Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Blood ; 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38820590

RESUMEN

Deficiency of X-linked Inhibitor of Apoptosis Protein (XIAP) is a rare genetic condition that can present with recurrent episodes of hemophagocytic lymphohistiocytosis (HLH), though the exact mechanisms leading to this hyperinflammatory disorder are unclear. Understanding its biology is critical to developing targeted therapies for this potentially fatal disease. Here we report on a novel multi-exonic intragenic duplication leading to XIAP deficiency with recurrent HLH that demonstrated complete response to interleukin (IL)-1b blockade. We further demonstrate using both primary patient cells and genetically modified THP-1 monocyte cell lines that, contrary to what has previously been shown in mouse cells, XIAP-deficient human macrophages do not produce excess IL-1b when stimulated under standard conditions. Instead, NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated hyperproduction of IL-1b is observed only when the XIAP-deficient cells are stimulated under autophagy-promoting conditions and this correlates with defective autophagic flux as measured by decreased accumulation of the early autophagy marker LC3-II. This work therefore highlights IL-1b blockade as a therapeutic option for patients with XIAP deficiency experiencing recurrent HLH and identifies a critical role for XIAP in promoting autophagy as a means of limiting IL-1b-mediated hyperinflammation during periods of cellular stress.

2.
J Cell Physiol ; 232(10): 2616-2625, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27306424

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Diabetes Mellitus Tipo 1/metabolismo , Endodermo/metabolismo , Fibroblastos/metabolismo , Glucosa/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Separación Celular/métodos , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Endodermo/patología , Células Nutrientes , Fibroblastos/patología , Regulación del Desarrollo de la Expresión Génica , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/patología , Secreción de Insulina , Células Secretoras de Insulina/patología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Desnudos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Organogénesis , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Transfección
3.
J Cell Physiol ; 231(9): 1994-2006, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26755186

RESUMEN

The generation of definitive endoderm (DE) from pluripotent stem cells (PSCs) is a fundamental stage in the formation of highly organized visceral organs, such as the liver and pancreas. Currently, there is a need for a comprehensive study that illustrates the involvement of different signaling pathways and their interactions in the derivation of DE cells from PSCs. This study aimed to identify signaling pathways that have the greatest influence on DE formation using analyses of transcriptional profiles, protein-protein interactions, protein-DNA interactions, and protein localization data. Using this approach, signaling networks involved in DE formation were constructed using systems biology and data mining tools, and the validity of the predicted networks was confirmed experimentally by measuring the mRNA levels of hub genes in several PSCs-derived DE cell lines. Based on our analyses, seven signaling pathways, including the BMP, ERK1-ERK2, FGF, TGF-beta, MAPK, Wnt, and PIP signaling pathways and their interactions, were found to play a role in the derivation of DE cells from PSCs. Lastly, the core gene regulatory network governing this differentiation process was constructed. The results of this study could improve our understanding surrounding the efficient generation of DE cells for the regeneration of visceral organs. J. Cell. Physiol. 231: 1994-2006, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Endodermo/citología , Redes Reguladoras de Genes , Páncreas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Activinas/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias Humanas/citología , Humanos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
4.
Cell Immunol ; 293(2): 59-66, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25569483

RESUMEN

Human Wharton's Jelly-derived Mesenchymal Stem Cells (hWJ-MSCs) are considered as an alternative for bone-marrow-derived MSCs. These cells have immunosuppressive properties. It was unclear whether the WJ-MSCs would sustain their immunomodulatory characteristics after lentiviral transduction or not. In this study, we evaluated immunomodulatory properties of WJ-MSCs after lentiviral transduction. HWJ-MSCs were transduced with lentiviral particles. Expression of transduced and un-transduced hWJ-MSCs surface molecules and secretion of IL-10, HGF, VEGF and TGF-ß was analyzed. Cell proliferation and frequency of CD4(+)CD25(+) CD127(low/neg) Foxp3(+) T regulatory cells was measured. There was no difference between the surface markers and secretion of IL-10, HGF, VEGF and TGF-ß in transduced and un-transduced hWJ-MSCs. Both cells inhibited the proliferation of PHA stimulated PBMCs, and improved the frequency of T regulatory cells. These findings suggest that lentiviral transduction does not alter the immunomodulatory function of hWJ-MSCs. However, lentiviral transduction may have a wide range of applications in gene therapy.


Asunto(s)
Diferenciación Celular/inmunología , Factores Inmunológicos/inmunología , Células Madre Mesenquimatosas/inmunología , Gelatina de Wharton/citología , Femenino , Citometría de Flujo , Factor de Crecimiento de Hepatocito/análisis , Factor de Crecimiento de Hepatocito/inmunología , Humanos , Factores Inmunológicos/genética , Interleucina-10/análisis , Interleucina-10/inmunología , Lentivirus/genética , Leucocitos Mononucleares , Células Madre Mesenquimatosas/citología , Embarazo , Transducción Genética , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/inmunología , Factor A de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/inmunología , Gelatina de Wharton/inmunología
5.
Cell Biol Int ; 39(5): 591-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25640312

RESUMEN

The first step in the formation of hepatocytes and beta cells is the generation of definitive endoderm (DE) which involves a central issue in developmental biology. Human induced pluripotent stem cells (hiPSCs) have the pluripotency to differentiate into all three germ layers in vitro and have been considered potent candidates for regenerative medicine as an unlimited source of cells for therapeutic applications. In this study, we investigated the differentiating potential of hiPSCs on poly (ε-caprolactone) (PCL) nanofibrous scaffold into DE cells. Here, we demonstrate directed differentiation of hiPSCs by factors such as Activin A and Wnt3a. The differentiation was determined by immunofluoresence staining with Sox17, FoxA2 and Goosecoid (Gsc) and also by qRT-PCR analysis. The results of this study showed that hiPSCs, as a new cell source, have the ability to differentiate into DE cells with a high capacity and also demonstrate that three dimension (3D) culture provides a suitable nanoenviroment for growth, proliferation and differentiation of hiPSCs. PCL nanofibrous scaffold with essential supplements, stimulating factors and EB-derived cells is able to provide a novel method for enhancing functional differentiation of hiPSCs into DE cells.


Asunto(s)
Activinas/farmacología , Diferenciación Celular/efectos de los fármacos , Endodermo/efectos de los fármacos , Poliésteres/química , Andamios del Tejido/química , Proteína Wnt3A/farmacología , Animales , Células Cultivadas , Galvanoplastia , Endodermo/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Ensayo de Materiales , Ratones , Nanofibras/química , Técnicas de Cultivo de Tejidos/instrumentación , Técnicas de Cultivo de Tejidos/métodos
6.
Exp Cell Res ; 322(1): 51-61, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24434355

RESUMEN

Due to pluripotency of induced pluripotent stem (iPS) cells, and the lack of immunological incompatibility and ethical issues, iPS cells have been considered as an invaluable cell source for future cell replacement therapy. This study was aimed first at establishment of novel iPS cells, ECiPS, which directly reprogrammed from human Eye Conjunctiva-derived Mesenchymal Stem Cells (EC-MSCs); second, comparing the inductive effects of Wnt3a/Activin A biomolecules to IDE1 small molecule in derivation of definitive endoderm (DE) from the ECiPS cells. To that end, first, the EC-MSCs were transduced by SOKM-expressing lentiviruses and characterized for endogenous expression of embryonic markers Then the established ECiPS cells were induced to DE formation by Wnt3a/Activin A or IDE1. Quantification of GSC, Sox17 and Foxa2 expression, as DE-specific markers, in both mRNA and protein levels revealed that induction of ECiPS cells by either Wnt3a/Activin A or IDE1 could enhance the expression level of the genes; however the levels of increase were higher in Wnt3a/Activin A induced ECiPS-EBs than IDE1 induced cells. Furthermore, the flow cytometry analyses showed no synergistic effect between Activin A and Wnt3a to derive DE-like cells from ECiPS cells. The comparative findings suggest that although both Wnt3a/Activin A signaling and IDE1 molecule could be used for differentiation of iPS into DE cells, the DE-inducing effect of Wnt3a/Activin A was statistically higher than IDE1.


Asunto(s)
Transdiferenciación Celular/fisiología , Conjuntiva/citología , Endodermo/fisiología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Activinas/genética , Activinas/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Reprogramación Celular/fisiología , Conjuntiva/metabolismo , Endodermo/citología , Células HEK293 , Humanos , Ratones , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
7.
Cell Biol Int ; 38(7): 825-34, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24687540

RESUMEN

In spite of certain clinical limitations, such as teratoma formation, the use of stem cells is considered as an appropriate source in cell therapy and tissue engineering. This study shows human endometrial stem cells (hEnSCs) has exceptional differentiation ability in hepatocyte formation. hEnSCs have high purification rate and immune-tolerance, and can be used as an appropriate substitute for hepatocytes in liver disorders. Differentiation required hepatogenic medium. Quantitative reverse transcription-polymerase chain reaction and immunofluorescent staining of hepatic genes and proteins including cytokeratin 18 (ck18), alpha-fetoprotein (afp), and albumin (alb) were used to assess differentiation. Cells differentiated with a hepatocyte-like morphology and expressed hepatic markers on 30 days of differentiation. The Periodic Acid-Schiff (PAS) reaction showed storage of glycogen, and albumin and afp secretions were also detected. In vitro hEnSCs behave like hepatocyte after differentiation and may be a suitable source of cells in liver regeneration.


Asunto(s)
Diferenciación Celular , Endometrio/citología , Hepatocitos/citología , Células Madre/citología , Adipogénesis , Adulto , Albúminas/metabolismo , Células Cultivadas , Femenino , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inmunohistoquímica , Queratina-18/metabolismo , Osteogénesis , Adulto Joven , alfa-Fetoproteínas/metabolismo
8.
J Immunother Cancer ; 12(3)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38490714

RESUMEN

BACKGROUND: In a prior report, we detailed the isolation and engineering of a bispecific killer cell engager, referred to as BiKE:E5C1. The BiKE:E5C1 exhibits high affinity/specificity for the CD16a activating receptor on natural killer (NK) cells and human epidermal growth factor receptor 2 (HER2) on cancer cells. In vitro studies have demonstrated that BiKE:E5C1 can activate the NK cells and induce the killing of HER2+ ovarian and breast cancer cells, surpassing the performance of the best-in-class monoclonal antibody, Trazimera (trastuzumab). To advance this BiKE technology toward clinical application, the objective of this research was to demonstrate the ability of BiKE:E5C1 to activate CD16+ immune cells such as NK cells and macrophages to kill cancer cells, and eradicate metastatic HER2+ tumors in NK humanized NOG mice. METHODS: We assessed BiKE:E5C1's potential to activate CD16-expressing peripheral blood (PB)-NK cells, laNK92 cells, and THP-1-CD16A monocyte-macrophages through flowcytometry and antibody-dependent cell-mediated cytotoxicity/phagocytosis (ADCC) assays. Subsequently, laNK92 cells were selected as effector cells and genetically modified to express the nanoluciferase gene, enabling the monitoring of their viability in NK humanized NOG mice using quantitative bioluminescent imaging (qBLI). To evaluate the functionality of BiKE:E5C1 in vivo, we introduced firefly luciferase-expressing ovarian cancer cells via intraperitoneal injection into hIL-15 and hIL-2 NOG mice, creating a model of ovarian cancer metastasis. Once tumor establishment was confirmed, we treated the mice with laNK92 cells plus BiKE:E5C1 and the response to therapy was assessed using qBLI. RESULTS: Our data demonstrate that BiKE:E5C1 activates not only laNK92 cells but also PB-NK cells and macrophages, significantly enhancing their anticancer activities. ADCC assay demonstrated that IgG1 Fc region had no impact on BiKE:E5C1's anticancer activity. In vivo results reveal that both hIL-15 and hIL-2 NOG mouse models support the viability and proliferation of laNK92 cells. Furthermore, it was observed that BiKE:E5C1 activates laNK92 cells in mice, leading to eradication of cancer metastasis in both NK humanized hIL-15 and hIL-2 NOG mouse models. CONCLUSIONS: Collectively, our in vivo findings underscore BiKE:E5C1's potential as an immune cell engager capable of activating immune cells for cancer cell elimination, thereby expanding the arsenal of available BiKEs for cancer immunotherapy.


Asunto(s)
Células Asesinas Naturales , Neoplasias Ováricas , Femenino , Ratones , Humanos , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Trastuzumab , Macrófagos , Neoplasias Ováricas/metabolismo
9.
Gut ; 61(8): 1187-96, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21948943

RESUMEN

OBJECTIVES: During pancreatitis, specific transcriptional programmes govern functional regeneration after injury. The objective of this study was to analyse the dynamic regulation of pancreatic genes and the role of transcriptional regulators during recovery from pancreatitis. DESIGN: Wild-type and genetically modified mice (Hnf1α(-/-) and Ptf1a(+/-)) were used. After caerulein or L-arginine induced pancreatitis, blood or pancreata were processed for enzymatic assays, ELISA, histology, immunohistochemistry, western blotting and quantitative reverse transcriptase-PCR. Nr5a2 promoter reporter and chromatin immunoprecipitation assays for Hnf1α were also performed. RESULTS: After caerulein pancreatic injury, expression of acinar and endocrine genes rapidly decreased, but eventually recovered, depicting distinct cell-type-specific patterns. Pdx1 and Hnf1α mRNAs underwent marked downregulation, matching endocrine/exocrine gene expression profiles. Ptf1a, Pdx1 and Hnf1α protein levels were also reduced and recovered gradually. These changes were associated with transient impairment of exocrine and endocrine function, including abnormal glucose tolerance. On l-arginine pancreatitis, changes in Ptf1a, Pdx1 and Hnf1α gene and protein expression were recapitulated. Reduced Hnf1α and Ptf1a levels after pancreatitis coincided with increased acinar cell proliferation, both in Hnf1α(-/-) and Ptf1a(+/-) mice. Moreover, Hnf1α(-/-) mice had reduced Ptf1a protein as well as transcripts for Ptf1a and digestive enzymes. Dispersed acini from Hnf1α(-/-) mice showed suboptimal secretory responses to caerulein. Bioinformatics analysis did not support a role for Hnf1α as a direct regulator of digestive enzyme genes. Instead, it was found that Hnf1α binds to, and regulates, the promoter of Nr5a2, coding an orphan nuclear receptor that regulates acinar gene expression. CONCLUSIONS: Dynamic changes in gene expression occur on pancreatitis induction, determining altered exocrine and endocrine function. This analysis uncovers roles for Hnf1α in the regulation of acinar cell determination and function. This effect may be mediated, in part, through direct regulation of Nr5a2.


Asunto(s)
Células Acinares/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Homeostasis/genética , Pancreatitis/genética , ARN Mensajero/genética , Células Acinares/patología , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Pancreatitis/metabolismo , Pancreatitis/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Nat Biomed Eng ; 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996616

RESUMEN

The immunogenicity of transplanted allogeneic cells and tissues is a major hurdle to the advancement of cell therapies. Here we show that the overexpression of eight immunomodulatory transgenes (Pdl1, Cd200, Cd47, H2-M3, Fasl, Serpinb9, Ccl21 and Mfge8) in mouse embryonic stem cells (mESCs) is sufficient to immunologically 'cloak' the cells as well as tissues derived from them, allowing their survival for months in outbred and allogeneic inbred recipients. Overexpression of the human orthologues of these genes in human ESCs abolished the activation of allogeneic human peripheral blood mononuclear cells and their inflammatory responses. Moreover, by using the previously reported FailSafe transgene system, which transcriptionally links a gene essential for cell division with an inducible and cell-proliferation-dependent kill switch, we generated cloaked tissues from mESCs that served as immune-privileged subcutaneous sites that protected uncloaked allogeneic and xenogeneic cells from rejection in immune-competent hosts. The combination of cloaking and FailSafe technologies may allow for the generation of safe and allogeneically accepted cell lines and off-the-shelf cell products.

11.
Biotechnol Appl Biochem ; 59(3): 205-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23586830

RESUMEN

Pancreatic duodenum homeobox protein-1 (PDX1) is a master regulatory gene in pancreatic development. Reprogramming of mesenchymal stem cells (MSCs) is a promising tool for producing insulin-producing cells. In this study, lentivirus harboring PDX1 (LV-PDX1) has been used for persistence gene expression in MSCs. The objective of this study was to evaluate the potential of lentivirus to introduce the PDX1 gene into MSCs to produce insulin-secreting cells and apply it for treatment of hyperglycemia in diabetic rats. MSCs were isolated from rat bone marrow, characterized, and transduced by LV-PDX1. Significant expressions of PDX1, neurogenin3, glucagon, glucose transporter2 (Glut2), and insulin were detected by quantitative reverse transcription-polymerase chain reaction (P < 0.05). PDX1 and insulin were detected at the protein level by immunofluorescence analysis. PDX1 could trigger a gene expression cascade that involved pancreatic endocrine differentiation and also revealed the glucose sensing ability by expressing Glut2 in high-glucose medium. The insulin secretion of MSCs(PDX1+) in the high-glucose medium was 1.75-fold higher than that secreted in the low-glucose medium (P < 0.05). MSCs(PDX1+) implanted into diabetic rats could decrease the blood glucose level from 485 mg/dL to the normal level in 3 days. This study showed MSCs(PDX1+) have the potential to be used as a viable resource in cell-based gene therapy of type 1 diabetes.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Homeodominio/metabolismo , Islotes Pancreáticos/citología , Lentivirus/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/virología , Transactivadores/metabolismo , Animales , Diabetes Mellitus Experimental/terapia , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Proteínas de Homeodominio/genética , Masculino , Reacción en Cadena de la Polimerasa , Ratas , Transactivadores/genética
12.
Gastroenterology ; 135(4): 1301-1310, 1310.e1-5, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18725222

RESUMEN

BACKGROUND & AIMS: Acinar cells constitute 90% of the pancreas epithelium, are polarized, and secrete digestive enzymes. These cells play a crucial role in pancreatitis and pancreatic cancer. However, there are limited models to study normal acinar cell differentiation in vitro. The aim of this work was to generate and characterize purified populations of pancreatic acinar cells from embryonic stem (ES) cells. METHODS: Reporter ES cells (Ela-pur) were generated that stably expressed both beta-galactosidase and puromycin resistance genes under the control of the elastase I promoter. Directed differentiation was achieved by incubation with conditioned media of cultured fetal pancreatic rudiments and adenoviral-mediated co-expression of p48/Ptf1a and Mist1, 2 basic helix-loop-helix transcription factors crucial for normal pancreatic acinar development and differentiation. RESULTS: Selected cells expressed multiple markers of acinar cells, including digestive enzymes and proteins of the secretory pathway, indicating activation of a coordinated differentiation program. The genes coding for digestive enzymes were not regulated as a single module, thus recapitulating what occurs during in vivo pancreatic development. The generated cells displayed transient agonist-induced Ca(2+) mobilization and showed a typical response to physiologic concentrations of secretagogues, including enzyme synthesis and secretion. Importantly, these effects did not imply the acquisition of a mixed acinar-ductal phenotype. CONCLUSIONS: These studies allow the generation of almost pure acinar-like cells from ES cells, providing a normal cell-based model for the study of the acinar differentiation program in vitro.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Páncreas Exocrino/citología , Páncreas Exocrino/embriología , Amilasas/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Carbacol/farmacología , Carboxipeptidasas A/genética , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , División Celular/fisiología , Células Cultivadas , Agonistas Colinérgicos/farmacología , Quimotripsinógeno/genética , Células Madre Embrionarias/ultraestructura , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Genes Reporteros , Ratones , Microscopía Inmunoelectrónica , Elastasa Pancreática/genética , Factores de Transcripción/genética , Transfección
13.
Curr Protoc Hum Genet ; 100(1): e71, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30335214

RESUMEN

The International Diabetic Federation estimated that 415 million adults currently have diabetes and 318 million adults had impaired glucose tolerance, putting them at high risk of developing diabetes in the future. In Type 1 Diabetes (T1D), the ß cells are lost because of autoimmune reactions. Although islet transplantation has been a promising therapy for T1D, it is greatly limited by pancreatic donors. Here, we describe a protocol to generate glucose- responsive pancreatic ß-like (GRPß-L) cells from human-induced pluripotent stem (iPS) cells. We recapitulate in vivo pancreas development by in vitro induction of differentiating human (iPS) cells with stage-specific signaling molecules and proteins. Inhibition of Tyrosine Kinase receptor AXL, TGF-ß, and Notch signaling pathways in the final stage of the five-stage protocol could efficiently generate GRPß-L from the endocrine progenitor. Differentiation of human iPS cells through the protocol could result in functional GRPß-L cells, which could be used in pharmaceutical and ß cell biology studies. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/metabolismo , Linaje de la Célula/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/terapia , Glucosa/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/patología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores Notch/antagonistas & inhibidores , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Tirosina Quinasa del Receptor Axl
14.
Int J Dev Biol ; 51(8): 723-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17939119

RESUMEN

Mesenchymal stem cells (MSCs) have been isolated based on the ability of adherence to plastic surfaces. The potential of these cells to differentiate along multiple lineages is the key to identifying stem cell populations in the absence of molecular markers. Here we describe a homogenous population of MSCs from mouse bone marrow isolated using a relatively straightforward and novel approach. This method is based on the combination of frequent medium change (FMC) and treatment of the primary cultures with trypsin. Cells isolated using this method demonstrated the MSCs characteristics including their ability to differentiate into mesenchymal lineages. MSCs retained the differentiation potentials in expanded cultures up to 10 passages. Isolated MSCs were reactive to the CD44, Sca-1, and CD90 cell surface markers. MSCs were negative for the hematopoietic surface markers such as CD34, CD11b, CD45, CD31, CD106, CD117 and CD135. The data presented in this report indicated that this method can result in efficient isolation of homogenous populations of MSCs from mouse bone marrow.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Animales , Diferenciación Celular , Células Cultivadas , Citometría de Flujo/métodos , Receptores de Hialuranos/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Monocitos/metabolismo , Células Madre , Tripsina/farmacología
15.
J Biomed Mater Res B Appl Biomater ; 105(8): 2516-2529, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27689849

RESUMEN

Liver tissue engineering (TE) is rapidly emerging as an effective technique which combines engineering and biological processes to compensate for the shortage of damaged or destroyed liver tissues. We examined the viability, differentiation, and integration of hepatocyte-like cells on an electrospun polyethersulfone (PES) scaffold, derived from human endometrial stem cells (hEnSCs). Natural polymers were separately grafted on plasma-treated PES nanofibers, that is, collagen, heparan sulfate (HS) and collagen-HS. Galactosilated PES (PES-Gal) nanofibrous were created. The engineering and cell growth parameters were considered and compared with each sample. The cellular studies revealed increased cell survival, attachment, and normal morphology on the bioactive natural polymer-grafted scaffolds after 30 days of hepatic differentiation. The chemical and molecular assays displayed hepatocyte differentiation. These cells were also functional, showing glycogen storage, α-fetoprotein, and albumin secretion. The HS nanoparticle-grafted PES nanofibers demonstrated a high rate of cell proliferation, differentiation, and integration. Based on the observations mentioned above, engineered tissue is a good option in the future, for the commercial production of three-dimensional liver tissues for clinical purposes. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2516-2529, 2017.


Asunto(s)
Diferenciación Celular , Colágeno/química , Endometrio/metabolismo , Glicosaminoglicanos/química , Hepatocitos/metabolismo , Nanofibras/química , Polímeros/química , Células Madre/metabolismo , Sulfonas/química , Andamios del Tejido/química , Técnicas de Cultivo de Célula , Células Cultivadas , Endometrio/citología , Femenino , Hepatocitos/citología , Humanos , Células Madre/citología
16.
ACS Appl Mater Interfaces ; 9(8): 6849-6864, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28116894

RESUMEN

In the embryonic heart, electrical impulses propagate in a unidirectional manner from the sinus venosus and appear to be involved in cardiogenesis. In this work, aligned and random polyaniline/polyetersulfone (PANI/PES) nanofibrous scaffolds doped by Camphor-10-sulfonic acid (ß) (CPSA) were fabricated via electrospinning and used to conduct electrical impulses in a unidirectional and multidirectional fashion, respectively. A bioreactor was subsequently engineered to apply electrical impulses to cells cultured on PANI/PES scaffolds. We established cardiovascular disease-specific induced pluripotent stem cells (CVD-iPSCs) from the fibroblasts of patients undergoing cardiothoracic surgeries. The CVD-iPSCs were seeded onto the scaffolds, cultured in cardiomyocyte-inducing factors, and exposed to electrical impulses for 1 h/day, over a 15-day time period in the bioreactor. The application of the unidirectional electrical stimulation to the cells significantly increased the number of cardiac Troponin T (cTnT+) cells in comparison to multidirectional electrical stimulation using random fibrous scaffolds. This was confirmed by real-time polymerase chain reaction for cardiac-related transcription factors (NKX2.5, GATA4, and NPPA) and a cardiac-specific structural gene (TNNT2). Here we report for the first time that applying electrical pulses in a unidirectional manner mimicking the unidirectional wave of electrical stimulation in the heart, could increase the derivation of cardiomyocytes from CVD-iPSCs.


Asunto(s)
Enfermedades Cardiovasculares , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Nanofibras , Andamios del Tejido
17.
PLoS One ; 11(3): e0150518, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26938987

RESUMEN

Induced pluripotent stem cells (iPSCs) provide a reliable source for the study of regenerative medicine, drug discovery, and developmental biology. Despite extensive studies on the reprogramming of mouse and human fibroblasts into iPSCs, the efficiency of reprogramming is still low. Here, we used a bioinformatics and systems biology approach to study the two gene regulatory waves governing the reprogramming of mouse and human fibroblasts into iPSCs. Our results revealed that the maturation phase of reprogramming was regulated by a more complex regulatory network of transcription factors compared to the initiation phase. Interestingly, in addition to pluripotency factors, the polycomb repressive complex 2 (PRC2) members Ezh2, Eed, Jarid2, Mtf2, and Suz12 are crucially recruited during the maturation phase of reprogramming. Moreover, we found that during the maturation phase of reprogramming, pluripotency factors, via the expression and induction of PRC2 complex members, could silence the lineage-specific gene expression program and maintain a ground state of pluripotency in human and mouse naïve iPSCs. The findings obtained here provide us a better understanding of the gene regulatory network (GRN) that governs reprogramming, and the maintenance of the naïve state of iPSCs.


Asunto(s)
Reprogramación Celular/genética , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Complejo Represivo Polycomb 2/metabolismo , Animales , Diferenciación Celular/genética , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Complejo Represivo Polycomb 2/genética , Mapas de Interacción de Proteínas/genética
18.
Neurosci Lett ; 626: 126-34, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27208834

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder, in which the nigro-striatal Dopaminergic (DAergic) neurons are selectively lost. Treatment of neurodegenerative diseases with Pluripotent Stem Cells (PSCs) is a big interest in cell therapy. Here, we used induced Pluripotent Stem Cells (iPSCs) expressing two master Dopaminergic (DAergic) transcription factors, i.e. Nurr1 and Pitx3, to generate functional in vitro DAergic-like neurons. After establishment and characterization of Doxycycline-inducible iPSCs from mouse fibroblasts, the cells were transduced by NURR1- and PITX3-harboring lentiviruses. The Nurr1/Pitx3 -iPSCs were differentiated through a five-stage protocol to generate DAergic-like neurons. The results confirmed the efficient expression of DAergic neuron markers in the end of protocol. Beside, the generated cells could exclusively synthesize and secrete Dopamine in response to secretagogues. In conclusion, overexpression of Nurr1 and Pitx3 in iPSCs could efficiently program iPSCs into functional DAergic-like neurons. This finding may have an impact on future stem cell therapy of PD.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Neuronas Dopaminérgicas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Vectores Genéticos , Células Madre Pluripotentes Inducidas/metabolismo , Lentivirus/fisiología , Ratones
19.
PLoS One ; 11(10): e0164457, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27755557

RESUMEN

The ability to yield glucose-responsive pancreatic beta-cells from human pluripotent stem cells in vitro will facilitate the development of the cell replacement therapies for the treatment of Type 1 Diabetes. Here, through the sequential in vitro targeting of selected signaling pathways, we have developed an abbreviated five-stage protocol (25-30 days) to generate human Embryonic Stem Cell-Derived Beta-like Cells (ES-DBCs). We showed that Geltrex, as an extracellular matrix, could support the generation of ES-DBCs more efficiently than that of the previously described culture systems. The activation of FGF and Retinoic Acid along with the inhibition of BMP, SHH and TGF-beta led to the generation of 75% NKX6.1+/NGN3+ Endocrine Progenitors. The inhibition of Notch and tyrosine kinase receptor AXL, and the treatment with Exendin-4 and T3 in the final stage resulted in 35% mono-hormonal insulin positive cells, 1% insulin and glucagon positive cells and 30% insulin and NKX6.1 co-expressing cells. Functionally, ES-DBCs were responsive to high glucose in static incubation and perifusion studies, and could secrete insulin in response to successive glucose stimulations. Mitochondrial metabolic flux analyses using Seahorse demonstrated that the ES-DBCs could efficiently metabolize glucose and generate intracellular signals to trigger insulin secretion. In conclusion, targeting selected signaling pathways for 25-30 days was sufficient to generate ES-DBCs in vitro. The ability of ES-DBCs to secrete insulin in response to glucose renders them a promising model for the in vitro screening of drugs, small molecules or genes that may have potential to influence beta-cell function.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células Secretoras de Insulina/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Células Cultivadas , Endodermo/citología , Endodermo/metabolismo , Exenatida , Glucosa/farmacología , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Análisis de Flujos Metabólicos , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Péptidos/farmacología , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Tretinoina/farmacología , Ponzoñas/farmacología
20.
J Biomed Mater Res A ; 103(1): 1-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24408884

RESUMEN

A very important obstacle in axonal regeneration after spinal cord injury is astroglial scaring. Noggin as bone morphogenic protein inhibitor plays a critical role in decreasing GFAP(+) cells and reducing the number of astrocytes in the site of injury. Human endometrial-derived stromal cells (hEnSCs) were isolated and cultured in two different neural inductive mediums consisting of neural progenitor maintenance medium (NPMM)/BDNF or NPMM/BDNF/Noggin in Matrigel 3D cell culture. Neural expression markers were investigated at the mRNA and protein level by real-time PCR and immunocytochemistry, respectively. The results showed that Noggin supplementation was able to increase the expression of Nestin, Tuj-1, and NF, whereas the expressions of GFAP, Bcl2, and Olig2 were decreased. In addition, DAPI staining demonstrated that lighter blue chromatin agreed with our observation of lower level of Bcl2 expression in the Noggin protocol in which over-expression of Bcl2 gene did not induce higher neurogenesis in poor Noggin medium. Our findings clearly demonstrated the neural differentiation potential of hEnSC in Matrigel and also Noggin supplementation was able to inhibit astrocyte formation.


Asunto(s)
Proteínas Portadoras/administración & dosificación , Colágeno , Endometrio/citología , Laminina , Nanofibras , Proteoglicanos , Células del Estroma/citología , Secuencia de Bases , Células Cultivadas , Medios de Cultivo , Cartilla de ADN , Combinación de Medicamentos , Femenino , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA