Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Front Neurosci ; 15: 636969, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33994920

RESUMEN

Retinitis pigmentosa (RP) is a rare, progressive disease that affects photoreceptors and retinal pigment epithelial (RPE) cells with blindness as a final outcome. Despite high medical and social impact, there is currently no therapeutic options to slow down the progression of or cure the disease. The development of effective therapies was largely hindered by high genetic heterogeneity, inaccessible disease tissue, and unfaithful model organisms. The fact that components of ubiquitously expressed splicing factors lead to the retina-specific disease is an additional intriguing question. Herein, we sought to correlate the retinal cell-type-specific disease phenotype with the splicing profile shown by a patient with autosomal recessive RP, caused by a mutation in pre-mRNA splicing factor 8 (PRPF8). In order to get insight into the role of PRPF8 in homeostasis and disease, we capitalize on the ability to generate patient-specific RPE cells and reveal differentially expressed genes unique to RPE cells. We found that spliceosomal complex and ribosomal functions are crucial in determining cell-type specificity through differential expression and alternative splicing (AS) and that PRPF8 mutation causes global changes in splice site selection and exon inclusion that particularly affect genes involved in these cellular functions. This finding corroborates the hypothesis that retinal tissue identity is conferred by a specific splicing program and identifies retinal AS events as a framework toward the design of novel therapeutic opportunities.

3.
Stem Cell Reports ; 16(3): 641-655, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33606988

RESUMEN

Increasing brown adipose tissue (BAT) mass and activation is a therapeutic strategy to treat obesity and complications. Obese and diabetic patients possess low amounts of BAT, so an efficient way to expand their mass is necessary. There is limited knowledge about how human BAT develops, differentiates, and is optimally activated. Accessing human BAT is challenging, given its low volume and anatomical dispersion. These constraints make detailed BAT-related developmental and functional mechanistic studies in humans virtually impossible. We have developed and characterized functionally and molecularly a new chemically defined protocol for the differentiation of human pluripotent stem cells (hPSCs) into brown adipocytes (BAs) that overcomes current limitations. This protocol recapitulates step by step the physiological developmental path of human BAT. The BAs obtained express BA and thermogenic markers, are insulin sensitive, and responsive to ß-adrenergic stimuli. This new protocol is scalable, enabling the study of human BAs at early stages of development.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis , Tejido Adiposo Pardo/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Reproducibilidad de los Resultados
4.
Stem Cell Res ; 48: 101956, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32905997

RESUMEN

The Spanish National Stem Cell Bank (Banco Nacional de Líneas Celulares, BNLC) was established in 2006 thanks to a change in the legislative framework in Spain. The Law 14/2006 updated the previous Assisted Reproduction Techniques Law (Law 45/2003) allowing the use of the surplus frozen embryos following IVF for research. The BNLC has a network structure with 3 nodes: the Regenerative Medicine Program (IDIBELL), the Principe Felipe Research Center (CIPF) in Valencia and the Andalusian Public Health System Biobank (SSPA Biobank) in Granada. The aim of the BNLC is to guarantee throughout the national territory the availability of human stem cell lines for biomedical research. At present time, there are 40 human embryonic stem cell lines (hESC) and 171 human induced pluripotent stem cell lines (hiPSC) registered in the BNLC. These lines are fully characterized and available in the context of research projects approved by the Technical Committee of the BNLC.


Asunto(s)
Regulación Gubernamental , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Diferenciación Celular , Línea Celular , Células Madre Embrionarias , Humanos , España , Bancos de Tejidos
5.
Sci Rep ; 10(1): 5426, 2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32214115

RESUMEN

Aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) is a photoreceptor-specific chaperone that stabilizes the effector enzyme of phototransduction, cGMP phosphodiesterase 6 (PDE6). Mutations in the AIPL1 gene cause a severe inherited retinal dystrophy, Leber congenital amaurosis type 4 (LCA4), that manifests as the loss of vision during the first year of life. In this study, we generated three-dimensional (3D) retinal organoids (ROs) from human induced pluripotent stem cells (hiPSCs) derived from an LCA4 patient carrying a Cys89Arg mutation in AIPL1. This study aimed to (i) explore whether the patient hiPSC-derived ROs recapitulate LCA4 disease phenotype, and (ii) generate a clinically relevant resource to investigate the molecular mechanism of disease and safely test novel therapies for LCA4 in vitro. We demonstrate reduced levels of the mutant AIPL1 and PDE6 proteins in patient organoids, corroborating the findings in animal models; however, patient-derived organoids maintained retinal cell cytoarchitecture despite significantly reduced levels of AIPL1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Ojo/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Organoides/metabolismo , Retina/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas del Ojo/genética , Humanos , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Mutación/genética
6.
Mol Vis ; 25: 663-678, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31814692

RESUMEN

PURPOSE: Retinal organoids generated from human pluripotent stem cells exhibit considerable variability during differentiation. Our goals are to assess developmental maturity of the neural retina in vitro and design improved protocols based on objective criteria. METHODS: We performed transcriptome analyses of developing retinal organoids from human embryonic and induced pluripotent stem cell lines and utilized multiple bioinformatic tools for comparative analysis. Immunohistochemistry, immunoblotting and electron microscopy were employed for validation. RESULTS: We show that the developmental variability in organoids was reflected in gene expression profiles and could be evaluated by molecular staging with the human fetal and adult retinal transcriptome data. We also demonstrate that the addition of 9-cis retinal, instead of the widely used all-trans retinoic acid, accelerated rod photoreceptor differentiation in organoid cultures, with higher rhodopsin expression and more mature mitochondrial morphology evident by day 120. CONCLUSION: Our studies provide an objective transcriptome-based modality for determining the differentiation state of retinal organoids and for comparisons across different stem cell lines and platforms, which should facilitate disease modeling and evaluation of therapies in vitro.


Asunto(s)
Diferenciación Celular , Diterpenos/farmacología , Células Madre Embrionarias Humanas/citología , Organoides/citología , Retina/citología , Células Fotorreceptoras Retinianas Bastones/citología , Retinaldehído/farmacología , Transcriptoma/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular , Forma de la Célula/efectos de los fármacos , Perfilación de la Expresión Génica , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Organoides/efectos de los fármacos , Organoides/ultraestructura , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Transcriptoma/efectos de los fármacos
7.
Stem Cell Res ; 38: 101455, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31082679

RESUMEN

Dermal fibroblasts from an autosomal recessive retinitis pigmentosa (RP) patient, homozygous for the mutation c.769 C>T, p.Arg257Ter, in CERKL (Ceramide Kinase-Like) gene, and a healthy sibling were derived and reprogrammed by Sendai virus. The generated human induced pluripotent stem cell (hiPSC) lines RP3-FiPS4F1 and Ctrl3-FiPS4F1, were free of genomically integrated reprogramming genes, showed stable karyotypes, expressed pluripotency markers and could be differentiated towards the three germ layers in vitro. These hiPSC lines offer a useful resource to study RP pathomechanisms, drug testing and therapeutic opportunities.


Asunto(s)
Homocigoto , Células Madre Pluripotentes Inducidas , Mutación Missense , Fosfotransferasas (Aceptor de Grupo Alcohol) , Retinitis Pigmentosa , Hermanos , Sustitución de Aminoácidos , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
8.
Stem Cell Res ; 34: 101341, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30612079

RESUMEN

The human induced pluripotent stem cell (hiPSC) line RP1-FiPS4F1 generated from the patient with autosomal recessive retinitis pigmentosa (arRP) caused by homozygous Ser331Cysfs*5 mutation in Mer tyrosine kinase receptor (MERTK) was genetically corrected using CRISPR/Cas9 system. Two isogenic hiPSCs lines, with heterozygous and homozygous correction of c.992_993delCA mutation in the MERTK gene were generated. These cell lines demonstrate normal karyotype, maintain a pluripotent state, and can differentiate toward three germ layers in vitro. These genetically corrected hiPSCs represent accurate controls to study the contribution of the specific genetic change to the disease, and potentially therapeutic material for cell-replacement therapy.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes Inducidas/patología , Mutación/genética , Retinitis Pigmentosa/patología , Reparación del Gen Blanco , Tirosina Quinasa c-Mer/genética , Secuencia de Bases , Línea Celular , Humanos
9.
Stem Cell Res ; 33: 151-155, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30366342

RESUMEN

The human induced pluripotent stem cell (hiPSC) line, derived from dermal fibroblasts from Leber congenital amaurosis patient with homozygous mutation c.265 T > C, p.Cys89Arg in aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) was generated by Sendai virus reprogramming. The generated hiPSC line was free of Sendai virus genes, had stable karyotype, carried the homozygous mutation, was immunopositive to pluripotency markers and able to generate all three germ layers upon embryoid body formation. Resource table.


Asunto(s)
Proteínas Portadoras/genética , Proteínas del Ojo/genética , Células Madre Pluripotentes Inducidas/metabolismo , Amaurosis Congénita de Leber/genética , Proteínas Adaptadoras Transductoras de Señales , Adulto , Femenino , Humanos , Mutación
13.
Stem Cells ; 36(4): 474-481, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29345014

RESUMEN

Hereditary retinal dystrophies, specifically retinitis pigmentosa (RP) are clinically and genetically heterogeneous diseases affecting primarily retinal cells and retinal pigment epithelial cells with blindness as a final outcome. Understanding the pathogenicity behind these diseases has been largely precluded by the unavailability of affected tissue from patients, large genetic heterogeneity and animal models that do not faithfully represent some human diseases. A landmark discovery of human induced pluripotent stem cells (hiPSCs) permitted the derivation of patient-specific cells. These cells have unlimited self-renewing capacity and the ability to differentiate into RP-affected cell types, allowing the studies of disease mechanism, drug discovery, and cell replacement therapies, both as individual cell types and organoid cultures. Together with precise genome editing, the patient specific hiPSC technology offers novel strategies for targeting the pathogenic mutations and design therapies toward retinal dystrophies. This study summarizes current hiPSC-based RP models and highlights key achievements and challenges of these cellular models, as well as questions that still remain unanswered. Stem Cells 2018;36:474-481.


Asunto(s)
Diferenciación Celular , Edición Génica , Genoma Humano , Células Madre Pluripotentes Inducidas/metabolismo , Retinitis Pigmentosa , Trasplante de Células Madre , Animales , Autoinjertos , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/terapia
15.
Neuroscientist ; 23(5): 554-566, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28281409

RESUMEN

Cerebellar ataxias are clinically and genetically heterogeneous diseases affecting primary cerebellar cells. The lack of availability of affected tissue from cerebellar ataxias patients is the main obstacle in investigating the pathogenicity of these diseases. The landmark discovery of human-induced pluripotent stem cells (hiPSC) has permitted the derivation of patient-specific cells with an unlimited self-renewing capacity. Additionally, their potential to differentiate into virtually any cell type of the human organism allows for large amounts of affected cells to be generated in culture, converting this hiPSC technology into a revolutionary tool in the study of the mechanisms of disease, drug discovery, and gene correction. In this review, we will summarize the current studies in which hiPSC were utilized to study cerebellar ataxias. Describing the currently available 2D and 3D hiPSC-based cellular models, and due to the fact that extracerebellar cells were used to model these diseases, we will discuss whether or not they represent a faithful cellular model and whether they have contributed to a better understanding of disease mechanisms.


Asunto(s)
Diferenciación Celular/fisiología , Ataxia Cerebelosa/cirugía , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Animales , Humanos , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología
16.
Stem Cells Transl Med ; 6(4): 1217-1226, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28213969

RESUMEN

Neural differentiation of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) can produce a valuable and robust source of human neural cell subtypes, holding great promise for the study of neurogenesis and development, and for treating neurological diseases. However, current hESCs and hiPSCs neural differentiation protocols require either animal factors or embryoid body formation, which decreases efficiency and yield, and strongly limits medical applications. Here we develop a simple, animal-free protocol for neural conversion of both hESCs and hiPSCs in adherent culture conditions. A simple medium formula including insulin induces the direct conversion of >98% of hESCs and hiPSCs into expandable, transplantable, and functional neural progenitors with neural rosette characteristics. Further differentiation of neural progenitors into dopaminergic and spinal motoneurons as well as astrocytes and oligodendrocytes indicates that these neural progenitors retain responsiveness to instructive cues revealing the robust applicability of the protocol in the treatment of different neurodegenerative diseases. The fact that this protocol includes animal-free medium and human extracellular matrix components avoiding embryoid bodies makes this protocol suitable for the use in clinic. Stem Cells Translational Medicine 2017;6:1217-1226.


Asunto(s)
Células Madre Pluripotentes/citología , Diferenciación Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Células Madre Embrionarias/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología
17.
Sci Rep ; 5: 12910, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26263531

RESUMEN

Retinitis pigmentosa (RP) represents a genetically heterogeneous group of retinal dystrophies affecting mainly the rod photoreceptors and in some instances also the retinal pigment epithelium (RPE) cells of the retina. Clinical symptoms and disease progression leading to moderate to severe loss of vision are well established and despite significant progress in the identification of causative genes, the disease pathology remains unclear. Lack of this understanding has so far hindered development of effective therapies. Here we report successful generation of human induced pluripotent stem cells (iPSC) from skin fibroblasts of a patient harboring a novel Ser331Cysfs*5 mutation in the MERTK gene. The patient was diagnosed with an early onset and severe form of autosomal recessive RP (arRP). Upon differentiation of these iPSC towards RPE, patient-specific RPE cells exhibited defective phagocytosis, a characteristic phenotype of MERTK deficiency observed in human patients and animal models. Thus we have created a faithful cellular model of arRP incorporating the human genetic background which will allow us to investigate in detail the disease mechanism, explore screening of a variety of therapeutic compounds/reagents and design either combined cell and gene- based therapies or independent approaches.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Modelos Biológicos , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Retinitis Pigmentosa/patología , Animales , Estudios de Casos y Controles , Diferenciación Celular , Humanos , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Retinitis Pigmentosa/genética , Tirosina Quinasa c-Mer
18.
Sci Rep ; 5: 9640, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25860664

RESUMEN

Spinal cord injury (SCI) results in neural loss and consequently motor and sensory impairment below the injury. There are currently no effective therapies for the treatment of traumatic SCI in humans. Various animal models have been developed to mimic human SCI. Widely used animal models of SCI are complete or partial transection or experimental contusion and compression, with both bearing controversy as to which one more appropriately reproduces the human SCI functional consequences. Here we present in details the widely used procedure of complete spinal cord transection as a faithful animal model to investigate neural and functional repair of the damaged tissue by exogenous human transplanted cells. This injury model offers the advantage of complete damage to a spinal cord at a defined place and time, is relatively simple to standardize and is highly reproducible.


Asunto(s)
Trasplante de Células , Modelos Animales de Enfermedad , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Investigación Biomédica Traslacional , Animales , Células Madre Embrionarias , Femenino , Humanos , Ratas , Traumatismos de la Médula Espinal/terapia
19.
Stem Cells ; 33(4): 1036-41, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25728093

RESUMEN

Spinal cord injury (SCI) usually results in long lasting locomotor and sensory neuron degeneration below the injury. Astrocytes normally play a decisive role in mechanical and metabolic support of neurons, but in the spinal cord they cause injury, exerting well-known detrimental effects that contribute to glial scar formation and inhibition of axon outgrowth. Cell transplantation is considered a promising approach for replacing damaged cells and promoting neuroprotective and neuroregenerative repair, but the effects of the grafted cells on local tissue and the regenerative properties of endogenous neural stem cells in the injured spinal cord are largely unknown. During the last 2 decades cumulative evidence from diverse animal models has indicated that reactive astrocytes in synergy with transplanted cells could be beneficial for injury in multiple ways, including neuroprotection and axonal growth. In this review, we specifically focus on the dual opposing roles of reactive astrocytes in SCI and how they contribute to the creation of a permissive environment when combined with transplanted cells as the influential components for a local regenerative niche. Modulation of reactive astrocyte function might represent an extremely attractive new therapy to enhance the functional outcomes in patients.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/trasplante , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos , Animales , Humanos , Regeneración Nerviosa/fisiología , Células Madre/metabolismo
20.
Front Genet ; 5: 132, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24860598

RESUMEN

Several studies have demonstrated the important role of non-coding RNAs as regulators of posttranscriptional processes, including stem cells self-renewal and neural differentiation. Human embryonic stem cells (hESCs) and induced pluripotent stem cells (ihPSCs) show enormous potential in regenerative medicine due to their capacity to differentiate to virtually any type of cells of human body. Deciphering the role of non-coding RNAs in pluripotency, self-renewal and neural differentiation will reveal new molecular mechanisms involved in induction and maintenances of pluripotent state as well as triggering these cells toward clinically relevant cells for transplantation. In this brief review we will summarize recently published studies which reveal the role of non-coding RNAs in pluripotency and neural differentiation of hESCs and ihPSC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA