Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
J Cancer Res Clin Oncol ; 148(1): 225-236, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34110489

RESUMO

PURPOSE: 177Lu-Dotatate is an emerging treatment modality for patients with unresectable or metastatic well-differentiated NETs. This study examines survival predictors in patients who received 177Lu-Dotatate. METHODS: A retrospective single-center review was conducted, examining 47 individuals with progressive well-differentiated NETs treated with 177Lu-Dotatate (four induction cycles of 5.5 GBq at 10-week intervals followed by eight maintenance cycles of 3.7 GBq at 6-month intervals). RESULTS: Median follow-up was 63.1 months with a median progression-free survival (PFS) of 34.1 months. However, median overall survival (OS) was not reached at the time of analysis. The presence of ≥ 5 bone metastases (hazard ratio HR 4.33; p = 0.015), non-gastroenteropancreatic (non-GEP) NETs (HR 3.22; p = 0.025) and development of interim ascites (HR 3.15; p = 0.047) independently predicted a worse OS. Patients with chromogranin A of ≥ 4 × upper limit of normal (ULN) had shorter OS (p < 0.001) and PFS (p = 0.004). Similarly, those with pre-existing ascites demonstrated a worse OS (p = 0.009) and PFS (p = 0.026). Liver metastases involving greater than 50% liver volume and the existence of unusual metastatic locations had a negative impact on OS (p = 0.033) and PFS (p = 0.026), respectively. CONCLUSION: High burden of skeletal and hepatic metastases, non-GEP-NETs, chromogranin A of ≥ 4 × ULN, unusual metastatic sites, pre-existing and interim ascites are predictors of poor outcomes in patients treated with 177Lu-Dotatate. These common indicators can be used for the risk stratification and identification of patients most likely to benefit from PRRT. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02236910, Retrospectively registered on September, 2014.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias Hepáticas/secundário , Tumores Neuroendócrinos/mortalidade , Tumores Neuroendócrinos/radioterapia , Octreotida/análogos & derivados , Compostos Organometálicos/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Antieméticos/uso terapêutico , Ascite/mortalidade , Ascite/patologia , Biomarcadores Tumorais/análise , Neoplasias Ósseas/mortalidade , Cromogranina A/análise , Endoderma/patologia , Feminino , Humanos , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade , Crista Neural/patologia , Tumores Neuroendócrinos/patologia , Octreotida/efeitos adversos , Octreotida/uso terapêutico , Compostos Organometálicos/efeitos adversos , Intervalo Livre de Progressão , Compostos Radiofarmacêuticos/efeitos adversos , Compostos Radiofarmacêuticos/uso terapêutico , Estudos Retrospectivos
2.
Stem Cell Reports ; 15(1): 242-255, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32619491

RESUMO

Individuals with the genetic disorder alpha-1 antitrypsin deficiency (AATD) are at risk of developing lung and liver disease. Patient induced pluripotent stem cells (iPSCs) have been found to model features of AATD pathogenesis but only a handful of AATD patient iPSC lines have been published. To capture the significant phenotypic diversity of the patient population, we describe here the establishment and characterization of a curated repository of AATD iPSCs with associated disease-relevant clinical data. To highlight the utility of the repository, we selected a subset of iPSC lines for functional characterization. Selected lines were differentiated to generate both hepatic and lung cell lineages and analyzed by RNA sequencing. In addition, two iPSC lines were targeted using CRISPR/Cas9 editing to accomplish scarless repair. Repository iPSCs are available to investigators for studies of disease pathogenesis and therapeutic discovery.


Assuntos
Acesso à Informação , Bases de Dados como Assunto , Células-Tronco Pluripotentes Induzidas/patologia , Deficiência de alfa 1-Antitripsina/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sistemas CRISPR-Cas/genética , Diferenciação Celular , Linhagem da Célula , Endoderma/patologia , Feminino , Edição de Genes , Loci Gênicos , Genótipo , Hepatócitos/patologia , Humanos , Pulmão/diagnóstico por imagem , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Mutação/genética , Fenótipo , Transcriptoma/genética , alfa 1-Antitripsina/genética , Deficiência de alfa 1-Antitripsina/diagnóstico por imagem , Deficiência de alfa 1-Antitripsina/genética
3.
World Neurosurg ; 143: 28-32, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32702494

RESUMO

BACKGROUND: Intracranial endodermal cysts are congenital lesions that generally develop in the cerebellopontine angle and ventral brainstem of the posterior fossa, whereas endodermal cysts in the quadrigeminal cistern are very rare. We report a rare case of an endodermal cyst in the quadrigeminal cistern with a non-enhancing nodule that developed in patient over 80 years of age. CASE DESCRIPTION: An 85-year-old man presented to our hospital with progressing gait disturbance and urinary incontinence. Preoperative images showed a cystic mass lesion with a nodule in the quadrigeminal cistern and hydrocephalus. There was no enhanced portion in the lesion, and the intensity of the cyst on magnetic resonance imaging revealed a high protein concentration. Subtotal resection was performed due to the adhesion of the cyst to the brainstem. It was diagnosed as an endodermal cyst. The postoperative course was uneventful, and hydrocephalus improved. CONCLUSIONS: This is a rare case of an intracranial endodermal cyst in terms of location and age of onset compared with previous reports. This case demonstrates that endodermal cysts should be considered as a differential diagnosis for lesions in the quadrigeminal cistern with high protein concentration in the cyst and nodule representing chronic inflammation, regardless of enhancing effects.


Assuntos
Cistos do Sistema Nervoso Central/diagnóstico por imagem , Endoderma/patologia , Espaço Subaracnóideo/diagnóstico por imagem , Idoso de 80 Anos ou mais , Cistos do Sistema Nervoso Central/complicações , Cistos do Sistema Nervoso Central/patologia , Cistos do Sistema Nervoso Central/cirurgia , Transtornos Neurológicos da Marcha/etiologia , Transtornos Neurológicos da Marcha/fisiopatologia , Humanos , Hidrocefalia/diagnóstico por imagem , Hidrocefalia/etiologia , Hidrocefalia/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Espaço Subaracnóideo/cirurgia , Incontinência Urinária/etiologia , Incontinência Urinária/fisiopatologia
4.
Congenit Anom (Kyoto) ; 59(3): 74-80, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30554442

RESUMO

Development of external genitalia and perineum is the subject of developmental biology as well as toxicology and teratology researches. Cloaca forms in the lower (caudal) end of endoderm. Such endodermal epithelia and surrounding mesenchyme interact with various signals to form the external genitalia. External genitalia (the anlage termed as genital tubercle: GT) formation shows prominent sexually dimorphic morphogenesis in late embryonic stages, which is an unexplored developmental research field because of many reasons. External genitalia develop adjacent to the cloaca which develops urethra and corporal bodies. Developmental regulators including growth factor signals are necessary for epithelia-mesenchyme interaction (EMI) in posterior embryos including the cloaca and urethra in the genitalia. In the case of male type urethra, formation of tubular urethra proceeds from the lower (ventral) side of external genitalia as a masculinization process in contrast to the case of female urethra. Mechanisms for its development are not elucidated yet due to the lack of suitable mutant mouse models. Because of the recent progresses of Cre (recombinase)-mediated conditional target gene modification analyses, many developmental regulatory genes become increasingly analyzed. Conditional gene knockout mouse approaches and tissue lineage approaches are expected to offer vital information for such sexually dimorphic developmental processes. This review aims to offer recent updates on the progresses of these emerging developmental processes for the research field of congenital anomalies.


Assuntos
Anormalidades Congênitas/genética , Regulação da Expressão Gênica no Desenvolvimento , Genitália/embriologia , Organogênese/genética , Períneo/embriologia , Animais , Anormalidades Congênitas/metabolismo , Anormalidades Congênitas/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Endoderma/crescimento & desenvolvimento , Endoderma/metabolismo , Endoderma/patologia , Feminino , Genitália/metabolismo , Genitália/patologia , Humanos , Fator de Transcrição MafB/genética , Fator de Transcrição MafB/metabolismo , Masculino , Camundongos , Camundongos Knockout , Períneo/patologia , Caracteres Sexuais , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Via de Sinalização Wnt
5.
Stem Cells ; 37(3): 306-317, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30471152

RESUMO

Directed differentiation of human induced pluripotent stem cells (iPSCs) toward hepatobiliary lineages has been increasingly used as models of human liver development/diseases. As protein kinases are important components of signaling pathways regulating cell fate changes, we sought to define the key molecular mediators regulating human liver development using inhibitors targeting tyrosine kinases during hepatic differentiation of human iPSCs. A library of tyrosine kinase inhibitors was used for initial screening during the multistage differentiation of human iPSCs to hepatic lineage. Among the 80 kinase inhibitors tested, only Src inhibitors suppressed endoderm formation while none had significant effect on later stages of hepatic differentiation. Transient inhibition of c-Src during endodermal induction of human iPSCs reduced endodermal commitment and expression of endodermal markers, including SOX17 and FOXA2, in a dose-dependent manner. Interestingly, the transiently treated cells later developed into profibrogenic cholangiocyte-like cells expressing both cholangiocyte markers, such as CK7 and CK19, and fibrosis markers, including Collagen1 and smooth muscle actin. Further analysis of these cells revealed colocalized expression of collagen and yes-associated protein (YAP; a marker associated with bile duct proliferation/fibrosis) and an increased production of interleukin-6 and tumor necrosis factor-α. Moreover, treatment with verteporfin, a YAP inhibitor, significantly reduced expression of fibrosis markers. In summary, these results suggest that c-Src has a critical role in cell fate determination during endodermal commitment of human iPSCs, and its alteration in early liver development in human may lead to increased production of abnormal YAP expressing profibrogenic proinflammatory cholangiocytes, similar to those seen in livers of patients with biliary fibrosis. Stem Cells 2019;37:306-317.


Assuntos
Proteína Tirosina Quinase CSK/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Endoderma/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Ductos Biliares/enzimologia , Ductos Biliares/patologia , Proteína Tirosina Quinase CSK/metabolismo , Endoderma/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/enzimologia , Células-Tronco Pluripotentes Induzidas/patologia , Fígado/enzimologia , Fígado/patologia
6.
Dev Cell ; 44(6): 679-693.e5, 2018 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-29587142

RESUMO

We show that the loss or gain of transcription factor programs that govern embryonic cell-fate specification is associated with a form of tumor plasticity characterized by the acquisition of alternative cell fates normally characteristic of adjacent organs. In human non-small cell lung cancers, downregulation of the lung lineage-specifying TF NKX2-1 is associated with tumors bearing features of various gut tissues. Loss of Nkx2-1 from murine alveolar, but not airway, epithelium results in conversion of lung cells to gastric-like cells. Superimposing oncogenic Kras activation enables further plasticity in both alveolar and airway epithelium, producing tumors that adopt midgut and hindgut fates. Conversely, coupling Nkx2-1 loss with foregut lineage-specifying SOX2 overexpression drives the formation of squamous cancers with features of esophageal differentiation. These findings demonstrate that elements of pathologic tumor plasticity mirror the normal developmental history of organs in that cancer cells acquire cell fates associated with developmentally related neighboring organs.


Assuntos
Linhagem da Célula , Neoplasias Esofágicas/patologia , Neoplasias Pulmonares/patologia , Fatores de Transcrição SOXB1/metabolismo , Neoplasias Gástricas/patologia , Fator Nuclear 1 de Tireoide/metabolismo , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/patologia , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Diferenciação Celular , Plasticidade Celular , Desenvolvimento Embrionário , Endoderma/metabolismo , Endoderma/patologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prognóstico , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Taxa de Sobrevida , Fator Nuclear 1 de Tireoide/genética
7.
Annu Rev Genet ; 51: 123-141, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29178821

RESUMO

Genetic mosaicism arises when a zygote harbors two or more distinct genotypes, typically due to de novo, somatic mutation during embryogenesis. The clinical manifestations largely depend on the differentiation status of the mutated cell; earlier mutations target pluripotent cells and generate more widespread disease affecting multiple organ systems. If gonadal tissue is spared-as in somatic genomic mosaicism-the mutation and its effects are limited to the proband, whereas mosaicism also affecting the gametes, such as germline or gonosomal mosaicism, is transmissible. Mosaicism is easily appreciated in cutaneous disorders, as phenotypically distinct mutant cells often give rise to lesions in patterns determined by the affected cell type. Genetic investigation of cutaneous mosaic disorders has identified pathways central to disease pathogenesis, revealing novel therapeutic targets. In this review, we discuss examples of cutaneous mosaicism, approaches to gene discovery in these disorders, and insights into molecular pathobiology that have potential for clinical translation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mosaicismo , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Dermatopatias Genéticas/genética , Ectoderma/metabolismo , Ectoderma/patologia , Embrião de Mamíferos , Endoderma/metabolismo , Endoderma/patologia , Humanos , Queratina-1/genética , Queratina-1/metabolismo , Queratina-10/genética , Queratina-10/metabolismo , Microdissecção e Captura a Laser , Mesoderma/metabolismo , Mesoderma/patologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Dermatopatias Genéticas/metabolismo , Dermatopatias Genéticas/patologia , Fatores de Tempo , Sequenciamento do Exoma
8.
Stem Cell Res ; 18: 60-63, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28395807

RESUMO

We describe here the reprogramming of CD34+ cells isolated from umbilical cord blood obtained after full term delivery of a healthy female child of Indian origin. The cells were nucleofected by episomal vectors expressing Oct4, Sox2, L-Myc, Klf4, Lin28 and p53DD (negative mutation in p53). Colonies were identified by alkaline phosphatase staining and characterized for expression of pluripotency markers at protein level by immunofluorescence, flow cytometry and at transcript level by PCR. Genomic stability of the cell line was checked by G-banded karyotype. The ability to differentiate to endoderm, mesoderm and ectoderm in vitro was confirmed by immunofluorescence staining.


Assuntos
Antígenos CD34/metabolismo , Reprogramação Celular , Sangue Fetal/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Antígenos CD34/genética , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Ectoderma/metabolismo , Ectoderma/patologia , Endoderma/metabolismo , Endoderma/patologia , Feminino , Sangue Fetal/metabolismo , Humanos , Índia , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariótipo , Fator 4 Semelhante a Kruppel , Mesoderma/metabolismo , Mesoderma/patologia , Microscopia de Fluorescência
9.
Cell Rep ; 19(4): 671-679, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28445719

RESUMO

Although histone-modifying enzymes are generally assumed to function in a manner dependent on their enzymatic activities, this assumption remains untested for many factors. Here, we show that the Tip60 (Kat5) lysine acetyltransferase (KAT), which is essential for embryonic stem cell (ESC) self-renewal and pre-implantation development, performs these functions independently of its KAT activity. Unlike ESCs depleted of Tip60, KAT-deficient ESCs exhibited minimal alterations in gene expression, chromatin accessibility at Tip60 binding sites, and self-renewal, thus demonstrating a critical KAT-independent role of Tip60 in ESC maintenance. In contrast, KAT-deficient ESCs exhibited impaired differentiation into mesoderm and endoderm, demonstrating a KAT-dependent function in differentiation. Consistent with this phenotype, KAT-deficient mouse embryos exhibited post-implantation developmental defects. These findings establish separable KAT-dependent and KAT-independent functions of Tip60 in ESCs and during differentiation, revealing a complex repertoire of regulatory functions for this essential chromatin remodeling complex.


Assuntos
Autorrenovação Celular/fisiologia , Lisina Acetiltransferase 5/metabolismo , Transativadores/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Endoderma/metabolismo , Endoderma/patologia , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/química , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Lisina Acetiltransferase 5/deficiência , Lisina Acetiltransferase 5/genética , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Regiões Promotoras Genéticas , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Transativadores/deficiência , Transativadores/genética
10.
Biochem Cell Biol ; 95(2): 251-262, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28177772

RESUMO

Mouse F9 cells differentiate into primitive endoderm (PrE) following the activation of the canonical WNT-ß-catenin pathway. The upregulation of Wnt6 and activation of ß-catenin-TCF-LEF-dependent transcription is known to accompany differentiation, but the Frizzled (FZD) receptor responsible for transducing the WNT6 signal is not known. Eight of the 10 Fzd genes were found to be expressed in F9 cells, with Fzd7 being the most highly expressed, and chosen for further analysis. To alter steady-state Fzd7 levels and test the effect this has on differentiation, siRNA and overexpression approaches were used to knock-down and ectopically express the Fzd7 message, respectively. siRNA knock-down of Fzd7 resulted in reduced DAB2 levels, and the overexpression activated a TCF-LEF reporter, but neither approach affected differentiation. Our focus turned to how canonical WNT6 signaling was attenuated to allow PrE cells to form parietal endoderm (PE). Dkk1, encoding a WNT antagonist, was examined and results showed that its expression increased in F9 cells treated with retinoic acid (RA) or overexpressing Wnt6. F9 cells overexpressing human DKK1 or treated with DKK1-conditioned medium and then treated with RA failed to differentiate, indicating that a negative feedback loop involving WNT6 and DKK1 attenuates canonical WNT-ß-catenin signaling, thereby allowing PE cells to differentiate.


Assuntos
Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Proto-Oncogênicas/genética , Receptores Acoplados a Proteínas G/genética , Teratocarcinoma/genética , Proteínas Wnt/genética , beta Catenina/genética , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Endoderma/metabolismo , Endoderma/patologia , Retroalimentação Fisiológica , Receptores Frizzled , Genes Reporter , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Teratocarcinoma/metabolismo , Teratocarcinoma/patologia , Tretinoína/farmacologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
11.
Mol Cell Endocrinol ; 457: 20-34, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-28111205

RESUMO

Epidemiological and experimental evidence associates the exposure to Bisphenol A with the increase of cancer risk in several organs, including prostate. BPA targets different pathways involved in carcinogenicity including the Nuclear Receptors (i.e. estrogen and androgen receptors), stress regulated proteins and, finally, epigenetic changes. Here, we analyse BPA-dependent carcinogenesis in endoderm-derived glands, thyroid, liver, pancreas and prostate focusing on cell signalling, DNA damage repair pathways and epigenetic modifications. Mainly, we gather molecular data evidencing harmful effects at doses relevant for human risk (low-doses). Since few molecular data are available, above all for the pancreas, we analysed transcriptomic data generated in our laboratory to suggest possible mechanisms of BPA carcinogenicity in endoderm-derived glands, discussing the role of nuclear receptors and stress/NF-kB pathways. We evidence that an in vitro toxicogenomic approach might suggest mechanisms of toxicity applicable to cells having the same developmental origin. Although we cannot draw firm conclusions, published data summarized in this review suggest that exposure to BPA, primarily during the developmental stages, represents a risk for carcinogenesis of endoderm-derived glands.


Assuntos
Compostos Benzidrílicos/toxicidade , Carcinogênese/genética , Carcinogênese/patologia , Endoderma/patologia , Exposição Ambiental/análise , Fenóis/toxicidade , Animais , Humanos , Neoplasias/genética , Neoplasias/patologia , Fatores de Risco
12.
J Cell Physiol ; 232(10): 2616-2625, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27306424

RESUMO

Human-induced pluripotent stem cells (hiPSCs) can potentially serve as an invaluable source for cell replacement therapy and allow the creation of patient- and disease-specific stem cells without the controversial use of embryos and avoids any immunological incompatibility. The generation of insulin-producing pancreatic ß-cells from pluripotent stem cells in vitro provides an unprecedented cell source for personal drug discovery and cell transplantation therapy in diabetes. A new five-step protocol was introduced in this study, effectively induced hiPSCs to differentiate into glucose-responsive insulin-producing cells. This process mimics in vivo pancreatic organogenesis by directing cells through stages resembling definitive endoderm, primitive gut-tube endoderm, posterior foregut, pancreatic endoderm, and endocrine precursor. Each stage of differentiation were characterized by stage-specific markers. The produced cells exhibited many properties of functional ß-cells, including expression of critical ß-cells transcription factors, the potency to secrete C-peptide in response to high levels of glucose and the presence of mature endocrine secretory granules. This high efficient differentiation protocol, established in this study, yielded 79.18% insulin-secreting cells which were responsive to glucose five times higher than the basal level. These hiPSCs-derived glucose-responsive insulin-secreting cells might provide a promising approach for the treatment of type I diabetes mellitus. J. Cell. Physiol. 232: 2616-2625, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular , Linhagem da Célula , Diabetes Mellitus Tipo 1/metabolismo , Endoderma/metabolismo , Fibroblastos/metabolismo , Glucose/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Separação Celular/métodos , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Endoderma/patologia , Células Alimentadoras , Fibroblastos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Secreção de Insulina , Células Secretoras de Insulina/patologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Nus , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Organogênese , Fenótipo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Teratoma/genética , Teratoma/metabolismo , Teratoma/patologia , Transfecção
13.
Cell Death Differ ; 24(1): 98-110, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27834953

RESUMO

Cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-regulated anion channel capable of conducting both Cl- and HCO3-, mutations of which cause cystic fibrosis (CF), a common autosomal recessive disease. Although CF patients are known to have varied degree of developmental problems, the biological role of CFTR in embryonic development remains elusive. Here, we show that CFTR is functionally expressed in mouse ESCs. CFTR-/- mESCs exhibit dramatic defect in mesendoderm differentiation. In addition, CFTR physically interacts with ß-catenin, defect of which leads to premature degradation of ß-catenin and suppressed activation of ß-catenin signaling. Furthermore, knockdown of CFTR retards the early development of Xenopus laevis with impaired mesoderm/endoderm differentiation and ß-catenin signaling. Our study reveals a previously undefined role of CFTR in controlling ESC differentiation and early embryonic development via its interaction with ß-catenin, and provides novel insights into the understanding of embryonic development.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Cloretos/análise , Colforsina/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Ectoderma/metabolismo , Ectoderma/patologia , Embrião não Mamífero/fisiologia , Desenvolvimento Embrionário , Endoderma/metabolismo , Endoderma/patologia , Feminino , Masculino , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Camundongos Knockout , Células-Tronco Embrionárias Murinas , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt3A/metabolismo , Proteínas de Xenopus/antagonistas & inibidores , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/crescimento & desenvolvimento
14.
Genes Dev ; 30(5): 535-52, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26944679

RESUMO

Faithful execution of developmental programs relies on the acquisition of unique cell identities from pluripotent progenitors, a process governed by combinatorial inputs from numerous signaling cascades that ultimately dictate lineage-specific transcriptional outputs. Despite growing evidence that metabolism is integrated with many molecular networks, how pathways that control energy homeostasis may affect cell fate decisions is largely unknown. Here, we show that AMP-activated protein kinase (AMPK), a central metabolic regulator, plays critical roles in lineage specification. Although AMPK-deficient embryonic stem cells (ESCs) were normal in the pluripotent state, these cells displayed profound defects upon differentiation, failing to generate chimeric embryos and preferentially adopting an ectodermal fate at the expense of the endoderm during embryoid body (EB) formation. AMPK(-/-) EBs exhibited reduced levels of Tfeb, a master transcriptional regulator of lysosomes, leading to diminished endolysosomal function. Remarkably, genetic loss of Tfeb also yielded endodermal defects, while AMPK-null ESCs overexpressing this transcription factor normalized their differential potential, revealing an intimate connection between Tfeb/lysosomes and germ layer specification. The compromised endolysosomal system resulting from AMPK or Tfeb inactivation blunted Wnt signaling, while up-regulating this pathway restored expression of endodermal markers. Collectively, these results uncover the AMPK pathway as a novel regulator of cell fate determination during differentiation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Lisossomos/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Células-Tronco Embrionárias , Endoderma/patologia , Camundongos , Mutação , Transdução de Sinais/genética , Via de Sinalização Wnt/genética
15.
Cell Rep ; 8(6): 1989-2002, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25242323

RESUMO

Setd2 is known as a histone-H3K36-specific methyltransferase. However, its role in physiological function remains unclear. In this study, we show that Setd2 mainly regulates differentiation of murine embryonic stem cells (mESCs) toward primitive endoderm. Furthermore, we show that downregulated endoderm-related genes in Setd2(-/-) mESCs are associated with an aberrantly low level of Erk activity and that enforced expression of Fgfr3 can rescue the defective Erk pathway in Setd2(-/-) mESCs. Interestingly, the transcriptional initiation of Fgfr3 is directly regulated through histone H3K36me3 modification in its distal promoter region by Setd2. These results indicate that Setd2 controls the primitive endoderm differentiation of mESCs by regulating the Fgfr3-Erk signaling.


Assuntos
Células-Tronco Embrionárias/citologia , Endoderma/patologia , Histona-Lisina N-Metiltransferase/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Imunoprecipitação da Cromatina , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Endoderma/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Histona-Lisina N-Metiltransferase/deficiência , Histona-Lisina N-Metiltransferase/genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Teratoma/patologia , Transcrição Gênica
16.
Dev Biol ; 393(1): 33-43, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25009997

RESUMO

Calumin is an endoplasmic reticulum (ER)-transmembrane protein, and little is known about its physiological roles. Here we showed that calumin homozygous mutant embryos die at embryonic days (E) 10.5-11.5. At mid-gestation, calumin was expressed predominantly in the yolk sac. Apoptosis was enhanced in calumin homozygous mutant yolk sacs at E9.5, pointing to a possible link to the embryonic lethality. Calumin co-immunoprecipitated with ERAD components such as p97, BIP, derlin-1, derlin-2 and VIMP, suggesting its involvement in ERAD. Indeed, calumin knockdown in HEK 293 cells resulted in ERAD being less efficient, as demonstrated by attenuation in both degradations of a misfolded α1-antitrypsin variant and the ER-to-cytosol dislocation of cholera toxin A1 subunit. In calumin homozygous mutant yolk sac endoderm cells, ER stress-associated alterations were observed, including lipid droplet accumulation, fragmentation of the ER and dissociation of ribosomes from the ER. In this context, the ER-overload response, assumed to be cytoprotective, was also triggered in the mutant endoderm cells, but seemed to fully counteract the excessive ER stress generated due to defective ERAD. Taken together, our findings suggested that calumin serves to maintain the yolk sac integrity through participation in the ERAD activity, contributing to embryonic development.


Assuntos
Degradação Associada com o Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/fisiologia , Saco Vitelino/metabolismo , Animais , Apoptose/genética , Linhagem Celular , Toxina da Cólera/metabolismo , Desenvolvimento Embrionário/genética , Endoderma/citologia , Endoderma/patologia , Estresse do Retículo Endoplasmático/genética , Células HEK293 , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Dobramento de Proteína , Interferência de RNA , RNA Interferente Pequeno , alfa 1-Antitripsina/metabolismo
17.
J Neurosurg Pediatr ; 12(3): 284-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23848288

RESUMO

Endodermal cyst is a rare developmental cyst of the CNS, such as a Rathke cleft and colloid cyst lined by columnar epithelium of presumed endodermal origin. Intracranial endodermal cysts are rare, and most are found in the posterior fossa. The authors report a case of petroclival endodermal cyst with extensive bone destruction. A 12-year-old boy presented with transient facial weakness and headache. Imaging revealed a 3 × 3 × 4-cm, partial rim, enhanced cystic lesion in the petroclival area that was isointense on T1-weighted imaging and hyperintense in T2-weighted imaging. The cyst wall was partially removed and the cyst was obliterated using a lateral approach. Histological examination revealed ciliated, simple-to-pseudostratified cuboidal epithelium with a basement membrane that was consistent with an endodermal cyst, with the rare finding of xanthogranulomatous changes.


Assuntos
Cistos do Sistema Nervoso Central/diagnóstico , Endoderma/patologia , Osso Petroso/patologia , Xantogranuloma Juvenil/diagnóstico , Cistos do Sistema Nervoso Central/patologia , Criança , Humanos , Masculino , Tomografia Computadorizada por Raios X , Xantogranuloma Juvenil/patologia
18.
Cell Transplant ; 22(5): 821-30, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22472700

RESUMO

Development of ß-cells from human embryonic stem cells (hESCs) could compensate for the shortage of islet donors required for diabetes therapy. Although pancreatic progenitors have been derived from hESCs using various protocols, no fully functional b-cells could be generated in vitro. We evaluated the in vivo growth and differentiation of PDX1+ pancreatic endoderm cells obtained from hESCs. Here we show site-specific survival and differentiation when comparing cells grafted in the epididymal fat pad or the subcutaneous space of NOD/SCID mice after 12 weeks follow-up. Subcutaneous grafts persisted and expressed PDX1 at all time points analyzed, showed PDX1 and NKX6.1 coexpression after 6 weeks, and contained NGN3+ cells after 12 weeks.These findings suggest that further specification along the pancreatic lineage occured at the subcutaneous site.In sharp contrast, in the fat pad grafts only a minority of PDX1+ cells remained after 2 weeks, and no further pancreatic differentiation was observed later on. In addition, contaminating mesenchymal cells present in the implants further developed into cartilage tissue after 6 weeks implantation in the fat pad, but not in the subcutaneous space. These findings indicate that the in vivo microenvironment plays a critical role in the further differentiation of transplanted pancreatic endoderm cells.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Endoderma/metabolismo , Pâncreas/metabolismo , Tecido Adiposo/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Células Cultivadas , Células-Tronco Embrionárias/transplante , Endoderma/citologia , Endoderma/patologia , Epididimo/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Injeções Subcutâneas , Células Secretoras de Insulina/citologia , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/patologia , Transativadores/metabolismo
19.
Reprod Biomed Online ; 25(3): 315-21, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22796234

RESUMO

This study has documented the major types of lineage progenitor cells at the second level of cell differentiation after the establishment of the primary germ layers in ectopic human embryos in vivo. These correspond to stages 8 and 9 of embryogenesis (weeks 3-4) in the Carnegie collection. The aim of this study was to provide images of fine structure of tissue progenitor cells to compare them with current imaging of their equivalent stem cells identified using fluorescent stem cell markers. These include neural, mesenchymal, endodermal, ectodermal (epidermal) and haematopoietic progenitor cells, including those for amniotic, yolk sac and chorionic tissues that are used in current stem cell research. Neural induction by the notochord has been imaged. This study should give valuable clues to understand the pattern of cell differentiation of embryonic stem cells (ESC) in vitro, which are more or less mimicked in ESC colonies, embryoid bodies and neurospheres as documented in the literature. The fine structure of week-3 and week-4 human ectopic embryos is presented to demonstrate progenitor tissue cells that will eventually form the brain, spinal cord, skin, gut, heart, blood, muscle, bone and other tissues of the human body later on in development. These images should help stem cell researchers using fluorescent markers and other techniques to identify embryonic and adult stem cells in culture.


Assuntos
Células-Tronco/citologia , Diferenciação Celular , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Endoderma/patologia , Epiderme/patologia , Feminino , Corantes Fluorescentes/farmacologia , Camadas Germinativas/patologia , Células-Tronco Hematopoéticas/citologia , Humanos , Mesoderma/patologia , Microscopia Eletrônica de Transmissão/métodos , Notocorda/patologia , Gravidez , Gravidez Ectópica , Somitos/patologia , Distribuição Tecidual , Saco Vitelino/citologia
20.
Stem Cells Dev ; 21(7): 1134-44, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21793658

RESUMO

Parthenogenetic embryonic stem cells (P-ESCs) offer an alternative source of pluripotent cells, which hold great promise for autologous transplantation and regenerative medicine. P-ESCs have been successfully derived from blastocysts of several mammalian species. However, compared with biparental embryonic stem cells (B-ESCs), P-ESCs are limited in their ability to fully differentiate into all 3 germ layers. For example, it has been observed that there is a differentiation bias toward ectoderm derivatives at the expense of endoderm and mesoderm derivatives-muscle in particular-in chimeric embryos, teratomas, and embryoid bodies. In the present study we found that H19 expression was highly upregulated in P-ESCs with more than 6-fold overexpression compared with B-ESCs. Thus, we hypothesized that manipulation of the H19 gene in P-ESCs would alleviate their limitations and allow them to function like B-ESCs. To test this hypothesis we employed a small hairpin RNA approach to reduce the amount of H19 transcripts in mouse P-ESCs. We found that downregulation of H19 led to an increase of mesoderm-derived muscle and endoderm in P-ESCs teratomas similar to that observed in B-ESCs teratomas. This phenomenon coincided with upregulation of mesoderm-specific genes such as Myf5, Myf6, and MyoD. Moreover, H19 downregulated P-ESCs differentiated into a higher percentage of beating cardiomyocytes compared with control P-ESCs. Collectively, these results suggest that P-ESCs are amenable to molecular modifications that bring them functionally closer to true ESCs.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Partenogênese , RNA não Traduzido/genética , Animais , Células Cultivadas , Ilhas de CpG/genética , Metilação de DNA , Regulação para Baixo , Ectoderma/metabolismo , Ectoderma/patologia , Corpos Embrioides/metabolismo , Corpos Embrioides/fisiologia , Células-Tronco Embrionárias/transplante , Endoderma/metabolismo , Endoderma/patologia , Endoderma/fisiologia , Feminino , Perfilação da Expressão Gênica , Genes Transgênicos Suicidas , Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Cariótipo , Mesoderma/patologia , Mesoderma/fisiologia , Camundongos , Músculos/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , RNA Longo não Codificante , RNA não Traduzido/metabolismo , Teratoma/metabolismo , Teratoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA