. Here we summarize recent progress towards understanding the regulation of canonical WNT/ß-catenin signaling activity through feedback regulatory loops involving the ligands, agonists and antagonists, the availability of intracellular pools of active ß-catenin and the cross-regulation of the WNT activity by ß-catenin independent pathway. We also review recent findings on the role of WNT/ß-catenin signaling in tissue lineage differentiation during embryogenesis and the maintenance and self renewal of embryo-derived stem cells in vitro.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Movimento Celular , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Glipicanas/metabolismo , Coração/embriologia , Coração/crescimento & desenvolvimento , Proteoglicanas de Heparan Sulfato/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Fenótipo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Proteínas Wnt/agonistas , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
Circ Cardiovasc Genet ; 4(4): 446-54, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21642241

RESUMO

BACKGROUND: Recently, elevation of circulating muscle-specific microRNA (miRNA) levels has been reported in patients with acute myocardial infarction. However, it is still unclear from which part of the myocardium or under what conditions miRNAs are released into circulating blood. The purpose of this study was to identify the source of elevated levels of circulating miRNAs and their function in cardiovascular diseases. METHODS AND RESULTS: Serum levels of miRNA (miR)-1 and miR-133a were increased significantly in patients not only with acute myocardial infarction but also with unstable angina pectoris and Takotsubo cardiomyopathy without elevation of serum creatine phosphokinase or cardiac troponin. MicroRNA microarray analysis of the heart from a mouse model of myocardial infarction indicated that the levels of miR-1, miR-133a, miR-208a, and miR-499 were significantly reduced in the infarcted myocardium. In situ hybridization of miR-133a also showed that miR-133a levels were very low in the infarcted and peri-infarcted myocardium. It has been shown that circulating miRNAs are localized inside exosomes, which are released after Ca(2+) stimulation. We stimulated H9c2 cardiomyoblasts with A23187 and measured miR-133a levels in the exosome fraction of the culture medium. A23187 induced a dose-dependent release of miR-133a, and significant elevation was observed only at concentrations where dead cells were detected. We also found that miR-133a-containing exosomes reduced the luciferase activity of 293FT cells transfected with an miR-133a sensor vector. CONCLUSIONS: These results suggest that elevated levels of circulating miR-133a in patients with cardiovascular diseases originate mainly from the injured myocardium. Circulating miR-133a can be used as a marker for cardiomyocyte death, and it may have functions in cardiovascular diseases.


Assuntos
Doenças Cardiovasculares/etiologia , MicroRNAs/sangue , Miocárdio/patologia , Animais , Biomarcadores/sangue , Cálcio , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/patologia , Morte Celular , Exossomos , Humanos , Camundongos , MicroRNAs/fisiologia , Miócitos Cardíacos/patologia
15.
J Mol Cell Cardiol ; 48(6): 1157-68, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19853610

RESUMO

Screening for cell surface proteins up-regulated under stress conditions may lead to the identification of new therapeutic targets. To search for genes whose expression was enhanced by treatment with oligomycin, a mitochondrial-F(0)F(1) ATP synthase inhibitor, signal sequence trapping was performed in H9C2 rat cardiac myoblasts. One of the genes identified was that for neural cell adhesion molecule (NCAM, CD56), a major regulator of development, cell survival, migration, and neurite outgrowth in the nervous system. Immunohistochemical analyses in a mouse myocardial infarction model revealed that NCAM was strongly expressed in residual cardiac myocytes in the infarcted region. Increased expression of NCAM was also found during the remodeling period in a rat model of hypertension-induced heart failure. Lentivirus-mediated knockdown of NCAM decreased the cell growth and survival following oligomycin treatment in H9C2 cells. In primary rat neonatal cardiac myocytes, NCAM was also found to be up-regulated and played a protective role following oligomycin treatment. Analyses of downstream signaling revealed that knockdown of NCAM significantly decreased the basal AKT phosphorylation level. In contrast, NCAM mimetic peptide P2d activated AKT and significantly reduced oligomycin-induced cardiomyocyte death, which was abolished by treatment with the PI3K inhibitor LY-294002 as well as overexpression of the dominant-negative AKT mutant. These findings demonstrate that NCAM is a cardioprotective factor up-regulated under metabolic stress in cardiomyocytes and augmentation of this signal improved survival.


Assuntos
Cardiotônicos/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Regulação para Cima , Animais , Membrana Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Miócitos Cardíacos/citologia , Oligomicinas/farmacologia , ATPases Translocadoras de Prótons/antagonistas & inibidores , Ratos
16.
Biochem Biophys Res Commun ; 390(3): 821-6, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19836354

RESUMO

Cardiac transcription factors play crucial roles in cardiac development and differentiation. To address the target genes they regulate is important for understanding the molecular mechanisms underlying these processes. In this study, ES cell lines harboring a neomycin resistance gene in the Nkx2-5 gene locus were generated and used to make purified ES cell-derived cardiomyocytes (ESCM) by in vitro differentiation and successive G418 treatment. Three lines with different Nkx2-5 gene expression levels were made and named as Nkx2-5 -/-, +/-, and overexpressing ESCMs. In order to investigate Nkx2-5-regulated gene expression in cardiomyocytes, the gene expression profiles were compared among these lines. The expression patterns of known Nkx2-5 downstream genes were consistent with the previous investigations in vivo. Several genes with Nkx2-5-dependent changes in their expression were detected and confirmed by real-time PCR. This study investigated Nkx2-5-regulated gene expression in ESCM and suggested potential targets of Nkx2-5 in cardiogenesis.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Proteínas de Homeodomínio/metabolismo , Miócitos Cardíacos/citologia , Organogênese/genética , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Separação Celular , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Camundongos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/genética
17.
Atherosclerosis ; 206(1): 102-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19321167

RESUMO

BACKGROUND: Progranulin (PGRN) is a unique growth factor that plays an important role in cutaneous wound healing. It has an anti-inflammatory effect and promotes cell proliferation. However, when it is degraded to granulin peptides (GRNs) by neutrophil proteases, a pro-inflammatory reaction occurs. Since injury, inflammation and repair are common features in the progression of atherosclerosis, it is conceivable that PGRN plays a role in atherogenesis. RESULTS: Immunohistochemical analysis of human carotid endoatherectomy specimens indicated that vascular smooth muscle cells (vSMCs) in the intima expressed PGRN. Some macrophages in the plaque also expressed PGRN. We assessed the effect of PGRN on a human monocytic leukemia cell line (THP-1) and human aortic smooth muscle cells (HASMCs). PGRN alone had no effect on HASMC or THP-1 proliferation or migration. However, when THP-1 cells were stimulated with MCP-1, the number of migrated cells decreased in a PGRN-dose-dependent manner. TNF-alpha-induced HASMC migration was enhanced only at 10nM of PGRN. Interleukin-8 (IL-8) secretion from HASMCs was reduced by forced expression of PGRN and increased by RNAi-mediated knockdown of PGRN. While exogenous treatment with recombinant PGRN decreased IL-8 secretion, degraded recombinant GRNs increased IL-8 secretion from HASMCs. CONCLUSIONS: The expression of PGRN mainly reduces inflammation and its degradation into GRNs enhances inflammation in atherosclerotic plaque and may contribute to the progression of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Doenças das Artérias Carótidas/fisiopatologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/farmacologia , Humanos , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/metabolismo , Progranulinas , Fator de Necrose Tumoral alfa/farmacologia , Túnica Íntima/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 28(6): 1077-83, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18388330

RESUMO

OBJECTIVE: CCN1 (Cyr61) is an extracellular matrix-associated protein involved in cell proliferation and survival. CCN1 is bound to vascular smooth muscle cells (VSMCs) via integrins and is expressed in VSMCs in atherosclerotic lesions, suggesting involvement in the regulation of vascular smooth muscle cell (VSMC) proliferation and atherosclerosis. We hypothesized that knockdown of CCN1 may inhibit VSMC proliferation and suppress neointimal hyperplasia. METHODS AND RESULTS: We examined the effect of the knockdown of CCN1 using rat cultured VSMCs and a rat balloon injury model. CCN1 stimulated adhesion and migration of VSMCs in a dose-dependent manner, and this was blocked by an antibody for integrin alpha(6)beta(1). Moreover, knockdown of endogenous CCN1 by lentiviral delivery of siRNA significantly inhibited proliferation of VSMCs and the uptake of 5-bromo-2'-deoxyuridine (BrdU). Replenishment with recombinant CCN1 reversed the effect of siRNA knockdown. Interestingly, knockdown of CCN1 significantly suppressed neointimal hyperplasia in a rat carotid artery balloon injury model at days 14 and 28 after injury. Gene transfer of CCN1 to smooth muscle reversed the effect of CCN1 knockdown on neointimal formation. These results suggest that endogenous CCN1 regulates proliferation of VSMCs and neointimal hyperplasia. CONCLUSIONS: Inhibition of CCN1 may provide a promising strategy for the prevention of restenosis after vascular interventions.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Angioplastia com Balão/efeitos adversos , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteína Rica em Cisteína 61 , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Hiperplasia/metabolismo , Hiperplasia/patologia , Proteínas Imediatamente Precoces/genética , Integrinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley
19.
Biochem Biophys Res Commun ; 355(3): 611-8, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17316559

RESUMO

CCN1 (Cyr61) is a secreted matricellular protein, mediating angiogenesis and cell survival through interaction with integrins. Although CCN1 expression is induced in the heart during ischemia and pressure overload, its function in cardiac myocytes remains to be elucidated. We hypothesized that CCN1 may not only induce angiogenesis but may also have a direct effect on cardiac myocytes during ischemia. In this study, we investigated the effect of CCN1 on survival of cardiac myocytes under oxidative stress and examined a signal transduction pathway downstream of CCN1. A solid-phase binding assay demonstrated that CCN1 was bound to cardiac myocytes in a dose-dependent, saturable manner. Inactivation of beta1 integrin in cardiac myocytes inhibited binding with CCN1, indicating that CCN1 was bound to cardiac myocytes via beta1 integrin. Knockdown of endogenous CCN1 decreased the number of surviving cells under oxidative stress, while pretreatment of cardiac myocytes with recombinant CCN1 significantly increased the number of surviving cells. Moreover, TUNEL staining showed that CCN1 significantly decreased apoptotic cells. Furthermore, treatment of cardiac myocytes with CCN1 induced phosphorylation of Akt and extracellular signal-regulated kinase (ERK). Inactivation of beta1 integrin inhibited CCN1-induced phosphorylation of these kinases and abolished the protective effect of CCN1. Moreover, pretreatment of cells with wortmannin completely blocked the protective effect of CCN1 on cardiac myocytes under oxidative stress, indicating that the protective effect of CCN1 was mainly mediated by activation of Akt. The antiapoptotic effect of CCN1 on cardiac myocytes together with its proangiogenic property could be beneficial in the treatment of ischemic heart disease.


Assuntos
Apoptose , Proteínas Imediatamente Precoces/farmacologia , Integrina beta1/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteína Rica em Cisteína 61 , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos
20.
Int Heart J ; 46(5): 927-31, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16272785

RESUMO

A 69-year-old female suffering from third-degree atrioventricular block with syncope underwent permanent pacemaker implantation. However, she developed shortness of breath 2 months after the implantation. Blood tests revealed elevated levels of LDH, CRP, BNP, and SIL-2R. Transthoracic echocardiography showed thickened left and right atrial walls with mild pericardial effusion. A diagnosis was made based on a CT scan and histology. Although most primary cardiac malignant lymphomas are associated with a poor prognosis, the patient was treated successfully with chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bloqueio Cardíaco/complicações , Neoplasias Cardíacas/tratamento farmacológico , Linfoma de Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Esquema de Medicação , Feminino , Bloqueio Cardíaco/terapia , Neoplasias Cardíacas/etiologia , Humanos , Linfoma de Células B/etiologia , Linfoma Difuso de Grandes Células B/etiologia , Marca-Passo Artificial , Prednisolona/administração & dosagem , Vincristina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...