Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Semin Cell Dev Biol ; 100: 29-51, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31862220

RESUMEN

The mammalian hearts have the least regenerative capabilities among tissues and organs. As such, heart regeneration has been and continues to be the ultimate goal in the treatment against acquired and congenital heart diseases. Uncovering such a long-awaited therapy is still extremely challenging in the current settings. On the other hand, this desperate need for effective heart regeneration has developed various forms of modern biotechnologies in recent years. These involve the transplantation of pluripotent stem cell-derived cardiac progenitors or cardiomyocytes generated in vitro and novel biochemical molecules along with tissue engineering platforms. Such newly generated technologies and approaches have been shown to effectively proliferate cardiomyocytes and promote heart repair in the diseased settings, albeit mainly preclinically. These novel tools and medicines give somehow credence to breaking down the barriers associated with re-building heart muscle. However, in order to maximize efficacy and achieve better clinical outcomes through these cell-based and/or cell-free therapies, it is crucial to understand more deeply the developmental cellular hierarchies/paths and molecular mechanisms in normal or pathological cardiogenesis. Indeed, the morphogenetic process of mammalian cardiac development is highly complex and spatiotemporally regulated by various types of cardiac progenitors and their paracrine mediators. Here we discuss the most recent knowledge and findings in cardiac progenitor cell biology and the major cardiogenic paracrine mediators in the settings of cardiogenesis, congenital heart disease, and heart regeneration.


Asunto(s)
Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Comunicación Paracrina , Células Madre Pluripotentes/metabolismo , Regeneración , Animales , Humanos , Miocardio/citología , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Ingeniería de Tejidos
2.
Stem Cells ; 38(6): 741-755, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32129551

RESUMEN

Cardiac progenitor formation is one of the earliest committed steps of human cardiogenesis and requires the cooperation of multiple gene sets governed by developmental signaling cascades. To determine the key regulators for cardiac progenitor formation, we have developed a two-stage genome-wide CRISPR-knockout screen. We mimicked the progenitor formation process by differentiating human pluripotent stem cells (hPSCs) into cardiomyocytes, monitored by two distinct stage markers of early cardiac mesodermal formation and commitment to a multipotent heart progenitor cell fate: MESP1 and ISL1, respectively. From the screen output, we compiled a list of 15 candidate genes. After validating seven of them, we identified ZIC2 as an essential gene for cardiac progenitor formation. ZIC2 is known as a master regulator of neurogenesis. hPSCs with ZIC2 mutated still express pluripotency markers. However, their ability to differentiate into cardiomyocytes was greatly attenuated. RNA-Seq profiling of the ZIC2-mutant cells revealed that the mutants switched their cell fate alternatively to the noncardiac cell lineage. Further, single cell RNA-seq analysis showed the ZIC2 mutants affected the apelin receptor-related signaling pathway during mesoderm formation. Our results provide a new link between ZIC2 and human cardiogenesis and document the potential power of a genome-wide unbiased CRISPR-knockout screen to identify the key steps in human mesoderm precursor cell- and heart progenitor cell-fate determination during in vitro hPSC cardiogenesis.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Estudio de Asociación del Genoma Completo/métodos , Corazón/fisiopatología , Mesodermo/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos , Ratones
3.
Mol Ther ; 26(7): 1644-1659, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29606507

RESUMEN

The generation of human pluripotent stem cell (hPSC)-derived ventricular progenitors and their assembly into a 3-dimensional in vivo functional ventricular heart patch has remained an elusive goal. Herein, we report the generation of an enriched pool of hPSC-derived ventricular progenitors (HVPs), which can expand, differentiate, self-assemble, and mature into a functional ventricular patch in vivo without the aid of any gel or matrix. We documented a specific temporal window, in which the HVPs will engraft in vivo. On day 6 of differentiation, HVPs were enriched by depleting cells positive for pluripotency marker TRA-1-60 with magnetic-activated cell sorting (MACS), and 3 million sorted cells were sub-capsularly transplanted onto kidneys of NSG mice where, after 2 months, they formed a 7 mm × 3 mm × 4 mm myocardial patch resembling the ventricular wall. The graft acquired several features of maturation: expression of ventricular marker (MLC2v), desmosomes, appearance of T-tubule-like structures, and electrophysiological action potential signature consistent with maturation, all this in a non-cardiac environment. We further demonstrated that HVPs transplanted into un-injured hearts of NSG mice remain viable for up to 8 months. Moreover, transplantation of 2 million HVPs largely preserved myocardial contractile function following myocardial infarction. Taken together, our study reaffirms the promising idea of using progenitor cells for regenerative therapy.


Asunto(s)
Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Proteínas con Homeodominio LIM/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Separación Celular/métodos , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/fisiología
6.
Nat Biomed Eng ; 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38831042

RESUMEN

The applicability of cytosine base editors is hindered by their dependence on sequence context and by off-target effects. Here, by using AlphaFold2 to predict the three-dimensional structure of 1,483 cytidine deaminases and by experimentally characterizing representative deaminases (selected from each structural cluster after categorizing them via partitional clustering), we report the discovery of a few deaminases with high editing efficiencies, diverse editing windows and increased ratios of on-target to off-target effects. Specifically, several deaminases induced C-to-T conversions with comparable efficiency at AC/TC/CC/GC sites, the deaminases could introduce stop codons in single-copy and multi-copy genes in mammalian cells without double-strand breaks, and some residue conversions at predicted DNA-interacting sites reduced off-target effects. Structure-based generative machine learning could be further leveraged to expand the applicability of base editors in gene therapies.

7.
Int J Biol Macromol ; 259(Pt 2): 129232, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38191104

RESUMEN

Ambrosia trifida (giant ragweed) is an invasive plant that can cause serious damage to natural ecosystems and severe respiratory allergies. However, the genomic basis of invasive adaptation and pollen allergens in Ambrosia species remain largely unknown. Here, we present a 1.66 Gb chromosome-scale reference genome for giant ragweed and identified multiple types of genome duplications, which are responsible for its rapid environmental adaptation and pollen development. The largest copies number and species-specific expansions of resistance-related gene families compared to Heliantheae alliance might contribute to resist stresses, pathogens and rapid adaptation. To extend the knowledge of evolutionary process of allergic pollen proteins, we predicted 26 and 168 potential pollen allergen candidates for giant ragweed and other Asteraceae plant species by combining machine learning and identity screening. Interestingly, we observed a specific tandemly repeated array for potential allergenic pectate lyases among Ambrosia species. Rapid evolutionary rates on putative pectate lyase allergens may imply a crucial role of nonsynonymous mutations on amino acid residues for plant biological function and allergenicity. Altogether, this study provides insight into the molecular ecological adaptation and putative pollen allergens prediction that will be helpful in promoting invasion genomic research and evolution of putative pollen allergy in giant ragweed.


Asunto(s)
Ambrosia , Hipersensibilidad , Ambrosia/genética , Antígenos de Plantas/genética , Ecosistema , Alérgenos/genética , Alérgenos/química , Polen/genética , Cromosomas
8.
Cardiovasc Res ; 119(5): 1202-1217, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36635482

RESUMEN

AIMS: Retinoic acid (RA) signalling is essential for heart development, and dysregulation of the RA signalling can cause several types of cardiac outflow tract (OFT) defects, the most frequent congenital heart disease (CHD) in humans. Matthew-Wood syndrome is caused by inactivating mutations of a transmembrane protein gene STRA6 that transports vitamin A (retinol) from extracellular into intracellular spaces. This syndrome shows a broad spectrum of malformations including CHD, although murine Stra6-null neonates did not exhibit overt heart defects. Thus, the detailed mechanisms by which STRA6 mutations could lead to cardiac malformations in humans remain unclear. Here, we investigated the role of STRA6 in the context of human cardiogenesis and CHD. METHODS AND RESULTS: To gain molecular signatures in species-specific cardiac development, we first compared single-cell RNA sequencing (RNA-seq) datasets, uniquely obtained from human and murine embryonic hearts. We found that while STRA6 mRNA was much less frequently expressed in murine embryonic heart cells derived from the Mesp1+ lineage tracing mice (Mesp1Cre/+; Rosa26tdTomato), it was expressed predominantly in the OFT region-specific heart progenitors in human developing hearts. Next, we revealed that STRA6-knockout human embryonic stem cells (hESCs) could differentiate into cardiomyocytes similarly to wild-type hESCs, but could not differentiate properly into mesodermal nor neural crest cell-derived smooth muscle cells (SMCs) in vitro. This is supported by the population RNA-seq data showing down-regulation of the SMC-related genes in the STRA6-knockout hESC-derived cells. Further, through machinery assays, we identified the previously unrecognized interaction between RA nuclear receptors RARα/RXRα and TBX1, an OFT-specific cardiogenic transcription factor, which would likely act downstream to STRA6-mediated RA signalling in human cardiogenesis. CONCLUSION: Our study highlights the critical role of human-specific STRA6 progenitors for proper induction of vascular SMCs that is essential for normal OFT formation. Thus, these results shed light on novel and human-specific CHD mechanisms, driven by STRA6 mutations.


Asunto(s)
Cardiopatías Congénitas , Músculo Liso Vascular , Humanos , Animales , Ratones , Músculo Liso Vascular/metabolismo , Corazón , Cardiopatías Congénitas/genética , Regulación de la Expresión Génica , Tretinoina/farmacología , Tretinoina/metabolismo , Vitamina A , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
9.
Nat Commun ; 14(1): 5435, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37669989

RESUMEN

Cardiogenic growth factors play important roles in heart development. Placental growth factor (PLGF) has previously been reported to have angiogenic effects; however, its potential role in cardiogenesis has not yet been determined. We analyze single-cell RNA-sequencing data derived from human and primate embryonic hearts and find PLGF shows a biphasic expression pattern, as it is expressed specifically on ISL1+ second heart field progenitors at an earlier stage and on vascular smooth muscle cells (SMCs) and endothelial cells (ECs) at later stages. Using chemically modified mRNAs (modRNAs), we generate a panel of cardiogenic growth factors and test their effects on enhancing cardiomyocyte (CM) and EC induction during different stages of human embryonic stem cell (hESC) differentiations. We discover that only the application of PLGF modRNA at early time points of hESC-CM differentiation can increase both CM and EC production. Conversely, genetic deletion of PLGF reduces generation of CMs, SMCs and ECs in vitro. We also confirm in vivo beneficial effects of PLGF modRNA for development of human heart progenitor-derived cardiac muscle grafts on murine kidney capsules. Further, we identify the previously unrecognized PLGF-related transcriptional networks driven by EOMES and SOX17. These results shed light on the dual cardiomyogenic and vasculogenic effects of PLGF during heart development.


Asunto(s)
Células Endoteliales , Miocardio , Femenino , Humanos , Animales , Ratones , Factor de Crecimiento Placentario , Miocitos Cardíacos , Diferenciación Celular
10.
Autophagy ; 17(11): 3577-3591, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33535890

RESUMEN

Scavenger receptors are pattern recognition receptors that recognize both foreign and self-ligands, and initiate different mechanisms of cellular activation, often as co-receptors. The function of scavenger receptor CD36 in the immune system has mostly been studied in macrophages but it is also highly expressed by innate type B cells where its function is less explored. Here we report that CD36 is involved in macro-autophagy/autophagy in B cells, and in its absence, the humoral immune response is impaired. We found that CD36-deficient B cells exhibit a significantly reduced plasma cell formation, proliferation, mitochondrial mobilization and oxidative phosphorylation. These changes were accompanied by impaired initiation of autophagy, and we found that CD36 regulated autophagy and colocalized with autophagosome membrane protein MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3). When we investigated T-cell-dependent immune responses, we found that mice with CD36 deficiency, specifically in B cells, exhibited attenuated germinal center responses, class switching, and antibody production as well as autophagosome formation. These findings establish a critical role for CD36 in B cell responses and may also contribute to our understanding of CD36-mediated autophagy in other cells as well as in B cell lymphomas that have been shown to express the receptor.Abbreviations: AICDA/AID: activation-induced cytidine deaminase; ATG5: autophagy related 5; ATP: adenosine triphosphate; BCR: B-cell receptor; CPG: unmethylated cytosine-guanosine; CQ: chloroquine; DC: dendritic cells; FOB: follicular B cells; GC: germinal center; Ig: immunoglobulin; LPS: lipopolysaccharide; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MFI: mean fluorescence intensity; MZB: marginal zone B cells; NP-CGG: 4-hydroxy-3-nitrophenylacetyl-chicken gamma globulin; OCR: oxygen consumption rate; oxLDL: oxidized low-density lipoprotein; PC: plasma cells; Rapa: rapamycin; SQSTM1/p62: sequestosome 1; SRBC: sheep red blood cells; Tfh: follicular helper T cells; TLR: toll-like receptor.


Asunto(s)
Autofagia , Linfocitos B/fisiología , Antígenos CD36/fisiología , Inmunidad Humoral , Proteínas Asociadas a Microtúbulos/fisiología , Animales , Autofagosomas/metabolismo , Autofagosomas/fisiología , Autofagia/fisiología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD36/metabolismo , Diferenciación Celular , Proliferación Celular , Humanos , Cambio de Clase de Inmunoglobulina , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Células Plasmáticas/fisiología , Linfocitos T/inmunología , Linfocitos T/fisiología
11.
Cancer Res ; 81(4): 956-967, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33293426

RESUMEN

The progression and metastatic capacity of solid tumors are strongly influenced by immune cells in the tumor microenvironment. In non-small cell lung cancer (NSCLC), accumulation of anti-inflammatory tumor-associated macrophages (TAM) is associated with worse clinical outcome and resistance to therapy. Here we investigated the immune landscape of NSCLC in the presence of protumoral TAMs expressing the macrophage receptor with collagenous structure (MARCO). MARCO-expressing TAM numbers correlated with increased occurrence of regulatory T cells and effector T cells and decreased natural killer (NK) cells in these tumors. Furthermore, transcriptomic data from the tumors uncovered a correlation between MARCO expression and the anti-inflammatory cytokine IL37. In vitro studies subsequently showed that lung cancer cells polarized macrophages to express MARCO and gain an immune-suppressive phenotype through the release of IL37. MARCO-expressing TAMs blocked cytotoxic T-cell and NK-cell activation, inhibiting their proliferation, cytokine production, and tumor killing capacity. Mechanistically, MARCO+ macrophages enhanced regulatory T (Treg) cell proliferation and IL10 production and diminished CD8 T-cell activities. Targeting MARCO or IL37 receptor (IL37R) by antibody or CRISPR knockout of IL37 in lung cancer cell lines repolarized TAMs, resulting in recovered cytolytic activity and antitumoral capacity of NK cells and T cells and downmodulated Treg cell activities. In summary, our data demonstrate a novel immune therapeutic approach targeting human TAMs immune suppression of NK- and T-cell antitumor activities. SIGNIFICANCE: This study defines tumor-derived IL37 and the macrophage scavenger receptor MARCO as potential therapeutic targets to remodel the immune-suppressive microenvironment in patients with lung cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/4/956/F1.large.jpg.


Asunto(s)
Receptores Inmunológicos , Receptores de Interleucina-1 , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Tolerancia Inmunológica/genética , Tolerancia Inmunológica/inmunología , Inmunoterapia/métodos , Interleucina-1/genética , Interleucina-1/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Ratones , Ratones Noqueados , Terapia Molecular Dirigida/métodos , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Linfocitos T Reguladores/patología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología
12.
Nat Commun ; 12(1): 5126, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34446705

RESUMEN

Embryonic development is largely conserved among mammals. However, certain genes show divergent functions. By generating a transcriptional atlas containing >30,000 cells from post-implantation non-human primate embryos, we uncover that ISL1, a gene with a well-established role in cardiogenesis, controls a gene regulatory network in primate amnion. CRISPR/Cas9-targeting of ISL1 results in non-human primate embryos which do not yield viable offspring, demonstrating that ISL1 is critically required in primate embryogenesis. On a cellular level, mutant ISL1 embryos display a failure in mesoderm formation due to reduced BMP4 signaling from the amnion. Via loss of function and rescue studies in human embryonic stem cells we confirm a similar role of ISL1 in human in vitro derived amnion. This study highlights the importance of the amnion as a signaling center during primate mesoderm formation and demonstrates the potential of in vitro primate model systems to dissect the genetics of early human embryonic development.


Asunto(s)
Amnios/metabolismo , Macaca fascicularis/embriología , Mesodermo/embriología , Amnios/embriología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Macaca fascicularis/genética , Macaca fascicularis/metabolismo , Mesodermo/metabolismo , Embarazo , Transducción de Señal
13.
Dev Cell ; 48(4): 475-490.e7, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30713072

RESUMEN

The morphogenetic process of mammalian cardiac development is complex and highly regulated spatiotemporally by multipotent cardiac stem/progenitor cells (CPCs). Mouse studies have been informative for understanding mammalian cardiogenesis; however, similar insights have been poorly established in humans. Here, we report comprehensive gene expression profiles of human cardiac derivatives from multipotent CPCs to intermediates and mature cardiac cells by population and single-cell RNA-seq using human embryonic stem cell-derived and embryonic/fetal heart-derived cardiac cells micro-dissected from specific heart compartments. Importantly, we discover a uniquely human subset of cono-ventricular region-specific CPCs, marked by LGR5. At 4 to 5 weeks of fetal age, the LGR5+ population appears to emerge specifically in the proximal outflow tract of human embryonic hearts and thereafter promotes cardiac development and alignment through expansion of the ISL1+TNNT2+ intermediates. The current study contributes to a deeper understanding of human cardiogenesis, which may uncover the putative origins of certain human congenital cardiac malformations.


Asunto(s)
Diferenciación Celular/fisiología , Miocitos Cardíacos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Análisis de la Célula Individual , Animales , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Ventrículos Cardíacos/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Proteínas con Homeodominio LIM/genética , Ratones Endogámicos C57BL , Células Madre Multipotentes , Miocardio/metabolismo , Organogénesis , Análisis de la Célula Individual/métodos
15.
Sci Rep ; 7(1): 1549, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28484230

RESUMEN

Human pluripotent stem cells (hPSCs) offer tremendous promise in tissue engineering and cell-based therapies due to their unique combination of two properties: pluripotency and unlimited proliferative capacity. However, directed differentiation of hPSCs to clinically relevant cell lineages is needed to achieve the goal of hPSC-based therapies. This requires a deep understanding of how cell signaling pathways converge on the nucleus to control differentiation and the ability to dissect gene function in a temporal manner. Here, we report the use of the PiggyBac transposon and a Tet-On 3G drug-inducible gene expression system to achieve versatile inducible gene expression in hPSC lines. Our new system, XLone, offers improvement over previous Tet-On systems with significantly reduced background expression and increased sensitivity to doxycycline. Transgene expression in hPSCs is tightly regulated in response to doxycycline treatment. In addition, the PiggyBac elements in our XLone construct provide a rapid and efficient strategy for generating stable transgenic hPSCs. Our inducible gene expression PiggyBac transposon system should facilitate the study of gene function and directed differentiation in human stem cells.


Asunto(s)
Elementos Transponibles de ADN/genética , Doxiciclina/farmacología , Células Madre Pluripotentes/metabolismo , Diferenciación Celular/efectos de los fármacos , Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Cinética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Plásmidos/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Factores de Tiempo , Transgenes
16.
Cell Reprogram ; 17(3): 221-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26053521

RESUMEN

Adipose tissue is an attractive source of easily accessible adult candidate cells for regenerative medicine. Adipose tissue-derived mesenchymal stem cells (ADSCs) have multipotency and strong proliferation and differentiation capabilities in vitro. However, as mesodermal multipotent stem cells, whether the ADSCs can convert into induced neural stem cells (NSCs) has so far not been demonstrated. In this study, we found that normally the naïve ADSCs cultured as either monolayer or spheres in NSC medium did not express Sox2 and Pax6 genes and proteins, and could not differentiate to neuron-like cells. However, when we introduced the Sox2 gene into ADSCs by retrovirus, they exhibited a typical NSC-like morphology, and could be passaged continuously, and expressed NSC specific markers Sox2 and Pax6. In addition, the ADSC-derived NSC-like cells displayed the ability to differentiate into neuron-like cells when switched to the differentiation culture medium, expressing neuronal markers, including Tuj1 and MAP2 genes and proteins. Our results suggest the ADSCs can be converted into induced NSC-like cells with a single transcription factor Sox2. This finding could provide another alternative cell source for cell therapy of neurological disorders.


Asunto(s)
Tejido Adiposo/fisiología , Transdiferenciación Celular , Células Madre Mesenquimatosas/citología , Células-Madre Neurales/citología , Factores de Transcripción SOXB1/genética , Tejido Adiposo/metabolismo , Animales , Células Madre Mesenquimatosas/metabolismo , Ratones , Transgenes
17.
Cell Cycle ; 14(8): 1282-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25692793

RESUMEN

Somatic cells can be reprogrammed into embryonic stem cells (ESCs) by nuclear transfer (NT-ESCs), or into induced pluripotent stem cells (iPSCs) by the "Yamanaka method." However, recent studies have indicated that mouse and human iPSCs are prone to epigenetic and transcriptional aberrations, and that NT-ESCs correspond more closely to ESCs derived from in vitro fertilized embryos than iPSCs. In addition, the procedure of NT-ESCs does not involve gene modification. Demonstration of generation of NT-ESCs using an easily-accessible source of adult cell types would be very important. Adipose tissue is a source of readily accessible donor cells and can be isolated from both males and females at different ages. Here we report that NT-ESCs can be generated from adipose tissue-derived cells (ADCs). At morphological, mRNA and protein levels, these NT-ESCs show classic ESC colonies, exhibit alkaline phosphatase (AP) activity, and display normal diploid karyotypes. Importantly, these cells express pluripotent markers including Oct4, Sox2, Nanog and SSEA-1. Furthermore, they can differentiate in vivo into various types of cells from 3 germinal layers by teratoma formation assays. This study demonstrates for the first time that ESCs can be generated from the adipose tissue by somatic cell nuclear transfer (SCNT) and suggests that ADCs can be a new donor-cell type for potential therapeutic cloning.


Asunto(s)
Tejido Adiposo/citología , Núcleo Celular/metabolismo , Células Madre Embrionarias de Ratones/citología , Técnicas de Transferencia Nuclear , Animales , Diferenciación Celular , Reprogramación Celular , Antígeno Lewis X/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción/metabolismo
18.
Cell Res ; 25(1): 67-79, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25475058

RESUMEN

Spermatogonial stem cells (SSCs) can produce numerous male gametes after transplantation into recipient testes, presenting a valuable approach for gene therapy and continuous production of gene-modified animals. However, successful genetic manipulation of SSCs has been limited, partially due to complexity and low efficiency of currently available genetic editing techniques. Here, we show that efficient genetic modifications can be introduced into SSCs using the CRISPR-Cas9 system. We used the CRISPR-Cas9 system to mutate an EGFP transgene or the endogenous Crygc gene in SCCs. The mutated SSCs underwent spermatogenesis after transplantation into the seminiferous tubules of infertile mouse testes. Round spermatids were generated and, after injection into mature oocytes, supported the production of heterozygous offspring displaying the corresponding mutant phenotypes. Furthermore, a disease-causing mutation in Crygc (Crygc(-/-)) that pre-existed in SSCs could be readily repaired by CRISPR-Cas9-induced nonhomologous end joining (NHEJ) or homology-directed repair (HDR), resulting in SSC lines carrying the corrected gene with no evidence of off-target modifications as shown by whole-genome sequencing. Fertilization using round spermatids generated from these lines gave rise to offspring with the corrected phenotype at an efficiency of 100%. Our results demonstrate efficient gene editing in mouse SSCs by the CRISPR-Cas9 system, and provide the proof of principle of curing a genetic disease via gene correction in SSCs.


Asunto(s)
Células Madre Adultas/metabolismo , Células Madre Adultas/trasplante , Sistemas CRISPR-Cas , Infertilidad Masculina/genética , Infertilidad Masculina/terapia , Mutagénesis , Animales , Células Cultivadas , Reparación del ADN por Unión de Extremidades , Femenino , Genes erbB-1 , Terapia Genética , Masculino , Ratones , Ratones Endogámicos BALB C , Espermatogénesis , Transgenes , gamma-Cristalinas/genética
19.
Stem Cells Dev ; 23(4): 363-71, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24083854

RESUMEN

White adipose tissue (WAT) is a very attractive source of mesenchymal stem cells (MSCs) because of its availability and ease of harvest. However, the current method of isolating adipose tissue-derived MSCs often relies on the adhesiveness of the cultured stromal-vascular fraction (SVF). Unfortunately, the SVF is a heterogeneous cell population containing many cell types, including adipocyte precursor cells, endothelial cells, pericytes, multipotent MSCs, erythrocytes, and hematopoietic cells. Here we systematically characterized the adipose tissue-derived lineage-negative (Lin(-)) cell population using various surface markers and a set of cell proliferation and differentiation assays. We demonstrate clearly that the Lin(-) cell population represents enriched MSCs, which were identified by their high expression of MSC surface markers, and that these cells are a robust population with a vigorous growth capability and delayed aging. This cell population also demonstrated a much higher capacity for differentiation into osteogenic, chondrogenic and adipogenic cell lineages related to MSCs than did the SVF. These cells promoted recovery from limb ischemia, likely via production of vascular endothelial growth factor, an angiogenic factor. Our study demonstrates that Lin(-) cells are enriched in MSCs and provides a reliable method for isolating purer MSCs than SVF cells from the WAT, especially for obtaining fresh MSCs for clinical applications. In summary, this study identified a new, reliable method for enrichment of WAT MSCs with regenerative repairing features.


Asunto(s)
Miembro Posterior/patología , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Tejido Adiposo Blanco/patología , Animales , Diferenciación Celular , Proliferación Celular , Separación Celular , Células Cultivadas , Citometría de Flujo , Miembro Posterior/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Fenotipo , Regeneración , Flujo Sanguíneo Regional , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Cell Res ; 23(1): 92-106, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23147797

RESUMEN

Induced pluripotent stem (iPS) cells generated using Yamanaka factors have great potential for use in autologous cell therapy. However, genomic abnormalities exist in human iPS cells, and most mouse iPS cells are not fully pluripotent, as evaluated by the tetraploid complementation assay (TCA); this is most likely associated with the DNA damage response (DDR) occurred in early reprogramming induced by Yamanaka factors. In contrast, nuclear transfer can faithfully reprogram somatic cells into embryonic stem (ES) cells that satisfy the TCA. We thus hypothesized that factors involved in oocyte-induced reprogramming may stabilize the somatic genome during reprogramming, and improve the quality of the resultant iPS cells. To test this hypothesis, we screened for factors that could decrease DDR signals during iPS cell induction. We determined that Zscan4, in combination with the Yamanaka factors, not only remarkably reduced the DDR but also markedly promoted the efficiency of iPS cell generation. The inclusion of Zscan4 stabilized the genomic DNA, resulting in p53 downregulation. Furthermore, Zscan4 also enhanced telomere lengthening as early as 3 days post-infection through a telomere recombination-based mechanism. As a result, iPS cells generated with addition of Zscan4 exhibited longer telomeres than classical iPS cells. Strikingly, more than 50% of iPS cell lines (11/19) produced via this "Zscan4 protocol" gave rise to live-borne all-iPS cell mice as determined by TCA, compared to 1/12 for lines produced using the classical Yamanaka factors. Our findings provide the first demonstration that maintaining genomic stability during reprogramming promotes the generation of high quality iPS cells.


Asunto(s)
Inestabilidad Genómica , Células Madre Pluripotentes Inducidas/citología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Reprogramación Celular , Reparación del ADN , Humanos , Ratones , Telómero/metabolismo , Teratoma/patología , Tetraploidía , Factores de Transcripción/genética , Transfección , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA