Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Diabetes ; 69(8): 1723-1734, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32457219

RESUMEN

The ß-cell protein synthetic machinery is dedicated to the production of mature insulin, which requires the proper folding and trafficking of its precursor, proinsulin. The complete network of proteins that mediate proinsulin folding and advancement through the secretory pathway, however, remains poorly defined. Here we used affinity purification and mass spectrometry to identify, for the first time, the proinsulin biosynthetic interaction network in human islets. Stringent analysis established a central node of proinsulin interactions with endoplasmic reticulum (ER) folding factors, including chaperones and oxidoreductases, that is remarkably conserved in both sexes and across three ethnicities. The ER-localized peroxiredoxin PRDX4 was identified as a prominent proinsulin-interacting protein. In ß-cells, gene silencing of PRDX4 rendered proinsulin susceptible to misfolding, particularly in response to oxidative stress, while exogenous PRDX4 improved proinsulin folding. Moreover, proinsulin misfolding induced by oxidative stress or high glucose was accompanied by sulfonylation of PRDX4, a modification known to inactivate peroxiredoxins. Notably, islets from patients with type 2 diabetes (T2D) exhibited significantly higher levels of sulfonylated PRDX4 than islets from healthy individuals. In conclusion, we have generated the first reference map of the human proinsulin interactome to identify critical factors controlling insulin biosynthesis, ß-cell function, and T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Peroxirredoxinas/metabolismo , Proinsulina/química , Proinsulina/metabolismo , Western Blotting , Diabetes Mellitus Tipo 2/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Inmunoprecipitación , Insulina/química , Masculino , Peroxirredoxinas/genética , Unión Proteica , Pliegue de Proteína , Espectrometría de Masas en Tándem
2.
Cell Mol Gastroenterol Hepatol ; 6(2): 181-198, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30003124

RESUMEN

BACKGROUND & AIMS: Oncogenic mutations in KRAS, coupled with inactivation of p53, CDKN2A/p16INK4A, and SMAD4, drive progression of pancreatic ductal adenocarcinoma (PDA). Overexpression of MYC and deregulation of retinoblastoma (RB) further promote cell proliferation and make identifying a means to therapeutically alter cell-cycle control pathways in PDA a significant challenge. We previously showed that the basic helix-loop-helix transcription factor E47 induced stable growth arrest in PDA cells in vitro and in vivo. Here, we identified molecular mechanisms that underlie E47-induced growth arrest in low-passage, patient-derived primary and established PDA cell lines. METHODS: RNA sequencing was used to profile E47-dependent transcriptomes in 5 PDA cell lines. Gene Ontology analysis identified cell-cycle control as the most altered pathway. Small interfering RNA/short hairpin RNA knockdown, small-molecule inhibitors, and viral expression were used to examine the function of E47-dependent genes in cell-cycle arrest. Cell morphology, expression of molecular markers, and senescence-associated ß-galactosidase activity assays identified cellular senescence. RESULTS: E47 uniformly inhibited PDA cell-cycle progression by decreasing expression of MYC, increasing the level of CDKN1B/p27KIP1, and restoring RB tumor-suppressor function. The molecular mechanisms by which E47 elicited these changes included altering both RNA transcript levels and protein stability of MYC and CDKN1B/p27KIP1. At the cellular level, E47 elicited a senescence-like phenotype characterized by increased senescence-associated ß-galactosidase activity and altered expression of senescence markers. CONCLUSIONS: E47 governs a highly conserved network of cell-cycle control genes, including MYC, CDKN1B/p27KIP1, and RB, which can induce a senescence-like program in PDA cells that lack CDKN2A/p16INK4A and wild-type p53. RNA sequencing data are available at the National Center for Biotechnology Information GEO at https://www.ncbi.nlm.nih.gov/geo/; accession number: GSE100327.

3.
Oncotarget ; 8(32): 53154-53167, 2017 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-28881801

RESUMEN

The average survival for patients with Pancreatic Ductal Adenocarcinoma (PDA) is merely 6 months, underscoring the need for new therapeutic approaches. During PDA progression, pancreatic acinar cells lose activity of the ClassI/II bHLH factors that regulate quiescence. We previously found that promoting transcriptional activity of the Class I bHLH factor E47 in highly aggressive PDA cells induced stable growth arrest in vitro and in vivo. To translate these findings for clinical utility, we developed a high throughput screening platform to identify small molecule inducers of Class I/II bHLH activity. A screen of 4,375 known drugs identified 70 bHLH activators. Prominent among the hits were members of the statin class of HMG-CoA reductase inhibitors, cholesterol lowering drugs that are also being evaluated in cancer. Studies with pitavastatin in primary patient derived tumor cells and established PDA lines, revealed dose dependent growth inhibition. At the molecular level, pitavastatin induced expression of the cyclin dependent kinase (CDK) inhibitor p21 in a cholesterol independent manner, blocked repressive phosphorylation of the Retinoblastoma tumor suppressor protein at CDK targeted sites, and reduced expression of E2F target genes required for progression through the G1/S boundary. Together, the data provide new insight into mechanisms by which statins constrain proliferation in cancer and establish the effectiveness of a novel screening platform to identify small molecules of clinical relevance in pancreatic cancer.

4.
J Tissue Eng Regen Med ; 10(6): 527-34, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-23897763

RESUMEN

Development of stem cell-based therapies for the treatment of type 1 diabetes would provide a renewable supply of human ß-cells. Human embryonic stem cells (ESCs) are considered to be one of the stem cell populations with sufficient proliferative capacity to achieve this goal. Currently, differentiation protocols directing ESCs toward a pancreatic fate employ a variety of expensive cytokines and inhibitors. With the known significance of microRNAs in islet development, we present a novel and cost-effective strategy in which miR-375 overexpression promotes pancreatic endocrine differentiation in hESCs in the absence of any extrinsic factors. miR-375 has been shown to be a key regulator of pancreatic development and function in zebrafish, mouse and human. In this study, hESCs were transduced with lentiviral vectors containing human miR-375 precursor and aggregated to form human embryoid bodies (hEBs) for up to 21 days. Morphological assessment, immunocytochemistry and DTZ staining confirmed that miR-375-induced hEBs have similar characteristics to those of mature islets. In addition, the dynamic expression profile of endodermal marker Foxa2 and endocrine-specific genes, including HNF4α, Pdx1, Pax6, Nkx6.1, Glut2 and insulin, were detected by quantitative real-time PCR. Finally, insulin release upon glucose stimulation was detected in our differentiated clusters. The data presented here demonstrate the feasibility of using microRNAs to direct differentiation into the pancreatic lineage. Copyright © 2013 John Wiley & Sons, Ltd.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Diferenciación Celular , Células Madre Embrionarias Humanas/metabolismo , Islotes Pancreáticos/metabolismo , MicroARNs/biosíntesis , Línea Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Islotes Pancreáticos/citología , MicroARNs/genética
5.
Pancreas ; 44(5): 718-27, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25894862

RESUMEN

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDA) initiates from quiescent acinar cells that attain a Kras mutation, lose signaling from basic helix-loop-helix (bHLH) transcription factors, undergo acinar-ductal metaplasia, and rapidly acquire increased growth potential. We queried whether PDA cells can be reprogrammed to revert to their original quiescent acinar cell state by shifting key transcription programs. METHODS: Human PDA cell lines were engineered to express an inducible form of the bHLH protein E47. Gene expression, growth, and functional studies were investigated using microarray, quantitative polymerase chain reaction, immunoblots, immunohistochemistry, small interfering RNA, chromatin immunoprecipitation analyses, and cell transplantation into mice. RESULTS: In human PDA cells, E47 activity triggers stable G0/G1 arrest, which requires the cyclin-dependent kinase inhibitor p21 and the stress response protein TP53INP1. Concurrently, E47 induces high level expression of acinar digestive enzymes and feed forward activation of the acinar maturation network regulated by the bHLH factor MIST1. Moreover, induction of E47 in human PDA cells in vitro is sufficient to inhibit tumorigenesis. CONCLUSIONS: Human PDA cells retain a high degree of plasticity, which can be exploited to induce a quiescent acinar cell state with reduced tumorigenic potential. Moreover, bHLH activity is a critical node coordinately regulating human PDA cell growth versus cell fate.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Reprogramación Celular , Senescencia Celular , Neoplasias Pancreáticas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/prevención & control , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Terapia Genética , Humanos , Ratones SCID , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/prevención & control , Fenotipo , Interferencia de ARN , Factores de Tiempo , Transfección , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cryobiology ; 70(3): 283-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25817378

RESUMEN

We have previously shown that human embryonic stem cell derived islet progenitors (hESC-IPs), encapsulated inside an immunoprotective device, mature in vivo and ameliorate diabetes in mice. The ability to cryopreserve hESC-IPs preloaded in these devices would enhance consistency and portability, but traditional 'slow freezing' methods did not work well for cells encapsulated in the device. Vitrification is an attractive alternative cryopreservation approach. To assess the tolerance of hESC-IPs to vitrification relevant conditions, we here are reporting cell survival following excursions in tonicity, exposure to fifteen 40% v/v combinations of 4 cryoprotectants, and varied methods for addition and elution. We find that 78% survival is achieved using a protocol in which cells are abruptly (in one step) exposed to a solution containing 10% v/v each dimethyl sulfoxide, propylene glycol, ethylene glycol, and glycerol on ice, and eluted step-wise with DPBS+0.5M sucrose at 37°C. Importantly, the hESC-IPs also maintain expression of the critical islet progenitor markers PDX-1, NKX6.1, NGN3 and NEURO-D1. Thus, hESC-IPs exhibit robust tolerance to exposure to vitrification solutions in relevant conditions.


Asunto(s)
Criopreservación/métodos , Crioprotectores/farmacología , Células Madre Embrionarias Humanas/citología , Islotes Pancreáticos/embriología , Vitrificación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Supervivencia Celular , Dimetilsulfóxido/farmacología , Glicol de Etileno/farmacología , Congelación , Glicerol/farmacología , Proteínas de Homeodominio/genética , Humanos , Islotes Pancreáticos/citología , Proteínas del Tejido Nervioso/genética , Propilenglicol/farmacología , Soluciones , Sacarosa/farmacología , Transactivadores/genética , Factores de Transcripción/genética
7.
Stem Cell Res ; 12(3): 807-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24788136

RESUMEN

There are several challenges to successful implementation of a cell therapy for insulin dependent diabetes derived from human embryonic stem cells (hESC). Among these are development of functional insulin producing cells, a clinical delivery method that eliminates the need for chronic immunosuppression, and assurance that hESC derived tumors do not form in the patient. We and others have shown that encapsulation of cells in a bilaminar device (TheraCyte) provides immunoprotection in rodents and primates. Here we monitored human insulin secretion and employed bioluminescent imaging (BLI) to evaluate the maturation, growth, and containment of encapsulated islet progenitors derived from CyT49 hESC, transplanted into mice. Human insulin was detectable by 7 weeks post-transplant and increased 17-fold over the course of 8 weeks, yet during this period the biomass of encapsulated cells remained constant. Remarkably, by 20 weeks post-transplant encapsulated cells secreted sufficient levels of human insulin to ameliorate alloxan induced diabetes. Further, bioluminescent imaging revealed for the first time that hESCs remained fully contained in encapsulation devices for up to 150 days, the longest period tested. Collectively, the data suggest that encapsulated hESC derived islet progenitors hold great promise as an effective and safe cell replacement therapy for insulin dependent diabetes.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Diabetes Mellitus/terapia , Células Madre Embrionarias/citología , Islotes Pancreáticos/crecimiento & desarrollo , Animales , Técnicas de Cultivo de Célula/instrumentación , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Células Madre Embrionarias/química , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/química , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Coloración y Etiquetado
8.
Mol Biol Rep ; 41(4): 2055-66, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24469711

RESUMEN

Islet transplantation is considered as an ultimate option for the treatment of type I diabetes. Human induced pluripotent stem cells (hiPSCs) have raised the possibility that patient-specific insulin-secreting cells might be derived from somatic cells through cell fate reprogramming. However, current protocols mostly rely on the use of several cytokines and inhibitors for directing differentiation towards pancreatic fate. Given the high manufacturing cost of these recombinant proteins, this approach is prohibitive for clinical applications. Knowing that microRNAs (miRNAs) are key players in various stages of pancreatic development, we present a novel and cost-effective strategy in which over-expression of miR-375 promotes pancreatic differentiation in hiPSCs in the absence of any other stimulator. We used a polycistronic viral vector expressing Sox2, Klf4, c-Myc, and Oct4 to drive hiPSCs from human foreskin fibroblasts. The established hiPSCs are similar to human embryonic stem cells in many aspects including morphology, passaging, surface and pluripotency markers, and gene expression. For differentiation induction, miR-375 was lentivirally overexpressed in these hiPSCs. Morphological assessment, immunocytochemistry, and expression analysis of islet marker genes confirmed that islet like cells were obtained in miR-375 transduced cells compared to controls. Our differentiated clusters secreted insulin in a glucose-dependant manner, showing in vitro functionality. We demonstrated for the first time that miRNAs might be ideal substitutes to induce pancreatic differentiation in hiPSCs. This work provides a new approach to study the role of miRNAs in pancreatic specification and increase the feasibility of using patient-specific iPSCs for beta cell replacement therapy for type I diabetes.


Asunto(s)
Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , MicroARNs/genética , Biomarcadores , Fibroblastos/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Insulina/biosíntesis , Factor 4 Similar a Kruppel , Fenotipo , Transducción Genética
9.
Int J Hematol Oncol Stem Cell Res ; 8(4): 20-9, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25774264

RESUMEN

BACKGROUND: The use of stem cells is considered as an appropriate source in cell therapy and tissue engineering. Differentiation of human induced Pluripotent Stem Cells (hiPSCs) to Hepatocyte-like Cells (HLCs) on mouse embryonic fibroblasts (MEFs) feeders is confronted with several problems that hinder the clinical applications of these differentiated cells for the treatment of liver injuries. Safe appropriate cells for stem cell-based therapies could create new hopes for liver diseases. This work focused on the determination of a capacity/efficiency for the differentiation of the hiPSCs into Hepatocyte-like Cells on a novel human adult bone marrow mesenchymal stem cells (hMSCs) feeder. MATERIALS AND METHODS: Undifferentiated human iPSCs were cultured on mitotically inactivated human adult bone marrow mesenchymal stem cells. A three-step differentiation process has been performed in presence of activin A which added for 3 days to induce a definitive endoderm formation. In the second step, medium was exchanged for six days. Subsequently, cells were treated with oncostatin M plus dexamethasone for 9 days to generate hepatic cells. Endodermic and liver-specific genes were assessed via quantitative reverse transcription-polymerase chain reaction and RT-PCR, moreover, immunocytochemical staining for liver proteins including albumin and alpha-fetoprotein. In addition, functional tests for glycogen storage, oil red examination, urea production and alpha-fetoprotein synthesis, as well as, cells differentiated with a hepatocyte-like morphology was also performed. RESULTS: Our results show that inactivated human adult bone marrow mesenchymal stem cell feeders could support the efficient differentiation of hiPSCs into HLCs. This process induced differentiation of iPSCs into definitive endocrine cells that expressed sox17, foxa2 and expression of the specific genes profiles in hepatic-like cells. In addition, immunocytochemical analysis confirmed albumin and alpha-fetoprotein protein expression, as well as, the hiPSCs-derived Hepatocyte-like Cells on human feeder exhibited a typical morphology. CONCLUSIONS: we suggested a successful and efficient culture for differentiation and maturation of hepatocytes on an alternative human feeders; this is an important step to generate safe and functional hepatocytes that is vital for regenerative medicine and transplantation on the cell-based therapies.

10.
Appl Biochem Biotechnol ; 172(4): 2055-69, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24326679

RESUMEN

Micro RNAs (miRNAs) are a novel class of non-coding regulatory RNA molecules that contribute to post-transcriptional gene regulation. Recent studies have demonstrated that specific miRNAs such as miR-150, miR-154, and miR-451 have key roles in erythropoiesis. To date, stimulatory cytokines are considered as unique effectors for in vitro differentiation of HSCs to erythropoietic lineage. However, the use of these factors is not cost-effective for clinical applications and therapeutic strategies. Here, we present a novel and cost-effective strategy in which miRNAs expression modulation promotes erythroid differentiation in HSCs in the absence of any extrinsic factors. Thus, CD133(+) hematopoietic stem cells purified from human umbilical cord blood were treated with pre-miR-451 containing lentiviruses, anti-miR-150 and anti-miR-154 in the absence of growth factors and cytokines. Obtained results indicated that miR-451 upregulation and miR-150 downregulation have positive effect on GATA-1, FOG-1, and EKLF, CD71 and CD235a genes expression and induce hemoglobinization efficiently. However, downregulation of miR-154 had no effect on erythropoiesis indexes compared to that observed in the control group. In conclusion, the data presented here for the first time demonstrate that expression modulation of miR-451 and miR-150 could be an efficient alternative to stimulatory cytokines for CD133(+) differentiation into erythroid lineage. Modulation of erythropoiesis in stem cells via miRNA holds promising potential for vascular tissue engineering and regenerative medicine applications.


Asunto(s)
Células Eritroides/citología , Células Eritroides/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , MicroARNs/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , MicroARNs/fisiología , Embarazo
11.
Iran J Basic Med Sci ; 16(6): 756-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23997901

RESUMEN

OBJECTIVE(S): Erythropoiesis is regulated by some extrinsic and intrinsic factors as microRNAs (miRNAs). miRNAs are endogenously small non-coding regulatory RNAs which play vital roles in the variety of cellular fate, critical processes; growth, apoptosis, metabolism, survival of the cells and specially differentiation. Several miRNAs such as miR-16 and miR-451 have been shown to be correlated with erythroid differentiation. Taking into account the importance of miRNAs in cellular differentiation, the goal of the present study was to examine the role of miRNAs in hematopoietic stem cells (HSC) differentiation into the erythroid cells in the absence of growth factors and stimulatory cytokines. MATERIALS AND METHODS: CD133+ stem cells were infected with lentiviruses containing miR-451/miR-16 precursor sequence, erythroid differentiation was evaluated using RT-PCR for hemoglobin chains and surface antigens, also by banzidine staining. RESULTS: MiR-451up-regulation, but not miR-16, could induce α, ß and γ-globin expression in CD133+ cells and have strong correlation with appearance of CD71 and CD235a markers in these cells. Moreover, miR-451 up-regulation increases the banzidine positive cells to ~ %40. CONCLUSION: Our results provide strong evidence that miR-451 up-regulation strongly induces erythroid differentiation and maturation of CD133+ stem cells. Hence, this method may provide a useful technique for the production of artificial blood RBC and be used as a new strategy for gene therapy of hemoglobinopathies, such as ß-thalassemias and sickle cell anemia.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA