Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Development ; 151(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38895963

RESUMEN

The sixth SY-Stem Symposium, jointly organized by the Research Institute of Molecular Pathology and the Institute of Molecular Biotechnology took place in Vienna in March 2024. Again, aspiring new group leaders were given a stage to present their work and vision of their labs. To round up the excellent program, the scientific organizers included renowned keynote speakers. Here, we provide a summary of the talks covering topics such as early embryogenesis, nervous system development and disease, regeneration and the latest technologies.


Asunto(s)
Desarrollo Embrionario , Humanos , Animales , Regeneración/fisiología , Células Madre/citología , Sistema Nervioso/embriología , Diferenciación Celular
2.
Life Sci Alliance ; 7(4)2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38238086

RESUMEN

The X-linked form of Opitz BBB/G syndrome (OS) is a monogenic disorder in which symptoms are established early during embryonic development. OS is caused by pathogenic variants in the X-linked gene MID1 Disease-associated variants are distributed across the entire gene locus, except for the N-terminal really interesting new gene (RING) domain that encompasses the E3 ubiquitin ligase activity. By using genome-edited human induced pluripotent stem cell lines, we here show that absence of isoforms containing the RING domain of MID1 causes severe patterning defects in human brain organoids. We observed a prominent neurogenic deficit with a reduction in neural tissue and a concomitant increase in choroid plexus-like structures. Transcriptome analyses revealed a deregulation of patterning pathways very early on, even preceding neural induction. Notably, the observed phenotypes starkly contrast with those observed in MID1 full-knockout organoids, indicating the presence of a distinct mechanism that underlies the patterning defects. The severity and early onset of these phenotypes could potentially account for the absence of patients carrying pathogenic variants in exon 1 of the MID1 gene coding for the N-terminal RING domain.


Asunto(s)
Esófago , Hipertelorismo , Hipospadias , Células Madre Pluripotentes Inducidas , Proteínas Nucleares , Humanos , Encéfalo/metabolismo , Esófago/anomalías , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de Microtúbulos/química , Proteínas Nucleares/genética , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Int J Biol Macromol ; 261(Pt 1): 129608, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38266846

RESUMEN

Natural fermentation with sun-drying is a modification that promotes the expansion capacity of starch, and its effects on potato starch have not been reported so far. The aim of this study was to evaluate the effects of the amylose content of potato (Solanum tuberosum L.) starches and natural fermentation followed by oven or sun drying on its properties. Cassava starch was also used a control. Native and fermented starches were evaluated based on their chemical composition, amylose, carboxyl and carbonyl content as well as their thermal, pasty, and morphological properties. The fermentation water was evaluated by pH and titratable acidity to control the process. Puffed balls were prepared to evaluate expandability, mass loss, porosity and texture. The fermentation intensity was greater for potato and cassava starch with low-amylose content than for potato starch with higher amylose content. In addition, the acidity of the fermentation water increased faster with cassava starch than with potato starches. The fermented potato starches with the highest amylose content had low acidity and low expansion capacity compared to the fermented potato and cassava starches with low-amylose content. Fermentation and sun-drying of low-amylose potato and cassava starches increased the expansion and reduced the hardness of the puffed balls.


Asunto(s)
Solanum tuberosum , Almidón , Almidón/química , Amilosa/química , Solanum tuberosum/química , Fermentación , Agua
4.
Stem Cell Reports ; 18(12): 2418-2433, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-37995703

RESUMEN

Although adult subependymal zone (SEZ) neural stem cells mostly generate GABAergic interneurons, a small progenitor population expresses the proneural gene Neurog2 and produces glutamatergic neurons. Here, we determined whether Neurog2 could respecify SEZ neural stem cells and their progeny toward a glutamatergic fate. Retrovirus-mediated expression of Neurog2 induced the glutamatergic lineage markers TBR2 and TBR1 in cultured SEZ progenitors, which differentiated into functional glutamatergic neurons. Likewise, Neurog2-transduced SEZ progenitors acquired glutamatergic neuron hallmarks in vivo. Intriguingly, they failed to migrate toward the olfactory bulb and instead differentiated within the SEZ or the adjacent striatum, where they received connections from local neurons, as indicated by rabies virus-mediated monosynaptic tracing. In contrast, lentivirus-mediated expression of Neurog2 failed to reprogram early SEZ neurons, which maintained GABAergic identity and migrated to the olfactory bulb. Our data show that NEUROG2 can program SEZ progenitors toward a glutamatergic identity but fails to reprogram their neuronal progeny.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células-Madre Neurales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neuronas/metabolismo , Células-Madre Neurales/metabolismo , Diferenciación Celular , Bulbo Olfatorio/metabolismo , Neurogénesis/fisiología
5.
Cell Reprogram ; 25(5): 212-223, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37366790

RESUMEN

Studying human somatic cell-to-neuron conversion using primary brain-derived cells as starting cell source is hampered by limitations and variations in human biopsy material. Thus, delineating the molecular variables that allow changing the identity of somatic cells, permit adoption of neuronal phenotypes, and foster maturation of induced neurons (iNs) is challenging. Based on our previous results that pericytes derived from the adult human cerebral cortex can be directly converted into iNs (Karow et al., 2018; Karow et al., 2012), we here introduce human induced pluripotent stem cell (hiPSC)-derived pericytes (hiPSC-pericytes) as a versatile and more uniform tool to study the pericyte-to-neuron conversion process. This strategy enables us to derive scalable cell numbers and allows for engineering of the starting cell population such as introducing reporter tools before differentiation into hiPSC-pericytes and subsequent iN conversion. Harvesting the potential of this approach, we established hiPSC-derived human-human neuronal cocultures that not only allow for independent manipulation of each coculture partner but also resulted in morphologically more mature iNs. In summary, we exploit hiPSC-based methods to facilitate the analysis of human somatic cell-to-neuron conversion.


Asunto(s)
Células Madre Pluripotentes Inducidas , Adulto , Humanos , Reprogramación Celular , Pericitos/fisiología , Neuronas , Diferenciación Celular/fisiología
6.
Curr Opin Genet Dev ; 70: 97-103, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34333231

RESUMEN

Direct lineage reprogramming challenges our traditional view on basic aspects of cellular identity, and in particular on processes crucial for identity acquisition. This is partly because in direct lineage reprogramming but not during natural differentiation processes changing cellular identity can occur in the absence of mitosis. Only recently, technologies emerged to deconstruct the cellular and molecular processes governing the transitory states a cell passes through on the journey from its original identity to the new target cell fate. Here we discuss arising concepts on the nature of these transitory states and the challenges and decisions cells must conquer to reach their new cellular identity.


Asunto(s)
Linaje de la Célula , Reprogramación Celular , Animales , Diferenciación Celular , Humanos
7.
Int J Mol Sci ; 22(16)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34445575

RESUMEN

The gut-brain axis is a bidirectional communication system driven by neural, hormonal, metabolic, immunological, and microbial signals. Signaling events from the gut can modulate brain function and recent evidence suggests that the gut-brain axis may play a pivotal role in linking gastrointestinal and neurological diseases. Accordingly, accumulating evidence has suggested a link between inflammatory bowel diseases (IBDs) and neurodegenerative, as well as neuroinflammatory diseases. In this context, clinical, epidemiological and experimental data have demonstrated that IBD predisposes a person to pathologies of the central nervous system (CNS). Likewise, a number of neurological disorders are associated with changes in the intestinal environment, which are indicative for disease-mediated gut-brain inter-organ communication. Although this axis was identified more than 20 years ago, the sequence of events and underlying molecular mechanisms are poorly defined. The emergence of precision medicine has uncovered the need to take into account non-intestinal symptoms in the context of IBD that could offer the opportunity to tailor therapies to individual patients. The aim of this review is to highlight recent findings supporting the clinical and biological link between the gut and brain, as well as its clinical significance for IBD as well as neurodegeneration and neuroinflammation. Finally, we focus on novel human-specific preclinical models that will help uncover disease mechanisms to better understand and modulate the function of this complex system.


Asunto(s)
Encéfalo/patología , Enfermedades del Sistema Nervioso Central/patología , Enfermedades Inflamatorias del Intestino/complicaciones , Animales , Encéfalo/metabolismo , Enfermedades del Sistema Nervioso Central/etiología , Enfermedades del Sistema Nervioso Central/metabolismo , Humanos
8.
Sci Adv ; 7(15)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33827819

RESUMEN

Neural cell diversity is essential to endow distinct brain regions with specific functions. During development, progenitors within these regions are characterized by specific gene expression programs, contributing to the generation of diversity in postmitotic neurons and astrocytes. While the region-specific molecular diversity of neurons and astrocytes is increasingly understood, whether these cells share region-specific programs remains unknown. Here, we show that in the neocortex and thalamus, neurons and astrocytes express shared region-specific transcriptional and epigenetic signatures. These signatures not only distinguish cells across these two brain regions but are also detected across substructures within regions, such as distinct thalamic nuclei, where clonal analysis reveals the existence of common nucleus-specific progenitors for neurons and astrocytes. Consistent with their shared molecular signature, regional specificity is maintained following astrocyte-to-neuron reprogramming. A detailed understanding of these regional-specific signatures may thus inform strategies for future cell-based brain repair.


Asunto(s)
Astrocitos , Neocórtex , Astrocitos/metabolismo , Epigenómica , Neuronas/fisiología , Tálamo
9.
Nat Neurosci ; 21(7): 932-940, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29915193

RESUMEN

Ectopic expression of defined transcription factors can force direct cell-fate conversion from one lineage to another in the absence of cell division. Several transcription factor cocktails have enabled successful reprogramming of various somatic cell types into induced neurons (iNs) of distinct neurotransmitter phenotype. However, the nature of the intermediate states that drive the reprogramming trajectory toward distinct iN types is largely unknown. Here we show that successful direct reprogramming of adult human brain pericytes into functional iNs by Ascl1 and Sox2 encompasses transient activation of a neural stem cell-like gene expression program that precedes bifurcation into distinct neuronal lineages. During this transient state, key signaling components relevant for neural induction and neural stem cell maintenance are regulated by and functionally contribute to iN reprogramming and maturation. Thus, Ascl1- and Sox2-mediated reprogramming into a broad spectrum of iN types involves the unfolding of a developmental program via neural stem cell-like intermediates.


Asunto(s)
Linaje de la Célula/fisiología , Reprogramación Celular/fisiología , Células-Madre Neurales/fisiología , Neuronas/fisiología , Pericitos/fisiología , Adulto , Anciano , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Células-Madre Neurales/citología , Neuronas/citología , Pericitos/citología , Factores de Transcripción SOXB1/genética , Adulto Joven
11.
Cell Stem Cell ; 18(3): 396-409, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26748418

RESUMEN

Despite the widespread interest in direct neuronal reprogramming, the mechanisms underpinning fate conversion remain largely unknown. Our study revealed a critical time point after which cells either successfully convert into neurons or succumb to cell death. Co-transduction with Bcl-2 greatly improved negotiation of this critical point by faster neuronal differentiation. Surprisingly, mutants with reduced or no affinity for Bax demonstrated that Bcl-2 exerts this effect by an apoptosis-independent mechanism. Consistent with a caspase-independent role, ferroptosis inhibitors potently increased neuronal reprogramming by inhibiting lipid peroxidation occurring during fate conversion. Genome-wide expression analysis confirmed that treatments promoting neuronal reprogramming elicit an anti-oxidative stress response. Importantly, co-expression of Bcl-2 and anti-oxidative treatments leads to an unprecedented improvement in glial-to-neuron conversion after traumatic brain injury in vivo, underscoring the relevance of these pathways in cellular reprograming irrespective of cell type in vitro and in vivo.


Asunto(s)
Técnicas de Reprogramación Celular , Reprogramación Celular , Neuroglía/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Transducción Genética , Animales , Ratones , Neuroglía/citología , Neuronas/citología , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/genética
12.
EMBO J ; 34(7): 832-4, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25691243

RESUMEN

Early development of the mammalian cerebral cortex proceeds via a sequence of proliferative and differentiative steps from neural stem cells toward neurons and glia. However, how these steps are molecularly orchestrated is still only partially understood. In this issue of The EMBO Journal, Artegiani and colleagues implicate Tox, a HMG-box transcription factor previously known only for its role in lymphocyte development, in early cortical development.


Asunto(s)
Calcineurina/metabolismo , Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/fisiología , Animales
13.
J Vis Exp ; (87)2014 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-24893711

RESUMEN

Direct lineage-reprogramming of non-neuronal cells into induced neurons (iNs) may provide insights into the molecular mechanisms underlying neurogenesis and enable new strategies for in vitro modeling or repairing the diseased brain. Identifying brain-resident non-neuronal cell types amenable to direct conversion into iNs might allow for launching such an approach in situ, i.e. within the damaged brain tissue. Here we describe a protocol developed in the attempt of identifying cells derived from the adult human brain that fulfill this premise. This protocol involves: (1) the culturing of human cells from the cerebral cortex obtained from adult human brain biopsies; (2) the in vitro expansion (approximately requiring 2-4 weeks) and characterization of the culture by immunocytochemistry and flow cytometry; (3) the enrichment by fluorescence-activated cell sorting (FACS) using anti-PDGF receptor-ß and anti-CD146 antibodies; (4) the retrovirus-mediated transduction with the neurogenic transcription factors sox2 and ascl1; (5) and finally the characterization of the resultant pericyte-derived induced neurons (PdiNs) by immunocytochemistry (14 days to 8 weeks following retroviral transduction). At this stage, iNs can be probed for their electrical properties by patch-clamp recording. This protocol provides a highly reproducible procedure for the in vitro lineage conversion of brain-resident pericytes into functional human iNs.


Asunto(s)
Reprogramación Celular/fisiología , Corteza Cerebral/citología , Neuronas/citología , Pericitos/citología , Técnicas de Cultivo de Célula , Linaje de la Célula , Citometría de Flujo , Humanos , Inmunohistoquímica , Neuronas/metabolismo , Técnicas de Placa-Clamp , Pericitos/metabolismo , Retroviridae/genética , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Transducción Genética
14.
PLoS One ; 9(4): e96279, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24781921

RESUMEN

A cell therapy strategy utilizing genetically-corrected induced pluripotent stem cells (iPSC) may be an attractive approach for genetic disorders such as muscular dystrophies. Methods for genetic engineering of iPSC that emphasize precision and minimize random integration would be beneficial. We demonstrate here an approach in the mdx mouse model of Duchenne muscular dystrophy that focuses on the use of site-specific recombinases to achieve genetic engineering. We employed non-viral, plasmid-mediated methods to reprogram mdx fibroblasts, using phiC31 integrase to insert a single copy of the reprogramming genes at a safe location in the genome. We next used Bxb1 integrase to add the therapeutic full-length dystrophin cDNA to the iPSC in a site-specific manner. Unwanted DNA sequences, including the reprogramming genes, were then precisely deleted with Cre resolvase. Pluripotency of the iPSC was analyzed before and after gene addition, and ability of the genetically corrected iPSC to differentiate into myogenic precursors was evaluated by morphology, immunohistochemistry, qRT-PCR, FACS analysis, and intramuscular engraftment. These data demonstrate a non-viral, reprogramming-plus-gene addition genetic engineering strategy utilizing site-specific recombinases that can be applied easily to mouse cells. This work introduces a significant level of precision in the genetic engineering of iPSC that can be built upon in future studies.


Asunto(s)
Reprogramación Celular , Distrofina/genética , Ingeniería Genética/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Integrasas/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Animales , Línea Celular , Terapia Genética/métodos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx/genética , Desarrollo de Músculos
15.
Bioessays ; 35(9): 771-4, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23757054

RESUMEN

Pericytes, typically attached to the walls of microvessels in almost all organs, interact with endothelial cells and take part in diverse biological processes, e.g. blood vessel regulation and tissue repair. This suggests that pericytes harbor a remarkable degree of cellular plasticity, which could potentially be employed for the treatment of diseases affecting diverse tissues such as the skeletal muscle and the central nervous system. Here, we follow pericytes on their journey across Waddington's epigenetic landscape, descending from their origin, along a path guided by environmental signals or ectopic transcription factors, at the end of which they acquire a new identity, e.g. muscle or nerve cells. The central theme of this review is the question of whether pericytes can be enticed to differentiate into whatever cell type is needed, and thus provide an endogenous cellular source for treating as yet incurable diseases--like a magic bullet.


Asunto(s)
Diferenciación Celular , Pericitos/citología , Cicatrización de Heridas/fisiología , Animales , Linaje de la Célula/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética , Humanos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Factores de Transcripción
16.
Cell Stem Cell ; 11(4): 471-6, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-23040476

RESUMEN

Reprogramming of somatic cells into neurons provides a new approach toward cell-based therapy of neurodegenerative diseases. A major challenge for the translation of neuronal reprogramming into therapy is whether the adult human brain contains cell populations amenable to direct somatic cell conversion. Here we show that cells from the adult human cerebral cortex expressing pericyte hallmarks can be reprogrammed into neuronal cells by retrovirus-mediated coexpression of the transcription factors Sox2 and Mash1. These induced neuronal cells acquire the ability of repetitive action potential firing and serve as synaptic targets for other neurons, indicating their capability of integrating into neural networks. Genetic fate-mapping in mice expressing an inducible Cre recombinase under the tissue-nonspecific alkaline phosphatase promoter corroborated the pericytic origin of the reprogrammed cells. Our results raise the possibility of functional conversion of endogenous cells in the adult human brain to induced neuronal fates.


Asunto(s)
Reprogramación Celular , Corteza Cerebral/citología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Neurogénesis , Neuronas/citología , Pericitos/citología , Potenciales de Acción , Adulto , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Células Cultivadas , Humanos , Ratones , Red Nerviosa , Enfermedades Neurodegenerativas/terapia , Retroviridae , Factores de Transcripción SOXB1/metabolismo , Trasplante de Células Madre , Transmisión Sináptica
17.
Circulation ; 126(11 Suppl 1): S20-8, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22965984

RESUMEN

BACKGROUND: Human cardiac progenitor cells (hCPCs) are a promising cell source for regenerative repair after myocardial infarction. Exploitation of their full therapeutic potential may require stable genetic modification of the cells ex vivo. Safe genetic engineering of stem cells, using facile methods for site-specific integration of transgenes into known genomic contexts, would significantly enhance the overall safety and efficacy of cellular therapy in a variety of clinical contexts. METHODS AND RESULTS: We used the phiC31 site-specific recombinase to achieve targeted integration of a triple fusion reporter gene into a known chromosomal context in hCPCs and human endothelial cells. Stable expression of the reporter gene from its unique chromosomal integration site resulted in no discernible genomic instability or adverse changes in cell phenotype. Namely, phiC31-modified hCPCs were unchanged in their differentiation propensity, cellular proliferative rate, and global gene expression profile when compared with unaltered control hCPCs. Expression of the triple fusion reporter gene enabled multimodal assessment of cell fate in vitro and in vivo using fluorescence microscopy, bioluminescence imaging, and positron emission tomography. Intramyocardial transplantation of genetically modified hCPCs resulted in significant improvement in myocardial function 2 weeks after cell delivery, as assessed by echocardiography (P=0.002) and MRI (P=0.001). We also demonstrated the feasibility and therapeutic efficacy of genetically modifying differentiated human endothelial cells, which enhanced hind limb perfusion (P<0.05 at day 7 and 14 after transplantation) on laser Doppler imaging. CONCLUSIONS: The phiC31 integrase genomic modification system is a safe, efficient tool to enable site-specific integration of reporter transgenes in progenitor and differentiated cell types.


Asunto(s)
Células Madre Fetales/trasplante , Terapia Genética/métodos , Miembro Posterior/irrigación sanguínea , Isquemia/cirugía , Mutagénesis Insercional/métodos , Infarto del Miocardio/cirugía , Animales , Diferenciación Celular , División Celular , Cromosomas Humanos Par 19/genética , Células Endoteliales/citología , Femenino , Corazón Fetal/citología , Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Integrasas , Péptidos y Proteínas de Señalización Intracelular , Isquemia/fisiopatología , Luciferasas de Luciérnaga/genética , Proteínas Luminiscentes/genética , Imagen por Resonancia Magnética , Ratones , Ratones SCID , Proteínas/genética , Distribución Aleatoria , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Timidina Quinasa/genética , Transgenes , Proteínas de Transporte Vesicular , Proteínas Virales/genética , Integración Viral , Proteína Fluorescente Roja
18.
Int J Biochem Cell Biol ; 44(11): 1970-82, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22871567

RESUMEN

Human mesenchymal stem cells (hMSC) are subjected to the control of several signal transduction pathways during regeneration processes, whereby Wnt/ß-catenin signaling is of pivotal importance. Since there exists only fragmentary knowledge concerning the molecular function of the Wnt-coreceptors LRP5 and LRP6 (low-density lipoprotein receptor-related protein) in hMSC, we studied their impact on Wnt/ß-catenin signal transduction by RNA interference. For monitoring changes in ß-catenin-dependent transcription in a highly sensitive and specific manner, hMSC were stably transfected with a TCF/LEF reporter gene plasmid. In the presence of the activator Wnt3a, knockdown of LRP6 led to a strong decreased Wnt/ß-catenin signaling, while RNAi against LRP5 exhibited no effect in this setting. In a reverse approach, ectopic expression of LRP6 resulted in a strong enhancement of Wnt/ß-catenin signaling, whereas overexpression of LRP5 exhibited no increased signaling capacity. Furthermore, only the ectopic expression of LRP6--but not that of LRP5--was able to restore Wnt3a-mediated ß-catenin signaling after knockdown of endogenously expressed LRP6. These results demonstrate LRP6 as the predominant Wnt3a LRP-receptor in hMSC, which cannot be substituted by LRP5. In addition, we observed enhanced differentiation toward the adipogenic lineage after RNAi against LRP6 which was associated with the induction of PPAR-γ and fat vacuole formation. Thus, LRP6 is not only indispensable for Wnt3a/ß-catenin signaling, but also for the suppression of differentiation of hMSC into the adipogenic lineage. Based on these observations, LRP6 may represent an attractive drug target for manipulating hMSC in cell and tissue regeneration approaches.


Asunto(s)
Adipogénesis , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Vía de Señalización Wnt , Proteína Axina/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Luciferasas de Luciérnaga/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , PPAR gamma/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción TCF/metabolismo , Vacuolas/metabolismo , beta Catenina/metabolismo
19.
Stem Cells ; 29(11): 1696-704, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21898697

RESUMEN

Induced pluripotent stem cells (iPSCs) have revolutionized the stem cell field. iPSCs are most often produced by using retroviruses. However, the resulting cells may be ill-suited for clinical applications. Many alternative strategies to make iPSCs have been developed, but the nonintegrating strategies tend to be inefficient, while the integrating strategies involve random integration. Here, we report a facile strategy to create murine iPSCs that uses plasmid DNA and single transfection with sequence-specific recombinases. PhiC31 integrase was used to insert the reprogramming cassette into the genome, producing iPSCs. Cre recombinase was then used for excision of the reprogramming genes. The iPSCs were demonstrated to be pluripotent by in vitro and in vivo criteria, both before and after excision of the reprogramming cassette. This strategy is comparable with retroviral approaches in efficiency, but is nonhazardous for the user, simple to perform, and results in nonrandom integration of a reprogramming cassette that can be readily deleted. We demonstrated the efficiency of this reprogramming and excision strategy in two accessible cell types, fibroblasts and adipose stem cells. This simple strategy produces pluripotent stem cells that have the potential to be used in a clinical setting.


Asunto(s)
ADN Nucleotidiltransferasas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Plásmidos/genética , Tejido Adiposo/citología , Animales , Southern Blotting , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/fisiología , ADN Nucleotidiltransferasas/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Reacción en Cadena de la Polimerasa
20.
Biol Chem ; 392(11): 1011-20, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21864196

RESUMEN

WNT/Frizzled receptor (FZD) signaling pathways are pivotal for physiological and pathophysiological processes. In humans, the complexity of WNT/FZD signaling is based on 19 WNTs, 10 FZDs and at least two (co)receptors (LRP5/6) mediating supposably four different signaling cascades. The detailed investigation of the specific function of the different initiating components is primarily hampered by the lack of most WNT proteins in a purified form. Therefore, we constructed and examined a chimeric protein of WNT3a and FZD4 as a suitable approach to overcome this obstacle for future studies of the specificity of other WNT/FZD combinations. Furthermore, we produced four different reporter HEK 293 cell lines to quantify the induced activation of the proposed signaling cascades, the ß-catenin-, the NFAT-, the AP-1- and the CRE-regulated pathways. The chimera WNT3aFZD4 efficiently induced ß-catenin-mediated luciferase activity. This activity was increased 40-fold compared with basal when LRP6 was stably cotransfected, proving that the chimera WNT3aFZD4 can also interact efficiently with LRP6. Our results demonstrate that the approach of using reporter gene cell lines in combination with WNT/FZD chimeras is efficient to study the ß-catenin-mediated pathway and should also allow clarifying the specificity of WNT/FZD combinations in the activation of the other pathways.


Asunto(s)
Receptores Frizzled/genética , Genes Reporteros , Proteínas Recombinantes de Fusión/genética , Proteínas Wnt/genética , Vía de Señalización Wnt , Secuencia de Bases , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...