Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Reproduction ; 155(2): 153-166, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29326135

RESUMO

Our overall goal is to create a three-dimensional human cell-based testicular model for toxicological and spermatogenesis studies. Methods to purify the major somatic testicular cells, namely Leydig cells (LCs), peritubular myoid cells (PCs) and Sertoli cells (SCs), from rats, mice and guinea pigs have been reported. In humans, the isolation of populations enriched for primary LCs, PCs or SCs also have described. One objective of this study was to determine if populations of cells enriched for all three of these cell types can be isolated from testes of single human donors, and we were successful in doing so from testes of three donors. Testes tissues were enzymatically digested, gravity sedimented and Percoll filtered to isolate populations enriched for LCs, PCs and SCs. LCs and PCs were identified by colorimetric detection of the expression of prototypical enzymes. Division of PCs and SCs in culture has been reported. We observed that primary human LCs could divide in culture by incorporation of 5-ethynyl-2'-deoxyuridine. SCs were identified and their functionality was demonstrated by the formation of tight junctions as shown by the expression of tight junction proteins, increased transepithelial electrical resistance, polarized secretion of biomolecules and inhibition of lucifer yellow penetration. Furthermore, we found that human SC feeder layers could facilitate germ cell progression of human embryonic stem cells (hESCs) by microarray analysis of gene expression.


Assuntos
Separação Celular/métodos , Técnicas de Cocultura/métodos , Células-Tronco Embrionárias/citologia , Células Intersticiais do Testículo/citologia , Células de Sertoli/citologia , Adulto , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Humanos , Células Intersticiais do Testículo/metabolismo , Masculino , Pessoa de Meia-Idade , Células de Sertoli/metabolismo , Espermatogênese , Testículo
2.
Hum Mol Genet ; 23(12): 3071-84, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24449759

RESUMO

Studies of human germ cell development are limited in large part by inaccessibility of germ cells during development. Moreover, although several studies have reported differentiation of mouse and human germ cells from pluripotent stem cells (PSCs) in vitro, differentiation of human germ cells from PSCs in vivo has not been reported. Here, we tested whether mRNA reprogramming in combination with xeno-transplantation may provide a viable system to probe the genetics of human germ cell development via use of induced pluripotent stem cells (iPSCs). For this purpose, we derived integration-free iPSCs via mRNA-based reprogramming with OCT3/4, SOX2, KLF4 and cMYC alone (OSKM) or in combination with the germ cell-specific mRNA, VASA (OSKMV). All iPSC lines met classic criteria of pluripotency. Moreover, global gene expression profiling did not distinguish large differences between undifferentiated OSKM and OSKMV iPSCs; however, some differences were observed in expression of pluripotency factors and germ cell-specific genes, and in epigenetic profiles and in vitro differentiation studies. In contrast, transplantation of undifferentiated iPSCs directly into the seminiferous tubules of germ cell-depleted immunodeficient mice revealed divergent fates of iPSCs produced with different factors. Transplantation resulted in morphologically and immunohistochemically recognizable germ cells in vivo, particularly in the case of OSKMV cells. Significantly, OSKMV cells also did not form tumors while OSKM cells that remained outside the seminiferous tubule proliferated extensively and formed tumors. Results indicate that mRNA reprogramming in combination with transplantation may contribute to tools for genetic analysis of human germ cell development.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Túbulos Seminíferos/metabolismo , Espermatozoides/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Epigênese Genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Camundongos Nus , Espermatozoides/citologia , Transplante Heterólogo/métodos
3.
Mol Cancer Ther ; 23(7): 949-960, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38507740

RESUMO

The activated B cell (ABC) subset of diffuse large B-cell lymphoma (DLBCL) is characterized by chronic B-cell receptor signaling and associated with poor outcomes when treated with standard therapy. In ABC-DLBCL, MALT1 is a core enzyme that is constitutively activated by stimulation of the B-cell receptor or gain-of-function mutations in upstream components of the signaling pathway, making it an attractive therapeutic target. We discovered a novel small-molecule inhibitor, ABBV-MALT1, that potently shuts down B-cell signaling selectively in ABC-DLBCL preclinical models leading to potent cell growth and xenograft inhibition. We also identified a rational combination partner for ABBV-MALT1 in the BCL2 inhibitor, venetoclax, which when combined significantly synergizes to elicit deep and durable responses in preclinical models. This work highlights the potential of ABBV-MALT1 monotherapy and combination with venetoclax as effective treatment options for patients with ABC-DLBCL.


Assuntos
Sinergismo Farmacológico , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , Proteínas Proto-Oncogênicas c-bcl-2 , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/antagonistas & inibidores , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Humanos , Animais , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Linhagem Celular Tumoral , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças
4.
Stem Cells ; 30(3): 441-51, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22162380

RESUMO

Our understanding of human germ cell development is limited in large part due to inaccessibility of early human development to molecular genetic analysis. Pluripotent human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) have been shown to differentiate to cells of all three embryonic germ layers, as well as germ cells in vitro, and thus may provide a model for the study of the genetics and epigenetics of human germline. Here, we examined whether intrinsic germ cell translational, rather than transcriptional, factors might drive germline formation and/or differentiation from human pluripotent stem cells in vitro. We observed that, with overexpression of VASA (DDX4) and/or DAZL (Deleted in Azoospermia Like), both hESCs and iPSCs differentiated to primordial germ cells, and maturation and progression through meiosis was enhanced. These results demonstrate that evolutionarily unrelated and divergent RNA-binding proteins can promote meiotic progression of human-derived germ cells in vitro. These studies describe an in vitro model for exploring specifics of human meiosis, a process that is remarkably susceptible to errors that lead to different infertility-related diseases.


Assuntos
RNA Helicases DEAD-box/fisiologia , Células Germinativas/metabolismo , Meiose , Células-Tronco Pluripotentes/fisiologia , Proteínas de Ligação a RNA/fisiologia , Animais , Antígenos de Diferenciação/metabolismo , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , RNA Helicases DEAD-box/metabolismo , Metilação de DNA , Perfilação da Expressão Gênica , Genes Reporter , Células Germinativas/citologia , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Células-Tronco Pluripotentes/metabolismo , RNA Longo não Codificante , RNA não Traduzido/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Análise de Sequência de DNA , Complexo Sinaptonêmico/metabolismo
5.
PLoS One ; 18(9): e0291029, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37751459

RESUMO

Neurodegenerative diseases encompass a group of debilitating conditions resulting from progressive nerve cell death. Of these, Alzheimer's disease (AD) occurs most frequently, but is currently incurable and has limited treatment success. Late onset AD, the most common form, is highly heritable but is caused by a combination of non-genetic risk factors and many low-effect genetic variants whose disease-causing mechanisms remain unclear. By mining the FinnGen study database of phenome-wide association studies, we identified a rare variant, rs148726219, enriched in the Finnish population that is associated with AD risk and dementia, and appears to have arisen on a common haplotype with older AD-associated variants such as rs429358. The rs148726219 variant lies in an overlapping intron of the FosB proto-oncogene (FOSB) and ERCC excision repair 1 (ERCC1) genes. To understand the impact of this SNP on disease phenotypes, we performed CRISPR/Cas9 editing in a human induced pluripotent stem cell (hiPSC) line to generate isogenic clones harboring heterozygous and homozygous alleles of rs148726219. hiPSC clones differentiated into induced excitatory neurons (iNs) did not exhibit detectable molecular or morphological variation in differentiation potential compared to isogenic controls. However, global transcriptome analysis showed differential regulation of nearby genes and upregulation of several biological pathways related to neuronal function, particularly synaptogenesis and calcium signaling, specifically in mature iNs harboring rs148726219 homozygous and heterozygous alleles. Functional differences in iN circuit maturation as measured by calcium imaging were observed across genotypes. Edited mature iNs also displayed downregulation of unfolded protein response and cell death pathways. This study implicates a phenotypic impact of rs148726219 in the context of mature neurons, consistent with its identification in late onset AD, and underscores a hiPSC-based experimental model to functionalize GWAS-identified variants.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Doença de Alzheimer/metabolismo , Polimorfismo de Nucleotídeo Único , Genótipo , Neurônios
6.
J Biol Chem ; 286(22): 19860-71, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21471197

RESUMO

During implantation, the uterine stromal cells undergo terminal differentiation into decidual cells, which support the proper progression of maternal-embryo interactions to successful establishment of pregnancy. The decidual cells synthesize extracellular matrix (ECM) components, such as laminins and collagens, which assemble into a unique basal lamina-like network that surrounds these cells. The functional significance of this matrix during implantation is unknown. We previously showed that the transcription factor CCAAT enhancer-binding protein ß (C/EBPß) critically regulates decidualization in the mouse. We now provide evidence that C/EBPß directly controls the Lamc1 gene, which encodes a predominant laminin constituent of the ECM produced by the decidual cells. Suppression of Lamc1 expression in mouse primary endometrial stromal cells prevented the assembly of this ECM and impaired stromal differentiation. Attenuation of expression of integrin ß1, a major constituent of the integrin receptors targeted by decidual laminins, also inhibited this differentiation process. Disruption of laminin-integrin interactions led to impaired activation of the focal adhesion kinase, an integrin-mediated regulator of cytoskeletal remodeling during decidualization. To further analyze the role of the decidual ECM in modulating maternal-embryo interactions, we monitored trophoblast invasion into differentiating uterine stromal monolayers, using a co-culture system. Silencing of stromal Lamc1 expression, which prevented formation of the basal lamina-like matrix, resulted in marked reduction in trophoblast outgrowth. Collectively, our findings identified C/EBPß as a critical regulator of the unique ECM that controls decidual cell architecture and differentiation, and it provided new insights into the mechanisms by which the uterine stromal microenvironment controls the progression of embryo implantation.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Decídua/metabolismo , Implantação do Embrião/fisiologia , Regulação da Expressão Gênica/fisiologia , Trofoblastos/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Decídua/citologia , Ativação Enzimática , Matriz Extracelular , Feminino , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Inativação Gênica , Integrinas/genética , Integrinas/metabolismo , Laminina/biossíntese , Laminina/genética , Troca Materno-Fetal/fisiologia , Camundongos , Gravidez , Células Estromais/citologia , Células Estromais/metabolismo , Trofoblastos/citologia
7.
Oncotarget ; 13: 1-12, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35018214

RESUMO

Inhibitors of poly(ADP)-ribose polymerase (PARP) exploit defective DNA repair pathways existing in several forms of cancer, such as those with BRCA mutations, and have proven clinical efficacy as chemosensitizers. However, platinum-based chemopotentiation by PARP inhibitors (PARPi), particularly for non-small cell lung cancer (NSCLC), has only been confirmed in a few preclinical models and the molecular mechanisms that drive PARPi combinatorial synergy with chemotherapeutics remains poorly defined. To better understand these mechanisms, we characterized cisplatin and veliparib efficacy in A549 and Calu6 NSCLC in vivo tumor xenograft models and observed combinatorial synergy in the Calu6 model. Transcriptome-wide analysis of xenografts revealed several differentially expressed genes (DEGs) between untreated and cisplatin + veliparib-treated groups, which were unique from genes identified in either of the single-agent treatment arms. Particularly at 10- and 21-days post-treatment, these DEGs were enriched within pathways involved in DNA damage repair, cell cycle regulation, and senescence. Furthermore, TGF-ß- and integrin-related pathways were enriched in the combination treatment arm, while pathways involved in cholesterol metabolism were identified at earlier time points in both the combination and cisplatin-only groups. These data advance the biological underpinnings of PARPi combined with platinum-based chemotherapy and provides additional insight into the diverse sensitivity of NSCLC models.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Difosfato de Adenosina , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Colesterol , Cisplatino , Humanos , Integrinas/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Platina/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Poli(ADP-Ribose) Polimerases/metabolismo , Ribose/uso terapêutico , Transcriptoma , Fator de Crescimento Transformador beta/genética
8.
Clin Cancer Res ; 27(18): 4983-4993, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131001

RESUMO

PURPOSE: Safety, efficacy, and exploratory biomarker analyses were evaluated in patients with advanced HER2-negative germline breast cancer susceptibility gene (gBRCA)-associated breast cancer enrolled in the BROCADE3 trial who received crossover veliparib monotherapy after disease progression on placebo plus carboplatin/paclitaxel. PATIENTS AND METHODS: Eligible patients (N = 513) were randomized 2:1 to veliparib plus carboplatin/paclitaxel or placebo plus carboplatin/paclitaxel; patients had variable platinum-free intervals (PFI) at progression. In the placebo arm, patients were eligible to receive crossover veliparib monotherapy (300-400 mg twice daily continuous). Antitumor activity and adverse events were assessed during crossover veliparib treatment. BRCA reversion mutations at crossover were analyzed retrospectively using next-generation sequencing on plasma circulating tumor DNA (ctDNA). RESULTS: Seventy-five patients in the placebo plus carboplatin/paclitaxel arm received ≥1 dose of crossover veliparib postprogression (mean treatment duration: 154 days). Eight of 50 (16%) patients with measurable disease had a RECIST v1.1 response. Activity was greater in patients with PFI ≥180 days compared with <180 days [responses in 23.1% (3/13) vs. 13.5% (5/37) of patients]. BRCA reversion mutations that restored protein function were detected in ctDNA from 4 of 28 patients tested, and the mean duration of crossover veliparib monotherapy was <1 month in these 4 patients versus 7.49 months in patients lacking reversion mutations. The most frequent adverse events were nausea (61%), vomiting (29%), and fatigue (24%). CONCLUSIONS: Crossover veliparib monotherapy demonstrated limited antitumor activity in patients who experienced disease progression on placebo plus carboplatin/paclitaxel. PFI appeared to affect veliparib activity. BRCA reversion mutations may promote cross-resistance and limit veliparib activity following progression on platinum.


Assuntos
Neoplasias da Mama , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Benzimidazóis , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Carboplatina/efeitos adversos , Feminino , Humanos , Mutação , Paclitaxel/efeitos adversos , Platina/uso terapêutico , Estudos Retrospectivos
9.
PLoS One ; 11(10): e0165268, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27768780

RESUMO

The mechanisms underlying human germ cell development are largely unknown, partly due to the scarcity of primordial germ cells and the inaccessibility of the human germline to genetic analysis. Human embryonic stem cells can differentiate to germ cells in vitro and can be genetically modified to study the genetic requirements for germ cell development. Here, we studied NANOS3 and DAZL, which have critical roles in germ cell development in several species, via their over expression in human embryonic stem cells using global transcriptional analysis, in vitro germ cell differentiation, and in vivo germ cell formation assay by xenotransplantation. We found that NANOS3 over expression prolonged pluripotency and delayed differentiation. In addition, we observed a possible connection of NANOS3 with inhibition of apoptosis. For DAZL, our results suggest a post-transcriptional regulation mechanism in hES cells. In addition, we found that DAZL suppressed the translation of OCT4, and affected the transcription of several genes associated with germ cells, cell cycle arrest, and cell migration. Furthermore, DAZL over expressed cells formed spermatogonia-like colonies in a rare instance upon xenotransplantation. These data can be used to further elucidate the role of NANOS3 and DAZL in germ cell development both in vitro and in vivo.


Assuntos
Células-Tronco Embrionárias/metabolismo , Proteínas de Ligação a RNA/metabolismo , Diferenciação Celular , Feminino , Expressão Gênica , Células Germinativas/citologia , Xenoenxertos , Humanos , Masculino , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Transcrição Gênica
10.
Sci Rep ; 5: 15041, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26456624

RESUMO

Deletions of the AZFa region (AZoospermia Factor-a) region of the human Y chromosome cause irreversible spermatogenic failure that presents clinically in men as Sertoli-cell only (SCO) pathology of the testis. Deletions of the AZFa region typically encompass two genes: DDX3Y and USP9Y. However, human genetic evidence indicates that SCO is most tightly linked to deletion of DDX3Y and that deletions/mutations of USP9Y can be transmitted from one generation to the next. Here, we generated stable iPSC lines with AZFa deletions, tested complementation via introduction of DDX3Y, and assessed ability to form germ cells in vivo in a xenotransplantation model. We observed a quantifiable improvement in formation of germ cell like cells (GCLCs) from complemented donor iPSCs. Moreover, expression of UTF1, a prospermatogonial protein, was restored in cells complemented by introduction of DDX3Y on the AZFa background. Whole-genome RNA sequencing of purified GCLCs revealed an enrichment of genes involved in translational suppression and transcriptional control in DDX3Y-rescued GCLCs over mutant GCLCs, which maintained a molecular phenotype more similar to undifferentiated iPSCs. This study demonstrates the ability to probe fundamental genetics of human germ cell formation by complementation and indicates that DDX3Y functions in the earliest stages of human germ cell development.


Assuntos
Cromossomos Humanos Y/metabolismo , RNA Helicases DEAD-box/genética , Células-Tronco Pluripotentes Induzidas/citologia , Espermatogênese/genética , Espermatozoides/metabolismo , Transcrição Gênica , Animais , Bussulfano/farmacologia , Diferenciação Celular , Cromossomos Humanos Y/química , RNA Helicases DEAD-box/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Teste de Complementação Genética , Vetores Genéticos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos Nus , Antígenos de Histocompatibilidade Menor , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pele/citologia , Pele/metabolismo , Espermatozoides/citologia , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/metabolismo , Transativadores/genética , Transativadores/metabolismo , Transplante Heterólogo , Proteína Vermelha Fluorescente
11.
Cell Rep ; 7(4): 1284-97, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24794432

RESUMO

Historically, spontaneous deletions and insertions have provided means to probe germline developmental genetics in Drosophila, mouse and other species. Here, induced pluripotent stem cell (iPSC) lines were derived from infertile men with deletions that encompass three Y chromosome azoospermia factor (AZF) regions and are associated with production of few or no sperm but normal somatic development. AZF-deleted iPSC lines were compromised in germ cell development in vitro. Undifferentiated iPSCs transplanted directly into murine seminiferous tubules differentiated extensively to germ-cell-like cells (GCLCs) that localized near the basement membrane, demonstrated morphology indistinguishable from fetal germ cells, and expressed germ-cell-specific proteins diagnostic of primordial germ cells. Alternatively, all iPSCs that exited tubules formed primitive tumors. iPSCs with AZF deletions produced significantly fewer GCLCs in vivo with distinct defects in gene expression. Findings indicate that xenotransplantation of human iPSCs directs germ cell differentiation in a manner dependent on donor genetic status.


Assuntos
Azoospermia/patologia , Fertilidade/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Túbulos Seminíferos/citologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Nus , Pele/citologia , Transplante Heterólogo/métodos
12.
PLoS One ; 9(4): e94231, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24718618

RESUMO

Data suggest that clinical applications of human induced pluripotent stem cells (hiPSCs) will be realized. Nonetheless, clinical applications will require hiPSCs that are free of exogenous DNA and that can be manufactured through Good Manufacturing Practice (GMP). Optimally, derivation of hiPSCs should be rapid and efficient in order to minimize manipulations, reduce potential for accumulation of mutations and minimize financial costs. Previous studies reported the use of modified synthetic mRNAs to reprogram fibroblasts to a pluripotent state. Here, we provide an optimized, fully chemically defined and feeder-free protocol for the derivation of hiPSCs using synthetic mRNAs. The protocol results in derivation of fully reprogrammed hiPSC lines from adult dermal fibroblasts in less than two weeks. The hiPSC lines were successfully tested for their identity, purity, stability and safety at a GMP facility and cryopreserved. To our knowledge, as a proof of principle, these are the first integration-free iPSCs lines that were reproducibly generated through synthetic mRNA reprogramming that could be putatively used for clinical purposes.


Assuntos
Técnicas de Cultura de Células/métodos , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , RNA Mensageiro/farmacologia , Adulto , Animais , Técnicas de Cultura de Células/normas , Diferenciação Celular , Linhagem Celular , Reprogramação Celular/efeitos dos fármacos , Corpos Embrioides , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Perfilação da Expressão Gênica , Camadas Germinativas/citologia , Proteínas de Fluorescência Verde/genética , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/transplante , Recém-Nascido , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Fator 3 de Transcrição de Octâmero/genética , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/síntese química , RNA Mensageiro/isolamento & purificação , Proteínas de Ligação a RNA/genética , Fatores de Transcrição SOXB1/genética , Pele/citologia , Teratoma/etiologia , Teratoma/patologia , Transfecção
13.
Stem Cell Res Ther ; 4(4): 87, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-23890092

RESUMO

INTRODUCTION: The reprogramming of a patient's somatic cells back into induced pluripotent stem cells (iPSCs) holds significant promise for future autologous cellular therapeutics. The continued presence of potentially oncogenic transgenic elements following reprogramming, however, represents a safety concern that should be addressed prior to clinical applications. The polycistronic stem cell cassette (STEMCCA), an excisable lentiviral reprogramming vector, provides, in our hands, the most consistent reprogramming approach that addresses this safety concern. Nevertheless, most viral integrations occur in genes, and exactly how the integration, epigenetic reprogramming, and excision of the STEMCCA reprogramming vector influences those genes and whether these cells still have clinical potential are not yet known. METHODS: In this study, we used both microarray and sensitive real-time PCR to investigate gene expression changes following both intron-based reprogramming and excision of the STEMCCA cassette during the generation of human iPSCs from adult human dermal fibroblasts. Integration site analysis was conducted using nonrestrictive linear amplification PCR. Transgene-free iPSCs were fully characterized via immunocytochemistry, karyotyping and teratoma formation, and current protocols were implemented for guided differentiation. We also utilized current good manufacturing practice guidelines and manufacturing facilities for conversion of our iPSCs into putative clinical grade conditions. RESULTS: We found that a STEMCCA-derived iPSC line that contains a single integration, found to be located in an intronic location in an actively transcribed gene, PRPF39, displays significantly increased expression when compared with post-excised stem cells. STEMCCA excision via Cre recombinase returned basal expression levels of PRPF39. These cells were also shown to have proper splicing patterns and PRPF39 gene sequences. We also fully characterized the post-excision iPSCs, differentiated them into multiple clinically relevant cell types (including oligodendrocytes, hepatocytes, and cardiomyocytes), and converted them to putative clinical-grade conditions using the same approach previously approved by the US Food and Drug Administration for the conversion of human embryonic stem cells from research-grade to clinical-grade status. CONCLUSION: For the first time, these studies provide a proof-of-principle for the generation of fully characterized transgene-free human iPSCs and, in light of the limited availability of current good manufacturing practice cellular manufacturing facilities, highlight an attractive potential mechanism for converting research-grade cell lines into putatively clinical-grade biologics for personalized cellular therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Diferenciação Celular , Reprogramação Celular , Expressão Gênica , Genômica , Humanos , Camundongos , Transgenes
14.
Mol Cell Biol ; 30(7): 1607-19, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20086097

RESUMO

Female mice lacking the transcription factor C/EBPbeta are infertile and display markedly reduced estrogen (E)-induced proliferation of the uterine epithelial lining during the reproductive cycle. The present study showed that E-stimulated luminal epithelial cells of a C/EBPbeta-null uterus are able to proceed through the G1 phase of the cell cycle before getting arrested in the S phase. This cell cycle arrest was accompanied by markedly reduced levels of expression of E2F3, an E2F family member, and a lack of nuclear localization of cyclin E, a critical regulator of cdk2. An increased nuclear accumulation of p27, an inhibitor of the cyclin E-cdk2 complex, was also observed for the mutant epithelium. Gene expression profiling of C/EBPbeta-null uterine epithelial cells revealed that the blockade of E-induced DNA replication triggers the activation of several well-known components of the DNA damage response pathway, such as ATM, ATR, histone H2AX, checkpoint kinase 1, and tumor suppressor p53. The activation of p53 by ATM/ATR kinase led to increased levels of expression of p21, an inhibitor of G1-S-phase progression, which helps maintain cell cycle arrest. Additionally, p53-dependent mechanisms contributed to an increased apoptosis of replication-defective cells in the C/EBPbeta-null epithelium. C/EBPbeta, therefore, is an essential mediator of E-induced growth and survival of uterine epithelial cells of cycling mice.


Assuntos
Apoptose/fisiologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Dano ao DNA , Replicação do DNA , Células Epiteliais/fisiologia , Útero/citologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Calnexina/metabolismo , Ciclo Celular/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Útero/efeitos dos fármacos
15.
Semin Reprod Med ; 28(1): 17-26, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20104425

RESUMO

In murine and human pregnancies, embryos implant by attaching to the luminal epithelium and invading into the stroma of the endometrium. Under the influence of the steroid hormones estrogen and progesterone, the stromal cells surrounding the implanting embryo undergo a remarkable transformation event. This process, known as decidualization, is an essential prerequisite for implantation. It comprises morphogenetic, biochemical, and vascular changes driven by the estrogen and progesterone receptors. The development of mutant mouse models lacking these receptors has firmly established the necessity of steroid signaling for decidualization. Genomic profiling of mouse and human endometrium has uncovered a complex yet highly conserved network of steroid-regulated genes that supports decidualization. To advance our understanding of the mechanisms regulating implantation and better address the clinical challenges of infertility and endometrial diseases such as endometriosis, it is important to integrate the information gained from the mouse and human models.


Assuntos
Decídua/fisiologia , Implantação do Embrião/fisiologia , Animais , Proteína Morfogenética Óssea 2/fisiologia , Conexina 43/fisiologia , Decídua/metabolismo , Modelos Animais de Doenças , Implantação do Embrião/genética , Endométrio/fisiologia , Feminino , Proteínas Homeobox A10 , Proteínas de Homeodomínio/fisiologia , Humanos , Interleucina-11/fisiologia , Camundongos , Chaperonas Moleculares/fisiologia , Gravidez , Receptores de Esteroides/fisiologia , Doenças Uterinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA