Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Biotechnol ; 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-38012450

RESUMEN

Clinical studies on the treatment of type 1 diabetes with device-encapsulated pancreatic precursor cells derived from human embryonic stem cells found that insulin output was insufficient for clinical benefit. We are conducting a phase 1/2, open-label, multicenter trial aimed at optimizing cell engraftment (ClinicalTrials.gov identifier: NCT03163511 ). Here we report interim, 1-year outcomes in one study group that received 2-3-fold higher cell doses in devices with an optimized membrane perforation pattern. ß cell function was measured by meal-stimulated plasma C-peptide levels at 3-month intervals, and the effect on glucose control was assessed by continuous glucose monitoring (CGM) and insulin dosing. Of 10 patients with undetectable baseline C-peptide, three achieved levels ≥0.1 nmol l-1 from month 6 onwards that correlated with improved CGM measures and reduced insulin dosing, indicating a glucose-controlling effect. The patient with the highest C-peptide (0.23 nmol l-1) increased CGM time-in-range from 55% to 85% at month 12; ß cell mass in sentinel devices in this patient at month 6 was 4% of the initial cell mass, indicating directions for improving efficacy.

2.
Cell Rep Med ; 2(12): 100466, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-35028608

RESUMEN

These preliminary data from an ongoing first-in-human phase 1/2, open-label study provide proof-of-concept that pluripotent stem cell-derived pancreatic endoderm cells (PEC-01) engrafted in type 1 diabetes patients become islet cells releasing insulin in a physiologically regulated fashion. In this study of 17 subjects aged 22-57 with type 1 diabetes, PEC-01 cells were implanted subcutaneously in VC-02 macroencapsulation devices, allowing for direct vascularization of the cells. Engraftment and insulin expression were observed in 63% of VC-02 units explanted from subjects at 3-12 months post-implant. Six of 17 subjects (35.3%) demonstrated positive C-peptide as early as 6 months post-implant. Most reported adverse events were related to surgical implant or explant procedures (27.9%) or to side-effects of immunosuppression (33.7%). Initial data suggest that pluripotent stem cells, which can be propagated to the desired biomass and differentiated into pancreatic islet-like tissue, may offer a scalable, renewable alternative to pancreatic islet transplants.


Asunto(s)
Péptido C/metabolismo , Células Inmovilizadas/citología , Diabetes Mellitus Tipo 1/terapia , Endodermo/citología , Insulina/metabolismo , Páncreas/citología , Trasplante de Células Madre , Células Madre/citología , Adolescente , Adulto , Anciano , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
Am J Transplant ; 21(6): 2090-2099, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33206461

RESUMEN

Detection of amyloid in intraportal islet implants of type 1 diabetes patients has been proposed as cause in their functional decline. The present study uses cultured adult human islets devoid of amyloid to examine conditions of its formation. After intraportal injection in patients, amyloid deposits <15 µm diameter were identified in 5%-12% of beta cell containing aggregates, 3-76 months posttransplant. Such deposits also formed in glucose-controlling islet implants in the kidney of diabetic mice but not in failing implants. Alginate-encapsulated islets formed amyloid during culture when functional, and in all intraperitoneal implants that corrected diabetes in mice, exhibiting larger sizes than in functioning nonencapsulated implants. After intraperitoneal injection in a patient, retrieved single capsules presented amyloid near living beta cells, whereas no amyloid occurred in clustered capsules with dead cells. Amyloid was also demonstrated in functional human stem cell-generated beta cell implants in subcutaneous devices of mice. Deposits up to 35 µm diameter were localized in beta cell-enriched regions and related to an elevated IAPP over insulin ratio in the newly generated beta cells. Amyloid in device-encapsulated human stem cell-generated beta cell implants marks the formation of a functional beta cell mass but also an imbalance between its activated state and its microenvironment.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Adulto , Amiloide , Animales , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Ratones , Células Madre
4.
Stem Cell Reports ; 10(3): 739-750, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29503087

RESUMEN

Human stem cells represent a potential source for implants that replace the depleted functional beta cell mass (FBM) in diabetes patients. Human embryonic stem cell-derived pancreatic endoderm (hES-PE) can generate implants with glucose-responsive beta cells capable of reducing hyperglycemia in mice. This study with device-encapsulated hES-PE (4 × 106 cells/mouse) determines the biologic characteristics at which implants establish metabolic control during a 50-week follow-up. A metabolically adequate FBM was achieved by (1) formation of a sufficient beta cell number (>0.3 × 106/mouse) at >50% endocrine purity and (2) their maturation to a functional state comparable with human pancreatic beta cells, as judged by their secretory responses during perifusion, their content in typical secretory vesicles, and their nuclear NKX6.1-PDX1-MAFA co-expression. Assessment of FBM in implants and its correlation with in vivo metabolic markers will guide clinical translation of stem cell-derived grafts in diabetes.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Endodermo/metabolismo , Endodermo/fisiología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Animales , Línea Celular , Glucosa/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transactivadores/metabolismo
5.
Diabetologia ; 60(1): 126-133, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27787618

RESUMEN

AIMS/HYPOTHESIS: To overcome the donor shortage in the treatment of advanced type 1 diabetes by islet transplantation, human embryonic stem cells (hESCs) show great potential as an unlimited alternative source of beta cells. hESCs may have immune privileged properties and it is important to determine whether these properties are preserved in hESC-derived cells. METHODS: We comprehensively investigated interactions of both innate and adaptive auto- and allo-immunity with hESC-derived pancreatic progenitor cells and hESC-derived endocrine cells, retrieved after in-vivo differentiation in capsules in the subcutis of mice. RESULTS: We found that hESC-derived pancreatic endodermal cells expressed relatively low levels of HLA endorsing protection from specific immune responses. HLA was upregulated when exposed to IFNγ, making these endocrine progenitor cells vulnerable to cytotoxic T cells and alloreactive antibodies. In vivo-differentiated endocrine cells were protected from complement, but expressed more HLA and were targets for alloreactive antibody-dependent cellular cytotoxicity and alloreactive cytotoxic T cells. After HLA compatibility was provided by transduction with HLA-A2, preproinsulin-specific T cells killed insulin-producing cells. CONCLUSIONS/INTERPRETATION: hESC-derived pancreatic progenitors are hypoimmunogenic, while in vivo-differentiated endocrine cells represent mature targets for adaptive immune responses. Our data support the need for immune intervention in transplantation of hESC-derived pancreatic progenitors. Cell-impermeable macro-encapsulation may suffice.


Asunto(s)
Células Madre Embrionarias Humanas/inmunología , Células Secretoras de Insulina/inmunología , Células Madre/metabolismo , Inmunidad Adaptativa/inmunología , Aloinjertos , Autoinmunidad , Células Cultivadas , Antígeno HLA-A2 , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Inmunidad Humoral/inmunología , Inmunidad Innata/inmunología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Interferón gamma/metabolismo
6.
Stem Cells Transl Med ; 4(10): 1214-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26304037

RESUMEN

UNLABELLED: The PEC-01 cell population, differentiated from human embryonic stem cells (hESCs), contains pancreatic progenitors (PPs) that, when loaded into macroencapsulation devices (to produce the VC-01 candidate product) and transplanted into mice, can mature into glucose-responsive insulin-secreting cells and other pancreatic endocrine cells involved in glucose metabolism. We modified the protocol for making PEC-01 cells such that 73%-80% of the cell population consisted of PDX1-positive (PDX1+) and NKX6.1+ PPs. The PPs were further differentiated to islet-like cells (ICs) that reproducibly contained 73%-89% endocrine cells, of which approximately 40%-50% expressed insulin. A large fraction of these insulin-positive cells were single hormone-positive and expressed the transcription factors PDX1 and NKX6.1. To preclude a significant contribution of progenitors to the in vivo function of ICs, we used a simple enrichment process to remove remaining PPs, yielding aggregates that contained 93%-98% endocrine cells and 1%-3% progenitors. Enriched ICs, when encapsulated and implanted into mice, functioned similarly to the VC-01 candidate product, demonstrating conclusively that in vitro-produced hESC-derived insulin-producing cells can mature and function in vivo in devices. A scaled version of our suspension culture was used, and the endocrine aggregates could be cryopreserved and retain functionality. Although ICs expressed multiple important ß cell genes, the cells contained relatively low levels of several maturity-associated markers. Correlating with this, the time to function of ICs was similar to PEC-01 cells, indicating that ICs required cell-autonomous maturation after delivery in vivo, which would occur concurrently with graft integration into the host. SIGNIFICANCE: Type 1 diabetes (T1D) affects approximately 1.25 million people in the U.S. alone and is deadly if not managed with insulin injections. This paper describes the production of insulin-producing cells in vitro and a new protocol for producing the cells, representing another potential cell source for a diabetes cell therapy. These cells can be loaded into a protective device that is implanted under the skin. The device is designed to protect the cells from immune rejection by the implant recipient. The implant can engraft and respond to glucose by secreting insulin, thus potentially replacing the ß cells lost in patients with T1D.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células Secretoras de Insulina/citología , Insulina/biosíntesis , Animales , Biomarcadores , Glucemia/análisis , Diferenciación Celular , Separación Celular/métodos , Células Cultivadas , Células Inmovilizadas/trasplante , Criopreservación , Perfilación de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Ratones , Proinsulina/metabolismo , Procesamiento Proteico-Postraduccional , Reproducibilidad de los Resultados , Transactivadores/biosíntesis
7.
Cell Stem Cell ; 16(2): 148-57, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25533131

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease caused by T cell-mediated destruction of insulin-producing ß cells in the islets of Langerhans. In most cases, reversal of disease would require strategies combining islet cell replacement with immunotherapy that are currently available only for the most severely affected patients. Here, we demonstrate that immunotherapies that target T cell costimulatory pathways block the rejection of xenogeneic human embryonic-stem-cell-derived pancreatic endoderm (hESC-PE) in mice. The therapy allowed for long-term development of hESC-PE into islet-like structures capable of producing human insulin and maintaining normoglycemia. Moreover, short-term costimulation blockade led to robust immune tolerance that could be transferred independently of regulatory T cells. Importantly, costimulation blockade prevented the rejection of allogeneic hESC-PE by human PBMCs in a humanized model in vivo. These results support the clinical development of hESC-derived therapy, combined with tolerogenic treatments, as a sustainable alternative strategy for patients with T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Endodermo/citología , Endodermo/inmunología , Células Madre Embrionarias Humanas/trasplante , Tolerancia Inmunológica/inmunología , Páncreas/citología , Linfocitos T Reguladores/inmunología , Animales , Diabetes Mellitus Tipo 1/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Páncreas/inmunología , Trasplante Heterólogo
8.
Am J Physiol Endocrinol Metab ; 307(9): E838-46, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25205822

RESUMEN

ß-Cells generated from large-scale sources can overcome current shortages in clinical islet cell grafts provided that they adequately respond to metabolic variations. Pancreatic (non)endocrine cells can develop from human embryonic stem (huES) cells following in vitro derivation to pancreatic endoderm (PE) that is subsequently implanted in immune-incompetent mice for further differentiation. Encapsulation of PE increases the proportion of endocrine cells in subcutaneous implants, with enrichment in ß-cells when they are placed in TheraCyte-macrodevices and predominantly α-cells when they are alginate-microencapsulated. At posttransplant (PT) weeks 20-30, macroencapsulated huES implants presented higher glucose-responsive plasma C-peptide levels and a lower proinsulin-over-C-peptide ratio than human islet cell implants under the kidney capsule. Their ex vivo analysis showed the presence of single-hormone-positive α- and ß-cells that exhibited rapid secretory responses to increasing and decreasing glucose concentrations, similar to isolated human islet cells. However, their insulin secretory amplitude was lower, which was attributed in part to a lower cellular hormone content; it was associated with a lower glucose-induced insulin biosynthesis, but not with lower glucagon-induced stimulation, which together is compatible with an immature functional state of the huES-derived ß-cells at PT weeks 20-30. These data support the therapeutic potential of macroencapsulated huES implants but indicate the need for further functional analysis. Their comparison with clinical-grade human islet cell grafts sets references for future development and clinical translation.


Asunto(s)
Células Inmovilizadas/trasplante , Diabetes Mellitus Tipo 1/cirugía , Células Madre Embrionarias/trasplante , Implantes Experimentales/efectos adversos , Trasplante de Islotes Pancreáticos/efectos adversos , Trasplante Heterólogo/efectos adversos , Trasplante Heterotópico/efectos adversos , Animales , Péptido C/sangre , Péptido C/metabolismo , Diferenciación Celular , Línea Celular , Células Inmovilizadas/citología , Células Inmovilizadas/metabolismo , Cruzamientos Genéticos , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Riñón , Membranas , Ratones Endogámicos NOD , Ratones SCID , Proinsulina/sangre , Proinsulina/metabolismo , Tejido Subcutáneo , Andamios del Tejido/efectos adversos
9.
Cell Stem Cell ; 12(2): 224-37, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23318056

RESUMEN

Embryonic development is characterized by dynamic changes in gene expression, yet the role of chromatin remodeling in these cellular transitions remains elusive. To address this question, we profiled the transcriptome and select chromatin modifications at defined stages during pancreatic endocrine differentiation of human embryonic stem cells. We identify removal of Polycomb group (PcG)-mediated repression on stage-specific genes as a key mechanism for the induction of developmental regulators. Furthermore, we discover that silencing of transitory genes during lineage progression associates with reinstatement of PcG-dependent repression. Significantly, in vivo- but not in vitro-differentiated endocrine cells exhibit close similarity to primary human islets in regard to transcriptome and chromatin structure. We further demonstrate that endocrine cells produced in vitro do not fully eliminate PcG-mediated repression on endocrine-specific genes, probably contributing to their malfunction. These studies reveal dynamic chromatin remodeling during developmental lineage progression and identify possible strategies for improving cell differentiation in culture.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Células Madre Embrionarias/citología , Páncreas/citología , Proteínas del Grupo Polycomb/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Ensamble y Desensamble de Cromatina/genética , Células Madre Embrionarias/metabolismo , Células Endocrinas/citología , Células Endocrinas/metabolismo , Endodermo/citología , Endodermo/metabolismo , Humanos , Ratones , Proteínas del Grupo Polycomb/genética
10.
PLoS One ; 7(5): e37004, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22623968

RESUMEN

Development of a human embryonic stem cell (hESC)-based therapy for type 1 diabetes will require the translation of proof-of-principle concepts into a scalable, controlled, and regulated cell manufacturing process. We have previously demonstrated that hESC can be directed to differentiate into pancreatic progenitors that mature into functional glucose-responsive, insulin-secreting cells in vivo. In this study we describe hESC expansion and banking methods and a suspension-based differentiation system, which together underpin an integrated scalable manufacturing process for producing pancreatic progenitors. This system has been optimized for the CyT49 cell line. Accordingly, qualified large-scale single-cell master and working cGMP cell banks of CyT49 have been generated to provide a virtually unlimited starting resource for manufacturing. Upon thaw from these banks, we expanded CyT49 for two weeks in an adherent culture format that achieves 50-100 fold expansion per week. Undifferentiated CyT49 were then aggregated into clusters in dynamic rotational suspension culture, followed by differentiation en masse for two weeks with a four-stage protocol. Numerous scaled differentiation runs generated reproducible and defined population compositions highly enriched for pancreatic cell lineages, as shown by examining mRNA expression at each stage of differentiation and flow cytometry of the final population. Islet-like tissue containing glucose-responsive, insulin-secreting cells was generated upon implantation into mice. By four- to five-months post-engraftment, mature neo-pancreatic tissue was sufficient to protect against streptozotocin (STZ)-induced hyperglycemia. In summary, we have developed a tractable manufacturing process for the generation of functional pancreatic progenitors from hESC on a scale amenable to clinical entry.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Diferenciación Celular/fisiología , Diabetes Mellitus Tipo 1/terapia , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Células Secretoras de Insulina/citología , Análisis de Varianza , Animales , Criopreservación/métodos , Células Madre Embrionarias/fisiología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones SCID , Estreptozocina
11.
Nat Biotechnol ; 29(8): 750-6, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804561

RESUMEN

Using a flow cytometry-based screen of commercial antibodies, we have identified cell-surface markers for the separation of pancreatic cell types derived from human embryonic stem (hES) cells. We show enrichment of pancreatic endoderm cells using CD142 and of endocrine cells using CD200 and CD318. After transplantation into mice, enriched pancreatic endoderm cells give rise to all the pancreatic lineages, including functional insulin-producing cells, demonstrating that they are pancreatic progenitors. In contrast, implanted, enriched polyhormonal endocrine cells principally give rise to glucagon cells. These antibodies will aid investigations that use pancreatic cells generated from pluripotent stem cells to study diabetes and pancreas biology.


Asunto(s)
Antígenos CD/metabolismo , Biomarcadores/metabolismo , Separación Celular/métodos , Células Madre Embrionarias/citología , Páncreas/citología , Animales , Anticuerpos/metabolismo , Células Cultivadas , Células Madre Embrionarias/metabolismo , Endodermo/citología , Citometría de Flujo , Humanos , Ratones , Ratones SCID , Microscopía Fluorescente , Trasplante Heterólogo
12.
Nat Biotechnol ; 26(4): 443-52, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18288110

RESUMEN

Development of a cell therapy for diabetes would be greatly aided by a renewable supply of human beta-cells. Here we show that pancreatic endoderm derived from human embryonic stem (hES) cells efficiently generates glucose-responsive endocrine cells after implantation into mice. Upon glucose stimulation of the implanted mice, human insulin and C-peptide are detected in sera at levels similar to those of mice transplanted with approximately 3,000 human islets. Moreover, the insulin-expressing cells generated after engraftment exhibit many properties of functional beta-cells, including expression of critical beta-cell transcription factors, appropriate processing of proinsulin and the presence of mature endocrine secretory granules. Finally, in a test of therapeutic potential, we demonstrate that implantation of hES cell-derived pancreatic endoderm protects against streptozotocin-induced hyperglycemia. Together, these data provide definitive evidence that hES cells are competent to generate glucose-responsive, insulin-secreting cells.


Asunto(s)
Técnicas de Cultivo de Célula/tendencias , Células Madre Embrionarias/citología , Glucosa/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ingeniería de Tejidos/tendencias , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Endodermo/citología , Endodermo/metabolismo , Humanos , Células Secretoras de Insulina/trasplante , Ratones , Páncreas Artificial/tendencias
13.
Nat Biotechnol ; 24(11): 1392-401, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17053790

RESUMEN

Of paramount importance for the development of cell therapies to treat diabetes is the production of sufficient numbers of pancreatic endocrine cells that function similarly to primary islets. We have developed a differentiation process that converts human embryonic stem (hES) cells to endocrine cells capable of synthesizing the pancreatic hormones insulin, glucagon, somatostatin, pancreatic polypeptide and ghrelin. This process mimics in vivo pancreatic organogenesis by directing cells through stages resembling definitive endoderm, gut-tube endoderm, pancreatic endoderm and endocrine precursor--en route to cells that express endocrine hormones. The hES cell-derived insulin-expressing cells have an insulin content approaching that of adult islets. Similar to fetal beta-cells, they release C-peptide in response to multiple secretory stimuli, but only minimally to glucose. Production of these hES cell-derived endocrine cells may represent a critical step in the development of a renewable source of cells for diabetes cell therapy.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Células Enteroendocrinas/fisiología , Islotes Pancreáticos/crecimiento & desarrollo , Hormonas Pancreáticas/biosíntesis , Hormonas Peptídicas/biosíntesis , Células Cultivadas , Ghrelina , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Páncreas/citología , Hormonas Pancreáticas/aislamiento & purificación
14.
Blood ; 108(2): 622-9, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16469876

RESUMEN

The Hoxa9 and Meis1 genes represent important oncogenic collaborators activated in a significant proportion of human leukemias with genetic alterations in the MLL gene. In this study, we show that the transforming property of Meis1 is modulated by 3 conserved domains, namely the Pbx interaction motif (PIM), the homeodomain, and the C-terminal region recently described to possess transactivating properties. Meis1 and Pbx1 interaction domain-swapping mutants are dysfunctional separately, but restore the full oncogenic activity of Meis1 when cotransduced in primary cells engineered to overexpress Hoxa9, thus implying a modular nature for PIM in Meis1-accelerated transformation. Moreover, we show that the transactivating domain of VP16 can restore, and even enhance, the oncogenic potential of the Meis1 mutant lacking the C-terminal 49 amino acids. In contrast to Meis1, the fusion VP16-Meis1 is spontaneously oncogenic, and all leukemias harbor genetic activation of endogenous Hoxa9 and/or Hoxa7, suggesting that Hoxa gene activation represents a key event required for the oncogenic activity of VP16-Meis1.


Asunto(s)
Transformación Celular Neoplásica , Células Madre Hematopoyéticas/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Leucemia/etiología , Proteínas de Neoplasias/fisiología , Activación Transcripcional/fisiología , Animales , Células Cultivadas , Proteína Vmw65 de Virus del Herpes Simple/fisiología , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/química , Leucemia/patología , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Estructura Terciaria de Proteína , Transducción Genética
15.
Nat Biotechnol ; 23(12): 1534-41, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16258519

RESUMEN

The potential of human embryonic stem (hES) cells to differentiate into cell types of a variety of organs has generated much excitement over the possible use of hES cells in therapeutic applications. Of great interest are organs derived from definitive endoderm, such as the pancreas. We have focused on directing hES cells to the definitive endoderm lineage as this step is a prerequisite for efficient differentiation to mature endoderm derivatives. Differentiation of hES cells in the presence of activin A and low serum produced cultures consisting of up to 80% definitive endoderm cells. This population was further enriched to near homogeneity using the cell-surface receptor CXCR4. The process of definitive endoderm formation in differentiating hES cell cultures includes an apparent epithelial-to-mesenchymal transition and a dynamic gene expression profile that are reminiscent of vertebrate gastrulation. These findings may facilitate the use of hES cells for therapeutic purposes and as in vitro models of development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Endodermo/citología , Endodermo/fisiología , Células Madre/citología , Células Madre/fisiología , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Ratones
16.
Blood ; 105(2): 784-93, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15454493

RESUMEN

The chromosomal translocation t(7; 11)(p15;p15), observed in human myeloid leukemia, results in a NUP98 and HOXA9 gene fusion. We generated a transgenic mouse line that specifically expressed the chimeric NUP98-HOXA9 gene in the myeloid lineage. While only 20% of the transgenic mice progressed to leukemia after a latency period, myeloid progenitor cells from nonleukemic transgenic mice still exhibited increased proliferative potential. This suggested that the NUP98-HOXA9 fusion induced a preleukemic phase, and other factors were required for complete leukemogenesis. NUP98-HOXA9 expression promoted the onset of retrovirus-induced BXH2 myeloid leukemia. This phenomenon was used to identify cooperative disease genes as common integration sites (CISs). Meis1, a known HOX cofactor, was identified as a CIS with a higher integration frequency in transgenic than in wild-type BXH2 mice. By the same means we identified further 4 candidate cooperative genes, Dnalc4, Fcgr2b, Fcrl, and Con1. These genes cooperated with NUP98-HOXA9 in transforming NIH 3T3 cells. The system described here is a powerful tool to identify cooperative oncogenes and will assist in the clarification of the multistep process of carcinogenesis.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/genética , Leucemia Mieloide/genética , Leucemia Mieloide/fisiopatología , Proteínas de Complejo Poro Nuclear/genética , Animales , División Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Mutagénesis Insercional , Células 3T3 NIH , Retroviridae/genética
17.
Nat Med ; 9(11): 1428-32, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14578881

RESUMEN

Hematopoietic stem cells (HSCs) can self-renew extensively after transplantation. The conditions supporting their in vitro expansion are still being defined. Retroviral overexpression of the human homeobox B4 (HOXB4) gene in mouse bone marrow cells enables over 40-fold expansion of HSCs in vitro. To circumvent the requirement for retroviral infection, we used recombinant human TAT-HOXB4 protein carrying the protein transduction domain of the HIV transactivating protein (TAT) as a potential growth factor for stem cells. HSCs exposed to TAT-HOXB4 for 4 d expanded by about four- to sixfold and were 8-20 times more numerous than HSCs in control cultures, indicating that HSC expansion induced by TAT-HOXB4 was comparable to that induced by the human HOXB4 retrovirus during a similar period of observation. Our results also show that TAT-HOXB4-expanded HSC populations retain their normal in vivo potential for differentiation and long-term repopulation. It is thus feasible to exploit recombinant HOXB4 protein for rapid and significant ex vivo expansion of normal HSCs.


Asunto(s)
División Celular/fisiología , Productos del Gen tat/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Proteínas Recombinantes de Fusión/genética , Factores de Transcripción/genética , Animales , Productos del Gen tat/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Retroviridae , Factores de Transcripción/metabolismo
18.
Blood ; 100(12): 4177-84, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12393433

RESUMEN

Chronic myelogenous leukemia (CML) is a clonal stem cell disease caused by the BCR-ABL oncoprotein and is characterized, in its early phase, by excessive accumulation of mature myeloid cells, which eventually leads to acute leukemia. The genetic events involved in CML's progression to acute leukemia remain largely unknown. Recent studies have detected the presence of the NUP98-HOXA9 fusion oncogene in acute leukemia derived from CML patients, which suggests that these 2 oncoproteins may interact and influence CML disease progression. Using in vitro purging of BCR-ABL-transduced mouse bone marrow cells, we can now report that recipients of bone marrow cells engineered to coexpress BCR-ABL with NUP98-HOXA9 develop acute leukemia within 7 to 10 days after transplantation. However, no disease is detected for more than 2 months in mice receiving bone marrow cells expressing either BCR-ABL or NUP98-HOXA9. We also provide evidence of high levels of HOXA9 expressed in leukemic blasts from acute-phase CML patients and that it interacts significantly on a genetic level with BCR-ABL in our in vivo CML model. Together, these studies support a causative, as opposed to a consequential, role for NUP98-HOXA9 (and possibly HOXA9) in CML disease progression.


Asunto(s)
Purgación de la Médula Ósea/métodos , Transformación Celular Neoplásica/genética , Proteínas de Fusión bcr-abl/genética , Proteínas de Homeodominio/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Técnicas de Cultivo de Célula/métodos , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Ratones , Ratones Endogámicos C57BL , Transducción Genética
19.
Blood ; 99(1): 121-9, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11756161

RESUMEN

Cytogenetic, genetic, and functional studies have demonstrated a direct link between deregulated Hoxa9 expression and acute myeloid leukemia (AML). Hoxa9 overexpression in mouse bone marrow cells invariably leads to AML within 3 to 10 months, suggesting the requirement for additional genetic events prior to AML. To gain further insight into how Hoxa9 affects hematopoietic development at the preleukemic stage, we have engineered its overexpression (1) in hematopoietic stem cells using retrovirus-mediated gene transfer and generated bone marrow transplantation chimeras and (2) in lymphoid cells using transgenic mice. Compared with controls, recipients of Hoxa9-transduced cells had an about 15-fold increase in transplantable lymphomyeloid long-term repopulating cells, indicating the capacity for this oncogene to confer a growth advantage to hematopoietic stem cells. In addition, overexpression of Hoxa9 in more mature cells enhanced granulopoiesis and partially blocked B lymphopoiesis at the pre-B-cell stage but had no detectable effect on T lymphoid development. Interestingly, despite specifically directing high expression of Hoxa9 in T and B lymphoid lineages, none of the Hoxa9 transgenic mice developed lymphoid malignancies for the observation period of more than 18 months.


Asunto(s)
Células de la Médula Ósea/metabolismo , Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Animales , Linfocitos B/patología , Células de la Médula Ósea/patología , Recuento de Células , División Celular , Regulación de la Expresión Génica , Granulocitos , Hematopoyesis , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Retroviridae/genética , Linfocitos T/patología , Transfección , Quimera por Trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA