Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
STAR Protoc ; 5(1): 102827, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38219151

RESUMEN

Here, we present a protocol to differentiate induced pluripotent stem cell (iPSC) into adherent hematopoietic progenitors that release floating CD14+ CD45+ monocytes into the culture medium. We describe steps for iPSC expansion, embryoid body (EB) formation, suspension culture, plating EBs, and recurring harvests of monocytes, a.k.a. "monocyte factory." We then describe detailed procedures for freezing/thawing of monocytes and differentiation into polarized M1 and M2 macrophages. This protocol provides foundation to study iPSC monocytes and their progenies such as macrophages, microglial, and dendritic cells. For complete details on the use and execution of this protocol, please refer to Karlson et al.1 and Panicker et al.2.


Asunto(s)
Células Madre Pluripotentes Inducidas , Monocitos , Humanos , Macrófagos , Diferenciación Celular , Cuerpos Embrioides
2.
STAR Protoc ; 4(2): 102292, 2023 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-37149860

RESUMEN

Here, we present a protocol for differentiating human-induced pluripotent stem cells into three distinct mesodermal cell types: vascular endothelial cells (ECs), pericytes, and fibroblasts. We describe steps for using monolayer serum-free differentiation and isolating ECs (CD31+) and mesenchymal pre-pericytes (CD31-) from a single differentiation set. We then differentiate pericytes into fibroblasts using a commercial fibroblast culture medium. The three cell types differentiated in this protocol are useful for vasculogenesis, drug testing, and tissue engineering applications. For complete details on the use and execution of this protocol, please refer to Orlova et al. (2014).1.

3.
Nat Methods ; 20(1): 149-161, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36550275

RESUMEN

Age-related macular degeneration (AMD), a leading cause of blindness, initiates in the outer-blood-retina-barrier (oBRB) formed by the retinal pigment epithelium (RPE), Bruch's membrane, and choriocapillaris. The mechanisms of AMD initiation and progression remain poorly understood owing to the lack of physiologically relevant human oBRB models. To this end, we engineered a native-like three-dimensional (3D) oBRB tissue (3D-oBRB) by bioprinting endothelial cells, pericytes, and fibroblasts on the basal side of a biodegradable scaffold and establishing an RPE monolayer on top. In this 3D-oBRB model, a fully-polarized RPE monolayer provides barrier resistance, induces choriocapillaris fenestration, and supports the formation of Bruch's-membrane-like structure by inducing changes in gene expression in cells of the choroid. Complement activation in the 3D-oBRB triggers dry AMD phenotypes (including subRPE lipid-rich deposits called drusen and choriocapillaris degeneration), and HIF-α stabilization or STAT3 overactivation induce choriocapillaris neovascularization and type-I wet AMD phenotype. The 3D-oBRB provides a physiologically relevant model to studying RPE-choriocapillaris interactions under healthy and diseased conditions.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Humanos , Epitelio Pigmentado de la Retina/metabolismo , Células Endoteliales , Coroides/metabolismo , Retina/metabolismo , Degeneración Macular/metabolismo
4.
Methods Mol Biol ; 2416: 133-156, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34870835

RESUMEN

Tankyrase/PARP inhibitor-regulated naïve human pluripotent stem cells (TIRN-hPSC) represent a new class of human stem cells for regenerative medicine that can differentiate into multi-lineage progenitors with improved in vivo functionality. Chemical reversion of conventional, primed hPSC to a TIRN-hPSC state alleviates dysfunctional epigenetic donor cell memory, lineage-primed gene expression, and potentially disease-associated aberrations in their differentiated progeny. Here, we provide methods for the reversion of normal or diseased patient-specific primed hPSC to TIRN-hPSC and describe their subsequent differentiation into embryonic-like pericytic-endothelial "naïve" vascular progenitors (N-VP). N-VP possess improved vascular functionality, high epigenetic plasticity, maintain greater genomic stability, and are more efficient in migrating to and re-vascularizing ischemic tissues than those generated from primed isogenic hPSC. We also describe detailed methods for the ocular transplantation and quantitation of vascular engraftment of N-VP into the ischemia-damaged neural retina of a humanized mouse model of ischemic retinopathy. The application of TIRN-hPSC-derived N-VP will advance vascular cell therapies of ischemic retinopathy, myocardial infarction, and cerebral vascular stroke.


Asunto(s)
Células Madre Pluripotentes , Animales , Diferenciación Celular/efectos de los fármacos , Humanos , Isquemia , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Enfermedades de la Retina , Tanquirasas
5.
Stem Cells Transl Med ; 10(7): 1081-1094, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33656802

RESUMEN

Gaucher disease (GD) is a lysosomal storage disorder caused by mutations in GBA1, the gene that encodes lysosomal ß-glucocerebrosidase (GCase). Mild mutations in GBA1 cause type 1 non-neuronopathic GD, whereas severe mutations cause types 2 and 3 neuronopathic GD (nGD). GCase deficiency results in the accumulation of glucosylceramide (GlcCer) and glucosylsphingosine (GlcSph). GlcSph is formed by deacylation of GlcCer by the lysosomal enzyme acid ceramidase. Brains from patients with nGD have high levels of GlcSph, a lipid believed to play an important role in nGD, but the mechanisms involved remain unclear. To identify these mechanisms, we used human induced pluripotent stem cell-derived neurons from nGD patients. We found that elevated levels of GlcSph activate mammalian target of rapamycin (mTOR) complex 1 (mTORC1), interfering with lysosomal biogenesis and autophagy, which were restored by incubation of nGD neurons with mTOR inhibitors. We also found that inhibition of acid ceramidase prevented both, mTOR hyperactivity and lysosomal dysfunction, suggesting that these alterations were caused by GlcSph accumulation in the mutant neurons. To directly determine whether GlcSph can cause mTOR hyperactivation, we incubated wild-type neurons with exogenous GlcSph. Remarkably, GlcSph treatment recapitulated the mTOR hyperactivation and lysosomal abnormalities in mutant neurons, which were prevented by coincubation of GlcSph with mTOR inhibitors. We conclude that elevated GlcSph activates an mTORC1-dependent pathogenic mechanism that is responsible for the lysosomal abnormalities of nGD neurons. We also identify acid ceramidase as essential to the pathogenesis of nGD, providing a new therapeutic target for treating GBA1-associated neurodegeneration.


Asunto(s)
Enfermedad de Gaucher , Células Madre Pluripotentes Inducidas , Diana Mecanicista del Complejo 1 de la Rapamicina , Neuronas , Psicosina/análogos & derivados , Ceramidasa Ácida/antagonistas & inhibidores , Enfermedad de Gaucher/tratamiento farmacológico , Enfermedad de Gaucher/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Lisosomas , Inhibidores mTOR , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neuronas/citología , Psicosina/sangre
6.
Nat Commun ; 11(1): 1195, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32139672

RESUMEN

Here, we report that the functionality of vascular progenitors (VP) generated from normal and disease-primed conventional human induced pluripotent stem cells (hiPSC) can be significantly improved by reversion to a tankyrase inhibitor-regulated human naïve epiblast-like pluripotent state. Naïve diabetic vascular progenitors (N-DVP) differentiated from patient-specific naïve diabetic hiPSC (N-DhiPSC) possessed higher vascular functionality, maintained greater genomic stability, harbored decreased lineage-primed gene expression, and were more efficient in migrating to and re-vascularizing the deep neural layers of the ischemic retina than isogenic diabetic vascular progenitors (DVP). These findings suggest that reprogramming to a stable naïve human pluripotent stem cell state may effectively erase dysfunctional epigenetic donor cell memory or disease-associated aberrations in patient-specific hiPSC. More broadly, tankyrase inhibitor-regulated naïve hiPSC (N-hiPSC) represent a class of human stem cells with high epigenetic plasticity, improved multi-lineage functionality, and potentially high impact for regenerative medicine.


Asunto(s)
Vasos Sanguíneos/patología , Diabetes Mellitus/patología , Células Madre Pluripotentes Inducidas/patología , Isquemia/terapia , Retina/patología , Células Madre/patología , Tanquirasas/antagonistas & inhibidores , Adulto , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Daño del ADN , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Código de Histonas , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Isquemia/patología , Ratones , Organoides/efectos de los fármacos , Organoides/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Regiones Promotoras Genéticas/genética , Células Madre/efectos de los fármacos , Células Madre/ultraestructura , Tanquirasas/metabolismo , Teratoma/patología , Transcripción Genética/efectos de los fármacos
7.
J Vis Exp ; (136)2018 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-29939183

RESUMEN

Naïve human pluripotent stem cells (N-hPSC) with improved functionality may have a wide impact in regenerative medicine. The goal of this protocol is to efficiently revert lineage-primed, conventional human pluripotent stem cells (hPSC) maintained on either feeder-free or feeder-dependent conditions to a naïve-like pluripotency with improved functionality. This chemical naïve reversion method employs the classical leukemia inhibitory factor (LIF), GSK3ß, and MEK/ERK inhibition cocktail (LIF-2i), supplemented with only a tankyrase inhibitor XAV939 (LIF-3i). LIF-3i reverts conventional hPSC to a stable pluripotent state adopting biochemical, transcriptional, and epigenetic features of the human pre-implantation epiblast. This LIF-3i method requires minimal cell culture manipulation and is highly reproducible in a broad repertoire of human embryonic stem cell (hESC) and transgene-free human induced pluripotent stem cell (hiPSC) lines. The LIF-3i method does not require a re-priming step prior to the differentiation; N-hPSC can be differentiated directly with extremely high efficiencies and maintain karyotypic and epigenomic stabilities (including at imprinted loci). To increase the universality of the method, conventional hPSC are first cultured in the LIF-3i cocktail supplemented with two additional small molecules that potentiate protein kinase A (forskolin) and sonic hedgehog (sHH) (purmorphamine) signaling (LIF-5i). This brief LIF-5i adaptation step significantly enhances the initial clonal expansion of conventional hPSC and permits them to be subsequently naïve-reverted with LIF-3i alone in bulk quantities, thus obviating the need for picking/subcloning rare N-hPSC colonies later. LIF-5i-stabilized hPSCs are subsequently maintained in LIF-3i alone without the need of anti-apoptotic molecules. Most importantly, LIF-3i reversion markedly improves the functional pluripotency of a broad repertoire of conventional hPSC by decreasing their lineage-primed gene expression and erasing the interline variability of directed differentiation commonly observed amongst independent hPSC lines. Representative characterizations of LIF-3i-reverted N-hPSC are provided, and experimental strategies for functional comparisons of isogenic hPSC in lineage-primed vs. naïve-like states are outlined.


Asunto(s)
Estratos Germinativos/metabolismo , Células Madre Pluripotentes/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Humanos , Células Madre Pluripotentes/citología , Transducción de Señal
8.
Stem Cell Reports ; 9(6): 1853-1867, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29198828

RESUMEN

Gaucher's disease (GD) is an autosomal recessive disorder caused by mutations in the GBA1 gene, which encodes acid ß-glucocerebrosidase (GCase). Severe GBA1 mutations cause neuropathology that manifests soon after birth, suggesting that GCase deficiency interferes with neuronal development. We found that neuronopathic GD induced pluripotent stem cell (iPSC)-derived neuronal progenitor cells (NPCs) exhibit developmental defects due to downregulation of canonical Wnt/ß-catenin signaling and that GD iPSCs' ability to differentiate to dopaminergic (DA) neurons was strikingly reduced due to early loss of DA progenitors. Incubation of the mutant cells with the Wnt activator CHIR99021 (CHIR) or with recombinant GCase restored Wnt/ß-catenin signaling and rescued DA differentiation. We also found that GD NPCs exhibit lysosomal dysfunction, which may be involved in Wnt downregulation by mutant GCase. We conclude that neuronopathic mutations in GCase lead to neurodevelopmental abnormalities due to a critical requirement of this enzyme for canonical Wnt/ß-catenin signaling at early stages of neurogenesis.


Asunto(s)
Diferenciación Celular/genética , Neuronas Dopaminérgicas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Neurogénesis/genética , Neuronas Dopaminérgicas/patología , Enfermedad de Gaucher/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lisosomas/genética , Lisosomas/patología , Mutación , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Piridinas/administración & dosificación , Pirimidinas/administración & dosificación , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
9.
Stem Cells Dev ; 26(16): 1141-1161, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28537488

RESUMEN

Although human embryonic stem cells (hESCs) were first derived almost 20 years ago, it was only recently acknowledged that they share closer molecular and functional identity to postimplantation lineage-primed murine epiblast stem cells than to naïve preimplantation inner cell mass-derived mouse ESCs (mESCs). A myriad of transcriptional, epigenetic, biochemical, and metabolic attributes have now been described that distinguish naïve and primed pluripotent states in both rodents and humans. Conventional hESCs and human induced pluripotent stem cells (hiPSCs) appear to lack many of the defining hallmarks of naïve mESCs. These include important features of the naïve ground state murine epiblast, such as an open epigenetic architecture, reduced lineage-primed gene expression, and chimera and germline competence following injection into a recipient blastocyst-stage embryo. Several transgenic and chemical methods were recently reported that appear to revert conventional human PSCs to mESC-like ground states. However, it remains unclear if subtle deviations in global transcription, cell signaling dependencies, and extent of epigenetic/metabolic shifts in these various human naïve-reverted pluripotent states represent true functional differences or alternatively the existence of distinct human pluripotent states along a spectrum. In this study, we review the current understanding and developmental features of various human pluripotency-associated phenotypes and discuss potential biological mechanisms that may support stable maintenance of an authentic epiblast-like ground state of human pluripotency.


Asunto(s)
Blastocisto/citología , Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Células Madre Pluripotentes/citología , Animales , Blastocisto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Células Madre Pluripotentes/metabolismo
10.
Stem Cells Int ; 2016: 3826249, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27688775

RESUMEN

Human induced pluripotent stem cells (hiPSCs) are reprogrammed from adult or progenitor somatic cells and must make substantial adaptations to ensure genomic stability in order to become "embryonic stem cell- (ESC-) like." The DNA damage response (DDR) is critical for maintenance of such genomic integrity. Herein, we determined whether cell of origin and reprogramming method influence the DDR of hiPSCs. We demonstrate that hiPSCs derived from cord blood (CB) myeloid progenitors (i.e., CB-iPSC) via an efficient high-fidelity stromal-activated (sa) method closely resembled hESCs in DNA repair gene expression signature and irradiation-induced DDR, relative to hiPSCs generated from CB or fibroblasts via standard methods. Furthermore, sa-CB-iPSCs also more closely resembled hESCs in accuracy of nonhomologous end joining (NHEJ), DNA double-strand break (DSB) repair, and C-MYC transcriptional signatures, relative to standard hiPSCs. Our data suggests that hiPSCs derived via more efficient reprogramming methods possess more hESC-like activated MYC signatures and DDR signaling. Thus, an authentic MYC molecular signature may serve as an important biomarker in characterizing the genomic integrity in hiPSCs.

11.
Development ; 143(23): 4368-4380, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27660325

RESUMEN

The derivation and maintenance of human pluripotent stem cells (hPSCs) in stable naïve pluripotent states has a wide impact in human developmental biology. However, hPSCs are unstable in classical naïve mouse embryonic stem cell (ESC) WNT and MEK/ERK signal inhibition (2i) culture. We show that a broad repertoire of conventional hESC and transgene-independent human induced pluripotent stem cell (hiPSC) lines could be reverted to stable human preimplantation inner cell mass (ICM)-like naïve states with only WNT, MEK/ERK, and tankyrase inhibition (LIF-3i). LIF-3i-reverted hPSCs retained normal karyotypes and genomic imprints, and attained defining mouse ESC-like functional features, including high clonal self-renewal, independence from MEK/ERK signaling, dependence on JAK/STAT3 and BMP4 signaling, and naïve-specific transcriptional and epigenetic configurations. Tankyrase inhibition promoted a stable acquisition of a human preimplantation ICM-like ground state via modulation of WNT signaling, and was most efficacious in efficiently reprogrammed conventional hiPSCs. Importantly, naïve reversion of a broad repertoire of conventional hiPSCs reduced lineage-primed gene expression and significantly improved their multilineage differentiation capacities. Stable naïve hPSCs with reduced genetic variability and improved functional pluripotency will have great utility in regenerative medicine and human disease modeling.


Asunto(s)
Diferenciación Celular/fisiología , Autorrenovación de las Células/fisiología , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Tanquirasas/antagonistas & inhibidores , Vía de Señalización Wnt/fisiología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Células Cultivadas , Reprogramación Celular/fisiología , Estratos Germinativos/embriología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Quinasas Janus/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Ratones , Factor de Transcripción STAT3/metabolismo
12.
Arthritis Rheumatol ; 68(10): 2540-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27159521

RESUMEN

OBJECTIVE: Scleroderma patients with autoantibodies to CENPs and/or interferon-inducible protein 16 (IFI-16) are at increased risk of severe vascular complications. This study was undertaken to determine whether these autoantigens are enriched in cells of the vasculature. METHODS: Successive stages of embryoid bodies (EBs) as well as vascular progenitors were used to evaluate the expression of scleroderma autoantigens IFI-16 and CENP by immunoblotting. CD31 was included to mark early blood vessels. IFI-16 and CD31 expression were defined in paraffin-embedded skin sections from scleroderma patients and from healthy controls. IFI-16 expression was determined by flow cytometric analysis in circulating endothelial cells (CECs) and circulating hematopoietic progenitor cells. RESULTS: Expression of CENP-A, IFI-16, and CD31 was enriched in EBs on days 10 and 12 of differentiation, and particularly in cultures enriched in vascular progenitors (IFI-16, CD31, and CENPs A and B). This pattern was distinct from that of comparator autoantigens. Immunohistochemical staining of paraffin-embedded skin sections showed enrichment of IFI-16 in CD31-positive vascular endothelial cells in biopsy specimens from scleroderma patients and normal controls. Flow cytometric analysis revealed IFI-16 expression in circulating hematopoietic progenitor cells but minimal expression in CECs. CONCLUSION: Our findings indicate that expression of the scleroderma autoantigens IFI-16 and CENPs, which are associated with severe vascular disease, is increased in vascular progenitors and mature endothelial cells. High level, lineage-enriched expression of autoantigens may explain the striking association between clinical phenotypes and the immune targeting of specific autoantigens.


Asunto(s)
Autoantígenos/inmunología , Proteína B del Centrómero/inmunología , Proteínas Cromosómicas no Histona/inmunología , Cuerpos Embrioides/inmunología , Células Endoteliales/inmunología , Células Progenitoras Endoteliales/inmunología , Proteínas Nucleares/inmunología , Fosfoproteínas/inmunología , Esclerodermia Sistémica/inmunología , Autoantígenos/metabolismo , Estudios de Casos y Controles , Linaje de la Célula , Proteína A Centromérica , Proteína B del Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cuerpos Embrioides/metabolismo , Células Endoteliales/metabolismo , Células Progenitoras Endoteliales/metabolismo , Citometría de Flujo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Leucocitos Mononucleares , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Esclerodermia Difusa/inmunología , Esclerodermia Difusa/metabolismo , Esclerodermia Limitada/inmunología , Esclerodermia Limitada/metabolismo , Esclerodermia Sistémica/metabolismo , Piel/inmunología , Piel/metabolismo
13.
Stem Cells Transl Med ; 4(8): 878-86, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26062980

RESUMEN

Gaucher disease (GD) is the most common lysosomal storage disease resulting from mutations in the lysosomal enzyme glucocerebrosidase (GCase). The hematopoietic abnormalities in GD include the presence of characteristic Gaucher macrophages that infiltrate patient tissues and cytopenias. At present, it is not clear whether these cytopenias are secondary to the pathological activity of Gaucher cells or a direct effect of GCase deficiency on hematopoietic development. To address this question, we differentiated induced pluripotent stem cells (iPSCs) derived from patients with types 1, 2, and 3 GD to CD34(+)/CD45(+)/CD43(+)/CD143(+) hematopoietic progenitor cells (HPCs) and examined their developmental potential. The formation of GD-HPCs was unaffected. However, these progenitors demonstrated a skewed lineage commitment, with increased myeloid differentiation and decreased erythroid differentiation and maturation. Interestingly, myeloid colony-formation assays revealed that GD-HPCs, but not control-HPCs, gave rise to adherent, macrophage-like cells, another indication of abnormal myelopoiesis. The extent of these hematologic abnormalities correlated with the severity of the GCase mutations. All the phenotypic abnormalities of GD-HPCs observed were reversed by incubation with recombinant GCase, indicating that these developmental defects were caused by the mutated GCase. Our results show that GCase deficiency directly impairs hematopoietic development. Additionally, our results suggest that aberrant myelopoiesis might contribute to the pathological properties of Gaucher macrophages, which are central to GD manifestations. The hematopoietic developmental defects we observed reflect hematologic abnormalities in patients with GD, demonstrating the utility of GD-iPSCs for modeling this disease.


Asunto(s)
Enfermedad de Gaucher/terapia , Hematopoyesis , Células Madre Pluripotentes Inducidas/trasplante , Mielopoyesis/fisiología , Diferenciación Celular , Linaje de la Célula , Enfermedad de Gaucher/patología , Células Madre Hematopoyéticas/patología , Humanos , Macrófagos/patología
14.
Stem Cells Dev ; 23(24): 2996-3010, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25027006

RESUMEN

Although the majority of Alzheimer's disease (AD) cases are sporadic, about 5% of cases are inherited in an autosomal dominant pattern as familial AD (FAD) and manifest at an early age. Mutations in the presenilin 1 (PSEN1) gene account for the majority of early-onset FAD. Here, we describe the generation of virus-free human induced pluripotent stem cells (hiPSCs) derived from fibroblasts of patients harboring the FAD PSEN1 mutation A246E and fibroblasts from healthy age-matched controls using nonintegrating episomal vectors. We have differentiated these hiPSC lines to the neuronal lineage and demonstrated that hiPSC-derived neurons have mature phenotypic and physiological properties. Neurons from mutant hiPSC lines express PSEN1-A246E mutations themselves and show AD-like biochemical features, that is, amyloidogenic processing of amyloid precursor protein (APP) indicated by an increase in ß-amyloid (Aß)42/Aß40 ratio. FAD hiPSCs harboring disease properties can be used as humanized models to test novel diagnostic methods and therapies and explore novel hypotheses for AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Células Madre Pluripotentes Inducidas/citología , Neurogénesis , Neuronas/citología , Potenciales de Acción , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Reprogramación Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Mutación Missense , Neuronas/metabolismo , Neuronas/fisiología , Presenilina-1/genética
15.
Nat Commun ; 5: 4047, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24915161

RESUMEN

Many forms of blindness result from the dysfunction or loss of retinal photoreceptors. Induced pluripotent stem cells (iPSCs) hold great potential for the modelling of these diseases or as potential therapeutic agents. However, to fulfill this promise, a remaining challenge is to induce human iPSC to recreate in vitro key structural and functional features of the native retina, in particular the presence of photoreceptors with outer-segment discs and light sensitivity. Here we report that hiPSC can, in a highly autonomous manner, recapitulate spatiotemporally each of the main steps of retinal development observed in vivo and form three-dimensional retinal cups that contain all major retinal cell types arranged in their proper layers. Moreover, the photoreceptors in our hiPSC-derived retinal tissue achieve advanced maturation, showing the beginning of outer-segment disc formation and photosensitivity. This success brings us one step closer to the anticipated use of hiPSC for disease modelling and open possibilities for future therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Fotorreceptoras de Vertebrados/fisiología , Retina/citología , Diferenciación Celular , Línea Celular , Humanos , Retina/fisiología
16.
Stem Cells ; 32(9): 2338-49, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24801745

RESUMEN

Gaucher disease (GD) is an autosomal recessive disorder caused by mutations in the acid ß-glucocerebrosidase (GCase; GBA) gene. The hallmark of GD is the presence of lipid-laden Gaucher macrophages, which infiltrate bone marrow and other organs. These pathological macrophages are believed to be the sources of elevated levels of inflammatory mediators present in the serum of GD patients. The alteration in the immune environment caused by GD is believed to play a role in the increased risk of developing multiple myeloma and other malignancies in GD patients. To determine directly whether Gaucher macrophages are abnormally activated and whether their functional defects can be reversed by pharmacological intervention, we generated GD macrophages by directed differentiation of human induced pluripotent stem cells (hiPSC) derived from patients with types 1, 2, and 3 GD. GD hiPSC-derived macrophages expressed higher levels of tumor necrosis factor α, IL-6, and IL-1ß than control cells, and this phenotype was exacerbated by treatment with lipopolysaccharide. In addition, GD hiPSC macrophages exhibited a striking delay in clearance of phagocytosed red blood cells, recapitulating the presence of red blood cell remnants in Gaucher macrophages from bone marrow aspirates. Incubation of GD hiPSC macrophages with recombinant GCase, or with the chaperones isofagomine and ambroxol, corrected the abnormal phenotypes of GD macrophages to an extent that reflected their known clinical efficacies. We conclude that Gaucher macrophages are the likely source of the elevated levels of inflammatory mediators in the serum of GD patients and that GD hiPSC are valuable new tools for studying disease mechanisms and drug discovery.


Asunto(s)
Citocinas/biosíntesis , Enfermedad de Gaucher/metabolismo , Enfermedad de Gaucher/patología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Femenino , Enfermedad de Gaucher/genética , Humanos , Células Madre Pluripotentes Inducidas/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID
17.
Curr Pathobiol Rep ; 2(1): 41-52, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24660130

RESUMEN

The cancer stem cell (CSC) paradigm presumes the existence of self-renewing cancer cells capable of regenerating all tumor compartments and exhibiting stem cell-associated phenotypes. Recent interpretations of the CSC hypothesis envision stemness as a dynamic trait of tumor-initiating cells rather than a defined and unique cell type. Bidirectional crosstalk between the tumor microenvironment and the cancer bulk is well described in the literature and the tumor-associated stroma, vasculature and immune infiltrate have all been implicated as direct contributors to tumor development. These non-neoplastic cell types have also been shown to organize specific niches within the tumor bulk where they can control the intra-tumor CSC content and alter the fate of CSCs and tumor progenitors during tumorigenesis to acquire phenotypic features for invasion, metastasis and dormancy. Despite the complexity of the tumor-stroma interactome, novel therapeutic approaches envision combining tumor-ablative treatment with manipulation of the tumor microenvironment. We will review the currently available literature that provides clues about the complex cellular network that regulate the CSC phenotype and its niches during tumor progression.

18.
Cell Mol Life Sci ; 71(8): 1353-74, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24158496

RESUMEN

Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.


Asunto(s)
Biomarcadores/metabolismo , Vasos Sanguíneos/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/clasificación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Pericitos/citología , Técnicas de Cultivo de Célula , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación
19.
Circulation ; 129(3): 359-72, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24163065

RESUMEN

BACKGROUND: The generation of vascular progenitors (VPs) from human induced pluripotent stem cells (hiPSCs) has great potential for treating vascular disorders such as ischemic retinopathies. However, long-term in vivo engraftment of hiPSC-derived VPs into the retina has not yet been reported. This goal may be limited by the low differentiation yield, greater senescence, and poor proliferation of hiPSC-derived vascular cells. To evaluate the potential of hiPSCs for treating ischemic retinopathies, we generated VPs from a repertoire of viral-integrated and nonintegrated fibroblast and cord blood (CB)-derived hiPSC lines and tested their capacity for homing and engrafting into murine retina in an ischemia-reperfusion model. METHODS AND RESULTS: VPs from human embryonic stem cells and hiPSCs were generated with an optimized vascular differentiation system. Fluorescence-activated cell sorting purification of human embryoid body cells differentially expressing endothelial/pericytic markers identified a CD31(+)CD146(+) VP population with high vascular potency. Episomal CB-induced pluripotent stem cells (iPSCs) generated these VPs with higher efficiencies than fibroblast-iPSC. Moreover, in contrast to fibroblast-iPSC-VPs, CB-iPSC-VPs maintained expression signatures more comparable to human embryonic stem cell VPs, expressed higher levels of immature vascular markers, demonstrated less culture senescence and sensitivity to DNA damage, and possessed fewer transmitted reprogramming errors. Luciferase transgene-marked VPs from human embryonic stem cells, CB-iPSCs, and fibroblast-iPSCs were injected systemically or directly into the vitreous of retinal ischemia-reperfusion-injured adult nonobese diabetic-severe combined immunodeficient mice. Only human embryonic stem cell- and CB-iPSC-derived VPs reliably homed and engrafted into injured retinal capillaries, with incorporation into damaged vessels for up to 45 days. CONCLUSIONS: VPs generated from CB-iPSCs possessed augmented capacity to home, integrate into, and repair damaged retinal vasculature.


Asunto(s)
Células Madre Embrionarias/citología , Sangre Fetal/citología , Células Madre Pluripotentes/citología , Daño por Reperfusión/terapia , Enfermedades de la Retina/terapia , Trasplante de Células Madre/métodos , Animales , Capilares/citología , Senescencia Celular , Daño del ADN , Modelos Animales de Enfermedad , Fibroblastos/citología , Supervivencia de Injerto , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Regeneración , Daño por Reperfusión/patología , Enfermedades de la Retina/patología , Transcriptoma
20.
Stem Cells Int ; 2013: 983059, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023546

RESUMEN

Mesenchymal stem/stromal cells (MSCs) and MSC-like multipotent stem/progenitor cells have been widely investigated for regenerative medicine and deemed promising in clinical applications. In order to further improve MSC-based stem cell therapeutics, it is important to understand the cellular kinetics and functional roles of MSCs in the dynamic regenerative processes. However, due to the heterogeneous nature of typical MSC cultures, their native identity and anatomical localization in the body have remained unclear, making it difficult to decipher the existence of distinct cell subsets within the MSC entity. Recent studies have shown that several blood-vessel-derived precursor cell populations, purified by flow cytometry from multiple human organs, give rise to bona fide MSCs, suggesting that the vasculature serves as a systemic reservoir of MSC-like stem/progenitor cells. Using individually purified MSC-like precursor cell subsets, we and other researchers have been able to investigate the differential phenotypes and regenerative capacities of these contributing cellular constituents in the MSC pool. In this review, we will discuss the identification and characterization of perivascular MSC precursors, including pericytes and adventitial cells, and focus on their cellular kinetics: cell adhesion, migration, engraftment, homing, and intercellular cross-talk during tissue repair and regeneration.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...