Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 257
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Med Mol Morphol ; 55(3): 174-186, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35461467

RESUMEN

Cleidocranial dysplasia (CCD) is a hereditary disorder associated with skeletal dysplasia and dental abnormalities. CCD arises from heterozygous loss of function mutations in the Runt-related transcription factor 2 (RUNX2) gene. Osteoporosis is often observed in CCD patients and conventional vitamin D supplementation is recommended. However, sufficient evidences have not been presented yet. This study investigated the role of RUNX2 in osteoblastic differentiation and sought to identify potential target genes for the treatment of osteoporosis associated with CCD, using induced pluripotent stem cell (iPSC) technology. We successfully established Runx2-/-, Runx2+/- and wild-type miPSCs from litter-matched mice and found poor Vdr expression in Runx2-/-cells. Significant down-regulation of osteoblastic differentiation in Runx2-/- miPSCs was observed. Gene expression array revealed unexpected results such as remarkable increase of Rankl expression and decrease of Vdr in Runx2-/- cells. Insufficient response to vitamin D in Runx2-/- cells was also observed. Our results suggest that RUNX2 functions as a regulator of Rankl and Vdr and thereby controls bone density. These findings also suggest that conventional vitamin D supplementation may not be as effective as previously expected, in the treatment of osteoporosis associated with CCD, and that inhibiting RANKL function might be worth considering as an alternative treatment strategy.


Asunto(s)
Displasia Cleidocraneal , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Células Madre Pluripotentes Inducidas , Osteoporosis , Vitamina D , Animales , Diferenciación Celular , Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Osteoporosis/genética , Vitamina D/farmacología
2.
Stem Cell Reports ; 16(12): 3050-3063, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34861166

RESUMEN

Hepatic stellate cells (HSCs) play a central role in the progression of liver fibrosis by producing extracellular matrices. The development of drugs to suppress liver fibrosis has been hampered by the lack of human quiescent HSCs (qHSCs) and an appropriate in vitro model that faithfully recapitulates HSC activation. In the present study, we developed a culture system to generate qHSC-like cells from human-induced pluripotent stem cells (hiPSCs) that can be converted into activated HSCs in culture. To monitor the activation process, a red fluorescent protein (RFP) gene was inserted in hiPSCs downstream of the activation marker gene actin alpha 2 smooth muscle (ACTA2). Using qHSC-like cells derived from RFP reporter iPSCs, we screened a repurposing chemical library and identified therapeutic candidates that prevent liver fibrosis. Hence, hiPSC-derived qHSC-like cells will be a useful tool to study the mechanism of HSC activation and to identify therapeutic agents.


Asunto(s)
Técnicas de Cultivo de Célula , Ciclo Celular , Descubrimiento de Drogas , Células Estrelladas Hepáticas/citología , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Animales , Evaluación Preclínica de Medicamentos , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL
3.
Front Immunol ; 12: 748519, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777360

RESUMEN

Inherited defects that abrogate the function of the adenosine deaminase (ADA) enzyme and consequently lead to the accumulation of toxic purine metabolites cause profound lymphopenia and severe combined immune deficiency. Additionally, neutropenia and impaired neutrophil function have been reported among ADA-deficient patients. However, due to the rarity of the disorder, the neutrophil developmental abnormalities and the mechanisms contributing to them have not been characterized. Induced pluripotent stem cells (iPSC) generated from two unrelated ADA-deficient patients and from healthy controls were differentiated through embryoid bodies into neutrophils. ADA deficiency led to a significant reduction in the number of all early multipotent hematopoietic progenitors. At later stages of differentiation, ADA deficiency impeded the formation of granulocyte colonies in methylcellulose cultures, leading to a significant decrease in the number of neutrophils generated from ADA-deficient iPSCs. The viability and apoptosis of ADA-deficient neutrophils isolated from methylcellulose cultures were unaffected, suggesting that the abnormal purine homeostasis in this condition interferes with differentiation or proliferation. Additionally, there was a significant increase in the percentage of hyperlobular ADA-deficient neutrophils, and these neutrophils demonstrated significantly reduced ability to phagocytize fluorescent microspheres. Supplementing iPSCs and methylcellulose cultures with exogenous ADA, which can correct adenosine metabolism, reversed all abnormalities, cementing the critical role of ADA in neutrophil development. Moreover, chemical inhibition of the ribonucleotide reductase (RNR) enzyme, using hydroxyurea or a combination of nicotinamide and trichostatin A in iPSCs from healthy controls, led to abnormal neutrophil differentiation similar to that observed in ADA deficiency, implicating RNR inhibition as a potential mechanism for the neutrophil abnormalities. In conclusion, the findings presented here demonstrate the important role of ADA in the development and function of neutrophils while clarifying the mechanisms responsible for the neutrophil abnormalities in ADA-deficient patients.


Asunto(s)
Adenosina Desaminasa/fisiología , Agammaglobulinemia/inmunología , Células Madre Pluripotentes Inducidas/citología , Neutrófilos/citología , Inmunodeficiencia Combinada Grave/inmunología , Adenosina Desaminasa/genética , Células Cultivadas , Cuerpos Embrioides/citología , Fibroblastos/enzimología , Granulocitos/citología , Humanos , Ácidos Hidroxámicos/farmacología , Hidroxiurea/farmacología , Lactante , Masculino , Mutación Missense , Mielopoyesis , Niacinamida/farmacología , Mutación Puntual , Ribonucleótido Reductasas/antagonistas & inhibidores
4.
Cells ; 10(7)2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34359837

RESUMEN

Induced pluripotent stem (iPS) cells constitute a perfect tool to study human embryo development processes such as myogenesis, thanks to their ability to differentiate into three germ layers. Currently, many protocols to obtain myogenic cells have been described in the literature. They differ in many aspects, such as media components, including signaling modulators, feeder layer constituents, and duration of culture. In our study, we compared three different myogenic differentiation protocols to verify, side by side, their efficiency. Protocol I was based on embryonic bodies differentiation induction, ITS addition, and selection with adhesion to collagen I type. Protocol II was based on strong myogenic induction at the embryonic bodies step with BIO, forskolin, and bFGF, whereas cells in Protocol III were cultured in monolayers in three special media, leading to WNT activation and TGF-ß and BMP signaling inhibition. Myogenic induction was confirmed by the hierarchical expression of myogenic regulatory factors MYF5, MYOD, MYF6 and MYOG, as well as the expression of myotubes markers MYH3 and MYH2, in each protocol. Our results revealed that Protocol III is the most efficient in obtaining myogenic cells. Furthermore, our results indicated that CD56 is not a specific marker for the evaluation of myogenic differentiation.


Asunto(s)
Técnicas de Cultivo de Célula , Medios de Cultivo/farmacología , Cuerpos Embrioides/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Colforsina/farmacología , Colágeno Tipo I/farmacología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica , Humanos , Indoles/farmacología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Insulina/farmacología , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Proteína MioD/genética , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Oximas/farmacología , Selenio/farmacología , Transferrina/farmacología
5.
Int J Mol Sci ; 22(16)2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34445332

RESUMEN

More than 85% of pre-clinically tested drugs fail during clinical trials, which results in a long, inefficient and costly process, suggesting that animal models are often poor predictors of human biology [...].


Asunto(s)
Descubrimiento de Drogas/métodos , Células Madre Pluripotentes Inducidas/fisiología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Descubrimiento de Drogas/tendencias , Evaluación Preclínica de Medicamentos/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos
6.
J Mol Cell Cardiol ; 160: 97-110, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34216608

RESUMEN

Angiotensin II (Ang II) presents a critical mediator in various pathological conditions such as non-genetic cardiomyopathy. Osmotic pump infusion in rodents is a commonly used approach to model cardiomyopathy associated with Ang II. However, profound differences in electrophysiology and pharmacokinetics between rodent and human cardiomyocytes may limit predictability of animal-based experiments. This study investigates the application of an Organ-on-a-chip (OOC) system in modeling Ang II-induced progressive cardiomyopathy. The disease model is constructed to recapitulate myocardial response to Ang II in a temporal manner. The long-term tissue cultivation and non-invasive functional readouts enable monitoring of both acute and chronic cardiac responses to Ang II stimulation. Along with mapping of cytokine secretion and proteomic profiles, this model presents an opportunity to quantitatively measure the dynamic pathological changes that could not be otherwise identified in animals. Further, we present this model as a testbed to evaluate compounds that target Ang II-induced cardiac remodeling. Through assessing the effects of losartan, relaxin, and saracatinib, the drug screening data implicated multifaceted cardioprotective effects of relaxin in restoring contractile function and reducing fibrotic remodeling. Overall, this study provides a controllable platform where cardiac activities can be explicitly observed and tested over the pathological process. The facile and high-content screening can facilitate the evaluation of potential drug candidates in the pre-clinical stage.


Asunto(s)
Angiotensina II/efectos adversos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Animales , Cardiomiopatías/patología , Cardiotónicos/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Evaluación Preclínica de Medicamentos/métodos , Fibroblastos/metabolismo , Fibrosis , Humanos , Células Madre Pluripotentes Inducidas/citología , Dispositivos Laboratorio en un Chip , Losartán/farmacología , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proyectos Piloto , Proteoma , Proteómica/métodos , Proteínas Recombinantes/farmacología , Relaxina/farmacología , Remodelación Ventricular/efectos de los fármacos
7.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800815

RESUMEN

In the last decade, different research groups in the academic setting have developed induced pluripotent stem cell-based protocols to generate three-dimensional, multicellular, neural organoids. Their use to model brain biology, early neural development, and human diseases has provided new insights into the pathophysiology of neuropsychiatric and neurological disorders, including microcephaly, autism, Parkinson's disease, and Alzheimer's disease. However, the adoption of organoid technology for large-scale drug screening in the industry has been hampered by challenges with reproducibility, scalability, and translatability to human disease. Potential technical solutions to expand their use in drug discovery pipelines include Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) to create isogenic models, single-cell RNA sequencing to characterize the model at a cellular level, and machine learning to analyze complex data sets. In addition, high-content imaging, automated liquid handling, and standardized assays represent other valuable tools toward this goal. Though several open issues still hamper the full implementation of the organoid technology outside academia, rapid progress in this field will help to prompt its translation toward large-scale drug screening for neurological disorders.


Asunto(s)
Descubrimiento de Drogas/métodos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Organoides/efectos de los fármacos , Animales , Automatización , Encéfalo/citología , Sistemas CRISPR-Cas , Técnicas de Cultivo de Célula , Colágeno , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/organización & administración , Predicción , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Madre Pluripotentes Inducidas/citología , Laminina , Aprendizaje Automático , Microscopía/métodos , Enfermedades del Sistema Nervioso/patología , Proteoglicanos , RNA-Seq , Reproducibilidad de los Resultados , Análisis de la Célula Individual
8.
J Steroid Biochem Mol Biol ; 211: 105881, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33766737

RESUMEN

Human hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) represent a promising cell source for the assessment of hepatotoxicity and pharmaceutical safety testing. However, the hepatic functionality of HLCs remains significantly inferior to primary human hepatocytes. The bioactive vitamin D (VD), calcitriol, promotes the differentiation of many types of cells, and its deficiency is correlated to the severity of liver diseases. Whether calcitriol contributes to the differentiation of HLCs needs to be explored. Here, we found that the supplementation of calcitriol improved the functionalities of hPSCs-derived HLCs in P450 activities, urea production, and albumin secretion. Moreover, calcitriol also enhanced mitochondrial respiratory function with increased protein expression levels of the subunit of respiratory enzyme complexes in HLCs. Further analyses showed that the mitochondrial biogenesis regulators and mitophagy were increased by calcitriol, thus improving the mitochondrial quality. These improvements in functionality and mitochondrial condition were dependent on vitamin D receptor (VDR) because the improvements were abolished under VDR-deficient conditions. Our finding provides a cost-effective chemical process for HLC maturation to meet the demand for basic research and potential clinic applications.


Asunto(s)
Calcitriol/farmacología , Diferenciación Celular , Hepatocitos/citología , Células Madre Pluripotentes Inducidas/citología , Mitocondrias/efectos de los fármacos , Mitofagia , Biogénesis de Organelos , Células Cultivadas , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Mitocondrias/metabolismo , Vitaminas/farmacología
9.
Sci Rep ; 11(1): 5654, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707655

RESUMEN

We hypothesized that an appropriate ratio of cardiomyocytes, fibroblasts, endothelial cells, and extracellular matrix (ECM) factors would be required for the development of three-dimensional cardiac tissues (3D-CTs) as drug screening systems. To verify this hypothesis, ECM-coated human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), ECM-coated cardiac fibroblasts (CFs), and uncoated cardiac endothelial cells (CEs) were mixed in the following ratios: 10:0:0 (10CT), 7:2:1 (7CT), 5:4:1 (5CT), and 2:7:1 (2CT). The expression of cardiac-, fibroblasts-, and endothelial-specific markers was assessed by FACS, qPCR, and immunostaining while that of ECM-, cell adhesion-, and ion channel-related genes was examined by qPCR. Finally, the contractile properties of the tissues were evaluated in the absence or presence of E-4031 and isoproterenol. The expression of ECM- and adhesion-related genes significantly increased, while that of ion channel-related genes significantly decreased with the CF proportion. Notably, 7CT showed the greatest contractility of all 3D-CTs. When exposed to E-4031 (hERG K channel blocker), 7CT and 5CT showed significantly decreased contractility and increased QT prolongation. Moreover, 10CT and 7CT exhibited a stronger response to isoproterenol than did the other 3D-CTs. Finally, 7CT showed the highest drug sensitivity among all 3D-CTs. In conclusion, 3D-CTs with an appropriate amount of fibroblasts/endothelial cells (7CT in this study) are suitable drug screening systems, e.g. for the detection of drug-induced arrhythmia.


Asunto(s)
Evaluación Preclínica de Medicamentos , Corazón/diagnóstico por imagen , Imagenología Tridimensional , Animales , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Isoproterenol/farmacología , Ratones , Contracción Miocárdica/fisiología , Piperidinas/farmacología , Piridinas/farmacología
10.
Sci Rep ; 11(1): 5437, 2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33686099

RESUMEN

Examining intestine-liver interactions is important for achieving the desired physiological drug absorption and metabolism response in in vitro drug tests. Multi-organ microphysiological systems (MPSs) constitute promising tools for evaluating inter-organ interactions in vitro. For coculture on MPSs, normal cells are challenging to use because they require complex maintenance and careful handling. Herein, we demonstrated the potential of coculturing normal cells on MPSs in the evaluation of intestine-liver interactions. To this end, we cocultured human-induced pluripotent stem cell-derived intestinal cells and fresh human hepatocytes which were isolated from PXB mice with medium circulation in a pneumatic-pressure-driven MPS with pipette-friendly liquid-handling options. The cytochrome activity, albumin production, and liver-specific gene expressions in human hepatocytes freshly isolated from a PXB mouse were significantly upregulated via coculture with hiPS-intestinal cells. Our normal cell coculture shows the effects of the interactions between the intestine and liver that may occur in vivo. This study is the first to demonstrate the coculturing of hiPS-intestinal cells and fresh human hepatocytes on an MPS for examining pure inter-organ interactions. Normal-cell coculture using the multi-organ MPS could be pursued to explore unknown physiological mechanisms of inter-organ interactions in vitro and investigate the physiological response of new drugs.


Asunto(s)
Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas , Animales , Técnicas de Cocultivo , Evaluación Preclínica de Medicamentos , Hepatocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Presión
11.
Cell Calcium ; 94: 102362, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33540322

RESUMEN

Progressive neuronal loss is a hallmark of many neurodegenerative diseases, including Alzheimer's and Parkinson's disease. These pathologies exhibit clear signs of inflammation, mitochondrial dysfunction, calcium deregulation, and accumulation of aggregated or misfolded proteins. Over the last decades, a tremendous research effort has contributed to define some of the pathological mechanisms underlying neurodegenerative processes in these complex brain neurodegenerative disorders. To better understand molecular mechanisms responsible for neurodegenerative processes and find potential interventions and pharmacological treatments, it is important to have robust in vitro and pre-clinical animal models that can recapitulate both the early biological events undermining the maintenance of the nervous system and early pathological events. In this regard, it would be informative to determine how different inherited pathogenic mutations can compromise mitochondrial function, calcium signaling, and neuronal survival. Since post-mortem analyses cannot provide relevant information about the disease progression, it is crucial to develop model systems that enable the investigation of early molecular changes, which may be relevant as targets for novel therapeutic options. Thus, the use of human induced pluripotent stem cells (iPSCs) represents an exceptional complementary tool for the investigation of degenerative processes. In this review, we will focus on two neurodegenerative diseases, Alzheimer's and Parkinson's disease. We will provide examples of iPSC-derived neuronal models and how they have been used to study calcium and mitochondrial alterations during neurodegeneration.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Mitocondrias/patología , Modelos Biológicos , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Animales , Señalización del Calcio , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
12.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530458

RESUMEN

A high-throughput drug screen identifies potentially promising therapeutics for clinical trials. However, limitations that persist in current disease modeling with limited physiological relevancy of human patients skew drug responses, hamper translation of clinical efficacy, and contribute to high clinical attritions. The emergence of induced pluripotent stem cell (iPSC) technology revolutionizes the paradigm of drug discovery. In particular, iPSC-based three-dimensional (3D) tissue engineering that appears as a promising vehicle of in vitro disease modeling provides more sophisticated tissue architectures and micro-environmental cues than a traditional two-dimensional (2D) culture. Here we discuss 3D based organoids/spheroids that construct the advanced modeling with evolved structural complexity, which propels drug discovery by exhibiting more human specific and diverse pathologies that are not perceived in 2D or animal models. We will then focus on various central nerve system (CNS) disease modeling using human iPSCs, leading to uncovering disease pathogenesis that guides the development of therapeutic strategies. Finally, we will address new opportunities of iPSC-assisted drug discovery with multi-disciplinary approaches from bioengineering to Omics technology. Despite technological challenges, iPSC-derived cytoarchitectures through interactions of diverse cell types mimic patients' CNS and serve as a platform for therapeutic development and personalized precision medicine.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ingeniería de Tejidos/métodos , Animales , COVID-19/patología , Enfermedades del Sistema Nervioso Central/patología , Descubrimiento de Drogas/instrumentación , Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Humanos , Células Madre Pluripotentes Inducidas/patología , Dispositivos Laboratorio en un Chip , Organoides/citología , Organoides/efectos de los fármacos , Organoides/patología , Ingeniería de Tejidos/instrumentación , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/patología , Tratamiento Farmacológico de COVID-19
14.
Biochem Biophys Res Commun ; 558: 231-238, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32113685

RESUMEN

Several groups have developed in vitro expansion cultures for mouse metanephric nephron progenitor cells (NPCs) using cocktails of small molecules and growth factors including BMP7. However, the detailed mechanisms by which BMP7 acts in the NPC expansion remain to be elucidated. Here, by performing chemical screening for BMP substitutes, we identified a small molecule, TCS21311, that can replace BMP7 and revealed a novel inhibitory role of BMP7 in JAK3-STAT3 signaling in NPC expansion culture. Further, we found that TCS21311 facilitates the proliferation of mouse embryonic NPCs and human induced pluripotent stem cell-derived NPCs when added to the expansion culture. These results will contribute to understanding the mechanisms of action of BMP7 in NPC proliferation in vitro and in vivo and to the stable supply of NPCs for regenerative therapy, disease modeling and drug discovery for kidney diseases.


Asunto(s)
Proteína Morfogenética Ósea 7/metabolismo , Inhibidores de las Cinasas Janus/farmacología , Nefronas/citología , Nefronas/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 7/administración & dosificación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Medios de Cultivo , Evaluación Preclínica de Medicamentos , Humanos , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Janus Quinasa 3/antagonistas & inhibidores , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Nefronas/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas
15.
Curr Eye Res ; 46(4): 504-514, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32777180

RESUMEN

PURPOSE: Maintaining mature and viable retinal pigment epithelial cells (RPE) in vitro has proven challenging. Investigating compounds that can promote RPE-viability and maturation is motivated by RPE transplantation research, the quest to understand RPE physiology, and a desire to modulate RPE in pathological states. We have previously reported that the silk protein sericin promotes viability, maturation, and pigmentation of human fetal RPE. In the present study, our aim was to uncover whether these effects can be seen in adult retinal pigment epithelial cell line-19 (ARPE-19) and induced pluripotent stem cell-derived RPE (iPSC-RPE). METHODS: ARPE-19 and iPSC-RPE were cultured with or without 10 mg/mL sericin. After 7 days, viability was assessed with calcein-acetoxymethyl ester (CAM) and ethidium homodimer-1 (EH-1) assays, flow cytometry, and morphometric analysis. Expression levels of RPE65, tyrosinase, and Pmel17 were quantified to compare maturation between the sericin-treated and control cultures. Light microscopy and staining of the tight junction protein zonula occludens protein 1 (ZO-1) were employed to study sericin's effects on RPE morphology. We also measured culture medium pH, glucose, lactate, and extracellular ion content. RESULTS: Sericin-supplemented RPE cultures demonstrated significantly better viability compared to control cultures. Sericin appeared to improve ARPE-19 maturation and morphology in vitro. No effects were seen on RPE pigmentation with the concentration of sericin and duration of cell culture herein reported. CONCLUSIONS: This is the first study to demonstrate that supplementing the culture media with sericin promotes the viability of iPSC-RPE and ARPE-19. Sericin's viability-promoting effects may have important implications for retinal therapeutics and regenerative medicine research.


Asunto(s)
Células Madre Pluripotentes Inducidas/efectos de los fármacos , Epitelio Pigmentado de la Retina/efectos de los fármacos , Sericinas/farmacología , Línea Celular , Supervivencia Celular/fisiología , Células Cultivadas , Citometría de Flujo , Glucosa/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ácido Láctico/metabolismo , Monofenol Monooxigenasa/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , cis-trans-Isomerasas/metabolismo , Antígeno gp100 del Melanoma/metabolismo
16.
Int J Hematol ; 114(5): 572-579, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32705572

RESUMEN

The use of allogeneic, pluripotent stem cell-derived immune cells for cancer immunotherapy has been the subject of recent research, including clinical trials. The use of pluripotent stem cells as the source for allogeneic immune cells facilitates stringent quality control of the final product, regarding efficacy, safety, and producibility. In this review, we have described the characteristics of natural killer (NK) cells from multiple cell sources, including pluripotent stem cells, the chimeric antigen receptor (CAR)-modification method and strategy for these NK cells, and the current and planned clinical trials of CAR-modified induced pluripotent stem cell-derived NK cells.


Asunto(s)
Expresión Génica , Inmunoterapia Adoptiva , Células Madre Pluripotentes Inducidas/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias/terapia , Receptores Quiméricos de Antígenos/genética , Animales , Antígenos de Neoplasias/inmunología , Estudios Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Células Asesinas Naturales/citología , Neoplasias/etiología , Pronóstico , Receptores Quiméricos de Antígenos/inmunología , Resultado del Tratamiento
17.
Methods Mol Biol ; 2180: 539-554, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32797433

RESUMEN

Current research in the field of transfusion medicine is focused on developing innovative approaches to generate populations of functional megakaryocytes (MKs) ex vivo. This may open perspectives to establish alternative therapies for donor platelet transfusion in the management of thrombocytopenic patients and pave the way for novel regenerative approaches. Efficient cryopreservation techniques can provide the opportunity for long-term storage and accumulation of necessary amounts of MKs in a ready-to-use manner. However, in this case, besides the viability, it is crucial to consider the recovery of functional MK properties after the impact of freezing. In this chapter, the possibility to cryopreserve iPSC-derived MKs is described. In particular, the methods for a comprehensive analysis of phenotypic and functional features of MKs after cryopreservation are proposed. The use of cryopreserved in vitro-produced MKs may benefit to the field of transfusion medicine to overcome the lack of sufficient blood donors.


Asunto(s)
Plaquetas/citología , Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Criopreservación/métodos , Crioprotectores/farmacología , Células Madre Pluripotentes Inducidas/citología , Megacariocitos/citología , Animales , Plaquetas/efectos de los fármacos , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Megacariocitos/efectos de los fármacos
18.
Stem Cells Dev ; 30(2): 79-90, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33256572

RESUMEN

Forced coexpression of the transcription factors Oct3/4, Klf4, Sox2, and c-Myc reprograms somatic cells into pluripotent stem cells (PSCs). Such induced PSCs (iPSCs) can generate any cell type of the adult body or indefinitely proliferate without losing their potential. Accordingly, iPSCs can serve as an unlimited cell source for the development of various disease models and regenerative therapies for animals and humans. Although canine peripheral blood mononuclear cells (PBMCs) can be easily obtained, they have a very low iPSC reprogramming efficiency. In this study, we determined the reprogramming efficiency of canine PBMCs under several conditions involving three types of media supplemented with small-molecule compounds. We found that canine iPSCs (ciPSCs) could be efficiently generated from PBMCs using N2B27 medium supplemented with leukemia inhibitory factor (LIF), basic fibroblast growth factor (bFGF), and a small-molecule cocktail (Y-27632, PD0325901, CHIR99021, A-83-01, Forskolin, and l-ascorbic acid). We generated five ciPSC lines that could be maintained in StemFit® medium supplemented with LIF. The SeVdp(KOSM)302L vectors were appropriately silenced in four ciPSC lines. Of the two lines characterized, both were positive for alkaline phosphatase activity and expressed pluripotency markers, including the Oct3/4, Sox2, and Nanog transcripts, as well as the octamer-binding transcription factor (OCT) 3/4 and NANOG proteins, and the SSEA-1 carbohydrate antigen. The ciPSCs could form embryoid bodies and differentiate into the three germ layers, as indicated by marker gene and protein expression. Furthermore, one ciPSC line formed teratomas comprising several tissues from every germ layer. Our ciPSC lines maintained a normal karyotype even after multiple passages. Moreover, our new reprogramming method was able to generate ciPSCs from multiple donor PBMCs. In conclusion, we developed an easy and efficient strategy for the generation of footprint-free ciPSCs from PBMCs. We believe that this strategy can be useful for disease modeling and regenerative medicine in the veterinary field.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Expresión Génica/genética , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares/metabolismo , Animales , Células Cultivadas , Técnicas de Reprogramación Celular/métodos , Medios de Cultivo/química , Medios de Cultivo/farmacología , Perros , Ectodermo/citología , Ectodermo/metabolismo , Endodermo/citología , Endodermo/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Leucocitos Mononucleares/citología , Mesodermo/citología , Mesodermo/metabolismo , Ratones Endogámicos ICR , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Reproducibilidad de los Resultados , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
19.
Stem Cell Reports ; 16(1): 168-181, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33338435

RESUMEN

Burgeoning applications of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in disease modeling, regenerative medicine, and drug screening have broadened the usage of hiPSC-CMs and entailed their long-term storage. Cryopreservation is the most common approach to store hiPSC-CMs. However, the effects of cryopreservation and recovery on hiPSC-CMs remain poorly understood. Here, we characterized the transcriptome, electro-mechanical function, and drug response of fresh hiPSC-CMs without cryopreservation and recovered hiPSC-CMs from cryopreservation. We found that recovered hiPSC-CMs showed upregulation of cell cycle genes, similar or reduced contractility, Ca2+ transients, and field potential duration. When subjected to treatment of drugs that affect electrophysiological properties, recovered hiPSC-CMs showed an altered drug response and enhanced propensity for drug-induced cardiac arrhythmic events. In conclusion, fresh and recovered hiPSC-CMs do not always show comparable molecular and physiological properties. When cryopreserved hiPSC-CMs are used for assessing drug-induced cardiac liabilities, the altered drug sensitivity needs to be considered.


Asunto(s)
Criopreservación , Miocitos Cardíacos/metabolismo , Potenciales de Acción , Antiarrítmicos/farmacología , Calcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Nifedipino/farmacología , Regulación hacia Arriba/efectos de los fármacos
20.
Cell ; 183(7): 1913-1929.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33333020

RESUMEN

Neurons in the cerebral cortex connect through descending pathways to hindbrain and spinal cord to activate muscle and generate movement. Although components of this pathway have been previously generated and studied in vitro, the assembly of this multi-synaptic circuit has not yet been achieved with human cells. Here, we derive organoids resembling the cerebral cortex or the hindbrain/spinal cord and assemble them with human skeletal muscle spheroids to generate 3D cortico-motor assembloids. Using rabies tracing, calcium imaging, and patch-clamp recordings, we show that corticofugal neurons project and connect with spinal spheroids, while spinal-derived motor neurons connect with muscle. Glutamate uncaging or optogenetic stimulation of cortical spheroids triggers robust contraction of 3D muscle, and assembloids are morphologically and functionally intact for up to 10 weeks post-fusion. Together, this system highlights the remarkable self-assembly capacity of 3D cultures to form functional circuits that could be used to understand development and disease.


Asunto(s)
Corteza Cerebral/fisiología , Corteza Motora/fisiología , Organoides/fisiología , Animales , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Vértebras Cervicales , Regulación de la Expresión Génica , Glutamatos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Músculos/fisiología , Mioblastos/metabolismo , Red Nerviosa/fisiología , Optogenética , Organoides/ultraestructura , Rombencéfalo/fisiología , Esferoides Celulares/citología , Médula Espinal/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA