Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 161(3): 459-469, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25910206

RESUMEN

Mitochondrial diseases include a group of maternally inherited genetic disorders caused by mutations in mtDNA. In most of these patients, mutated mtDNA coexists with wild-type mtDNA, a situation known as mtDNA heteroplasmy. Here, we report on a strategy toward preventing germline transmission of mitochondrial diseases by inducing mtDNA heteroplasmy shift through the selective elimination of mutated mtDNA. As a proof of concept, we took advantage of NZB/BALB heteroplasmic mice, which contain two mtDNA haplotypes, BALB and NZB, and selectively prevented their germline transmission using either mitochondria-targeted restriction endonucleases or TALENs. In addition, we successfully reduced human mutated mtDNA levels responsible for Leber's hereditary optic neuropathy (LHOND), and neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP), in mammalian oocytes using mitochondria-targeted TALEN (mito-TALENs). Our approaches represent a potential therapeutic avenue for preventing the transgenerational transmission of human mitochondrial diseases caused by mutations in mtDNA. PAPERCLIP.


Asunto(s)
Marcación de Gen , Enfermedades Mitocondriales/genética , Animales , Fusión Celular , ADN Mitocondrial , Embrión de Mamíferos/metabolismo , Endonucleasas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NZB , Enfermedades Mitocondriales/prevención & control , Mutación , Oocitos/metabolismo
2.
Appl Microbiol Biotechnol ; 108(1): 409, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38970663

RESUMEN

Vitamin D deficiencies are linked to multiple human diseases. Optimizing its synthesis, physicochemical properties, and delivery systems while minimizing side effects is of clinical relevance and is of great medical and industrial interest. Biotechnological techniques may render new modified forms of vitamin D that may exhibit improved absorption, stability, or targeted physiological effects. Novel modified vitamin D derivatives hold promise for developing future therapeutic approaches and addressing specific health concerns related to vitamin D deficiency or impaired metabolism, such as avoiding hypercalcemic effects. Identifying and engineering key enzymes and biosynthetic pathways involved, as well as developing efficient cultures, are therefore of outmost importance and subject of intense research. Moreover, we elaborate on the critical role that microbial bioconversions might play in the a la carte design, synthesis, and production of novel, more efficient, and safer forms of vitamin D and its analogs. In summary, the novelty of this work resides in the detailed description of the physiological, medical, biochemical, and epidemiological aspects of vitamin D supplementation and the steps towards the enhanced and simplified industrial production of this family of bioactives relying on microbial enzymes. KEY POINTS: • Liver or kidney pathologies may hamper vitamin D biosynthesis • Actinomycetes are able to carry out 1α- or 25-hydroxylation on vitamin D precursors.


Asunto(s)
Biotransformación , Vitamina D , Vitamina D/metabolismo , Humanos , Vías Biosintéticas/genética , Ingeniería Metabólica/métodos , Actinobacteria/metabolismo , Actinobacteria/genética , Biotecnología/métodos , Bacterias/metabolismo , Bacterias/genética , Hidroxilación
4.
Immunity ; 32(2): 240-52, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20153221

RESUMEN

Injury to the central nervous system initiates an uncontrolled inflammatory response that results in both tissue repair and destruction. Here, we showed that, in rodents and humans, injury to the spinal cord triggered surface expression of CD95 ligand (CD95L, FasL) on peripheral blood myeloid cells. CD95L stimulation of CD95 on these cells activated phosphoinositide 3-kinase (PI3K) and metalloproteinase-9 (MMP-9) via recruitment and activation of Syk kinase, ultimately leading to increased migration. Exclusive CD95L deletion in myeloid cells greatly decreased the number of neutrophils and macrophages infiltrating the injured spinal cord or the inflamed peritoneum after thioglycollate injection. Importantly, deletion of myeloid CD95L, but not of CD95 on neural cells, led to functional recovery of spinal injured animals. Our results indicate that CD95L acts on peripheral myeloid cells to induce tissue damage. Thus, neutralization of CD95L should be considered as a means to create a controlled beneficial inflammatory response.


Asunto(s)
Movimiento Celular , Proteína Ligando Fas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Mieloides/metabolismo , Peritonitis/inmunología , Proteínas Tirosina Quinasas/metabolismo , Animales , Células Cultivadas , Proteína Ligando Fas/genética , Proteína Ligando Fas/inmunología , Humanos , Inflamación , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/patología , Peritoneo/inmunología , Peritoneo/patología , Peritonitis/inducido químicamente , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Médula Espinal/inmunología , Médula Espinal/patología , Quinasa Syk , Tioglicolatos/administración & dosificación
5.
Nature ; 491(7425): 603-7, 2012 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-23075850

RESUMEN

Nuclear-architecture defects have been shown to correlate with the manifestation of a number of human diseases as well as ageing. It is therefore plausible that diseases whose manifestations correlate with ageing might be connected to the appearance of nuclear aberrations over time. We decided to evaluate nuclear organization in the context of ageing-associated disorders by focusing on a leucine-rich repeat kinase 2 (LRRK2) dominant mutation (G2019S; glycine-to-serine substitution at amino acid 2019), which is associated with familial and sporadic Parkinson's disease as well as impairment of adult neurogenesis in mice. Here we report on the generation of induced pluripotent stem cells (iPSCs) derived from Parkinson's disease patients and the implications of LRRK2(G2019S) mutation in human neural-stem-cell (NSC) populations. Mutant NSCs showed increased susceptibility to proteasomal stress as well as passage-dependent deficiencies in nuclear-envelope organization, clonal expansion and neuronal differentiation. Disease phenotypes were rescued by targeted correction of the LRRK2(G2019S) mutation with its wild-type counterpart in Parkinson's disease iPSCs and were recapitulated after targeted knock-in of the LRRK2(G2019S) mutation in human embryonic stem cells. Analysis of human brain tissue showed nuclear-envelope impairment in clinically diagnosed Parkinson's disease patients. Together, our results identify the nucleus as a previously unknown cellular organelle in Parkinson's disease pathology and may help to open new avenues for Parkinson's disease diagnoses as well as for the potential development of therapeutics targeting this fundamental cell structure.


Asunto(s)
Proteínas Mutantes/metabolismo , Células-Madre Neurales/patología , Enfermedad de Parkinson/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Apoptosis , Diferenciación Celular , División Celular , Línea Celular , Células Clonales/metabolismo , Células Clonales/patología , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Técnicas de Sustitución del Gen , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Proteínas Mutantes/genética , Mutación , Células-Madre Neurales/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Estrés Fisiológico
6.
Nature ; 472(7342): 221-5, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21346760

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal human premature ageing disease, characterized by premature arteriosclerosis and degeneration of vascular smooth muscle cells (SMCs). HGPS is caused by a single point mutation in the lamin A (LMNA) gene, resulting in the generation of progerin, a truncated splicing mutant of lamin A. Accumulation of progerin leads to various ageing-associated nuclear defects including disorganization of nuclear lamina and loss of heterochromatin. Here we report the generation of induced pluripotent stem cells (iPSCs) from fibroblasts obtained from patients with HGPS. HGPS-iPSCs show absence of progerin, and more importantly, lack the nuclear envelope and epigenetic alterations normally associated with premature ageing. Upon differentiation of HGPS-iPSCs, progerin and its ageing-associated phenotypic consequences are restored. Specifically, directed differentiation of HGPS-iPSCs to SMCs leads to the appearance of premature senescence phenotypes associated with vascular ageing. Additionally, our studies identify DNA-dependent protein kinase catalytic subunit (DNAPKcs, also known as PRKDC) as a downstream target of progerin. The absence of nuclear DNAPK holoenzyme correlates with premature as well as physiological ageing. Because progerin also accumulates during physiological ageing, our results provide an in vitro iPSC-based model to study the pathogenesis of human premature and physiological vascular ageing.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Envejecimiento/metabolismo , Envejecimiento/patología , Envejecimiento/fisiología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Envejecimiento Prematuro/fisiopatología , Proteínas de Unión al Calcio/análisis , Diferenciación Celular , Línea Celular , Reprogramación Celular , Senescencia Celular , Proteína Quinasa Activada por ADN/metabolismo , Epigénesis Genética , Fibroblastos/patología , Holoenzimas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lamina Tipo A , Proteínas de Microfilamentos/análisis , Modelos Biológicos , Músculo Liso Vascular/patología , Membrana Nuclear/patología , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Progeria/genética , Progeria/patología , Progeria/fisiopatología , Precursores de Proteínas/análisis , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Especificidad por Sustrato , Calponinas
7.
Proc Natl Acad Sci U S A ; 111(14): 5076-82, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706886

RESUMEN

Induced pluripotent stem cells (iPSCs) are created by the reprogramming of somatic cells via overexpression of certain transcription factors, such as the originally described Yamanaka factors: Oct4, Sox2, Klf4, and c-Myc (OSKM). Here we discuss recent advancements in iPSC reprogramming and introduce mathematical approaches to help map the landscape between cell states during reprogramming. Our modelization indicates that OSKM expression diminishes and/or changes potential barriers between cell states and that epigenetic remodeling facilitate these transitions. From a practical perspective, the modeling approaches outlined here allow us to predict the time necessary to create a given number of iPSC colonies or the number of reprogrammed cells generated in a given time. Additional investigations will help to further refine modeling strategies, rendering them applicable toward the study of the development and stability of cancer cells or even other reprogramming processes such as lineage conversion. Ultimately, a quantitative understanding of cell state transitions might facilitate the establishment of regenerative medicine strategies and enhance the translation of reprogramming technologies into the clinic.


Asunto(s)
Diferenciación Celular , Modelos Biológicos , Células Madre Pluripotentes/citología , Factores de Transcripción/metabolismo , Humanos , Factor 4 Similar a Kruppel , Células Madre Pluripotentes/metabolismo
8.
Circulation ; 131(14): 1278-1290, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25739401

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs) have emerged as critical epigenetic regulators with important functions in development and disease. Here, we sought to identify and functionally characterize novel lncRNAs critical for vertebrate development. METHODS AND RESULTS: By relying on human pluripotent stem cell differentiation models, we investigated lncRNAs differentially regulated at key steps during human cardiovascular development with a special focus on vascular endothelial cells. RNA sequencing led to the generation of large data sets that serve as a gene expression roadmap highlighting gene expression changes during human pluripotent cell differentiation. Stage-specific analyses led to the identification of 3 previously uncharacterized lncRNAs, TERMINATOR, ALIEN, and PUNISHER, specifically expressed in undifferentiated pluripotent stem cells, cardiovascular progenitors, and differentiated endothelial cells, respectively. Functional characterization, including localization studies, dynamic expression analyses, epigenetic modification monitoring, and knockdown experiments in lower vertebrates, as well as murine embryos and human cells, confirmed a critical role for each lncRNA specific for each analyzed developmental stage. CONCLUSIONS: We have identified and functionally characterized 3 novel lncRNAs involved in vertebrate and human cardiovascular development, and we provide a comprehensive transcriptomic roadmap that sheds new light on the molecular mechanisms underlying human embryonic development, mesodermal commitment, and cardiovascular specification.


Asunto(s)
Sistema Cardiovascular/crecimiento & desarrollo , Células Endoteliales/química , Regulación del Desarrollo de la Expresión Génica/genética , Miocitos Cardíacos/química , Células Madre Pluripotentes/química , ARN Largo no Codificante/aislamiento & purificación , Vertebrados/genética , Animales , Sistema Cardiovascular/metabolismo , Diferenciación Celular , Linaje de la Célula , Mapeo Cromosómico , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Corazón Fetal/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Datos de Secuencia Molecular , Morfolinos/farmacocinética , Miocitos Cardíacos/citología , ARN Largo no Codificante/fisiología , Análisis de Secuencia de ARN , Transcriptoma , Vertebrados/crecimiento & desarrollo , Pez Cebra/embriología
9.
Biochem Biophys Res Commun ; 473(3): 693-7, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-26655812

RESUMEN

Despite the profound and rapid advancements in reprogramming technologies since the generation of the first induced pluripotent stem cells (iPSCs) in 2006[1], the molecular basics of the process and its implications are still not fully understood. Recent work has suggested that a subset of TFs, so called "Pioneer TFs", play an important role during the stochastic phase of iPSC reprogramming [2-6]. Pioneer TFs activities differ from conventional transcription factors in their mechanism of action. They bind directly to condensed chromatin and elicit a series of chromatin remodeling events that lead to opening of the chromatin. Chromatin decondensation by pioneer factors progressively occurs during cell division and in turn exposes specific gene promoters in the DNA to which TFs can now directly bind to promoters that are readily accessible[2, 6]. Here, we will summarize recent advancements on our understanding of the molecular mechanisms underlying reprogramming to iPSC as well as the implications that pioneer Transcription Factor activities might play during different lineage conversion processes.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Animales , Diferenciación Celular , Linaje de la Célula , Cromatina/química , Células Madre Embrionarias/citología , Epigénesis Genética , Regulación de la Expresión Génica , Humanos , Medicina Regenerativa/métodos , Factores de Transcripción/metabolismo
10.
Nat Methods ; 10(1): 77-83, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23202434

RESUMEN

Lineage conversion of one somatic cell type to another is an attractive approach for generating specific human cell types. Lineage conversion can be direct, in the absence of proliferation and multipotent progenitor generation, or indirect, by the generation of expandable multipotent progenitor states. We report the development of a reprogramming methodology in which cells transition through a plastic intermediate state, induced by brief exposure to reprogramming factors, followed by differentiation. We use this approach to convert human fibroblasts to mesodermal progenitor cells, including by non-integrative approaches. These progenitor cells demonstrated bipotent differentiation potential and could generate endothelial and smooth muscle lineages. Differentiated endothelial cells exhibited neo-angiogenesis and anastomosis in vivo. This methodology for indirect lineage conversion to angioblast-like cells adds to the armamentarium of reprogramming approaches aimed at the study and treatment of ischemic pathologies.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Reprogramación Celular , Endotelio Vascular/citología , Fibroblastos/citología , Miocitos del Músculo Liso/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Western Blotting , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
11.
Stem Cells ; 33(3): 713-25, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25385436

RESUMEN

c-Myc and phosphatidylinositol 3-OH kinase (PI3K) both participate in diverse cellular processes, including cell cycle control and tumorigenic transformation. They also contribute to preserving embryonic stem cell (ESC) characteristics. However, in spite of the vast knowledge, the molecular relationship between c-Myc and PI3K in ESCs is not known. Herein, we demonstrate that c-Myc and PI3K function cooperatively but independently to support ESC self-renewal when murine ESCs are cultured under conventional culture condition. Interestingly, culture of ESCs in 2i-condition including a GSK3ß and MEK inhibitor renders both PI3K and Myc signaling dispensable for the maintenance of pluripotent properties. These results suggest that the requirement for an oncogenic proliferation-dependent mechanism sustained by Myc and PI3K is context dependent and that the 2i-condition liberates ESCs from the dependence of this mechanism.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/biosíntesis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Sistema de Señalización de MAP Quinasas , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética
12.
Stem Cells ; 32(11): 2923-2938, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25175072

RESUMEN

Reprogramming technologies have emerged as a promising approach for future regenerative medicine. Here, we report on the establishment of a novel methodology allowing for the conversion of human fibroblasts into hematopoietic progenitor-like cells with macrophage differentiation potential. SOX2 overexpression in human fibroblasts, a gene found to be upregulated during hematopoietic reconstitution in mice, induced the rapid appearance of CD34+ cells with a concomitant upregulation of mesoderm-related markers. Profiling of cord blood hematopoietic progenitor cell populations identified miR-125b as a factor facilitating commitment of SOX2-generated CD34+ cells to immature hematopoietic-like progenitor cells with grafting potential. Further differentiation toward the monocytic lineage resulted in the appearance of CD14+ cells with functional phagocytic capacity. In vivo transplantation of SOX2/miR-125b-generated CD34+ cells facilitated the maturation of the engrafted cells toward CD45+ cells and ultimately the monocytic/macrophage lineage. Altogether, our results indicate that strategies combining lineage conversion and further lineage specification by in vivo or in vitro approaches could help to circumvent long-standing obstacles for the reprogramming of human cells into hematopoietic cells with clinical potential.


Asunto(s)
Diferenciación Celular/fisiología , Fibroblastos/citología , Monocitos/citología , Células Madre/citología , Animales , Antígenos CD34/metabolismo , Linaje de la Célula/fisiología , Células Cultivadas , Humanos , Antígenos Comunes de Leucocito/metabolismo , Ratones
14.
J Biol Chem ; 287(29): 24131-8, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22613719

RESUMEN

The tubular epithelium of the kidney is susceptible to injury from a number of different causes, including inflammatory and immune disorders, oxidative stress, and nephrotoxins, among others. Primary renal epithelial cells remain one of the few tools for studying the biochemical and physiological characteristics of the renal tubular system. Nevertheless, differentiated primary cells are not suitable for recapitulation of disease properties that might arise during embryonic kidney formation and further maturation. Thus, cellular systems resembling kidney characteristics are in urgent need to model disease as well as to establish reliable drug-testing platforms. Induced pluripotent stem cells (iPSCs) bear the capacity to differentiate into every cell lineage comprising the adult organism. Thus, iPSCs bring the possibility for recapitulating embryonic development by directed differentiation into specific lineages. iPSC differentiation ultimately allows for both disease modeling in vitro and the production of cellular products with potential for regenerative medicine. Here, we describe the rapid, reproducible, and highly efficient generation of iPSCs derived from endogenous kidney tubular renal epithelial cells with only two transcriptional factors, OCT4 and SOX2. Kidney-derived iPSCs may provide a reliable cellular platform for the development of kidney differentiation protocols allowing drug discovery studies and the study of kidney pathology.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Persona de Mediana Edad , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/genética
15.
Cell Rep ; 40(7): 111200, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35977506

RESUMEN

Apolipoprotein E4 (APOEε4) is the major allelic risk factor for late-onset sporadic Alzheimer's disease (sAD). Inflammation is increasingly considered as critical in sAD initiation and progression. Identifying brain molecular mechanisms that could bridge these two risk factors remain unelucidated. Leveraging induced pluripotent stem cell (iPSC)-based strategies, we demonstrate that APOE controls inflammation in human astrocytes by regulating Transgelin 3 (TAGLN3) expression and, ultimately, nuclear factor κB (NF-κB) activation. We uncover that APOE4 specifically downregulates TAGLN3, involving histone deacetylases activity, which results in low-grade chronic inflammation and hyperactivated inflammatory responses. We show that APOE4 exerts a dominant negative effect to prime astrocytes toward a pro-inflammatory state that is pharmacologically reversible by TAGLN3 supplementation. We further confirm that TAGLN3 is downregulated in the brain of patients with sAD. Our findings highlight the APOE-TAGLN3-NF-κB axis regulating neuroinflammation in human astrocytes and reveal TAGLN3 as a molecular target to modulate neuroinflammation, as well as a potential biomarker for AD.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Apolipoproteínas E/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Alzheimer/metabolismo , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Apolipoproteínas E/genética , Astrocitos/metabolismo , Humanos , Inflamación/metabolismo , FN-kappa B/metabolismo
17.
J Med Chem ; 63(8): 3868-3880, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31940200

RESUMEN

Farnesoid X receptor (FXR) agonists are emerging as important potential therapeutics for the treatment of nonalcoholic steatohepatitis (NASH) patients, as they exert positive effects on multiple aspects of the disease. FXR agonists reduce lipid accumulation in the liver, hepatocellular inflammation, hepatic injury, and fibrosis. While there are currently no approved therapies for NASH, the bile acid-derived FXR agonist obeticholic acid (OCA; 6-ethyl chenodeoxycholic acid) has shown promise in clinical studies. Previously, we described the discovery of tropifexor (LJN452), the most potent non-bile acid FXR agonist currently in clinical investigation. Here, we report the discovery of a novel chemical series of non-bile acid FXR agonists based on a tricyclic dihydrochromenopyrazole core from which emerged nidufexor (LMB763), a compound with partial FXR agonistic activity in vitro and FXR-dependent gene modulation in vivo. Nidufexor has advanced to Phase 2 human clinical trials in patients with NASH and diabetic nephropathy.


Asunto(s)
Benzotiazoles/uso terapéutico , Ácido Quenodesoxicólico/análogos & derivados , Dieta Alta en Grasa/efectos adversos , Isoxazoles/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Benzotiazoles/química , Ácido Quenodesoxicólico/química , Ácido Quenodesoxicólico/uso terapéutico , Perros , Humanos , Isoxazoles/química , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/etiología , Estructura Terciaria de Proteína , Ratas , Resultado del Tratamiento
18.
Protein Cell ; 10(7): 485-495, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31041783

RESUMEN

Identification of the precise molecular pathways involved in oncogene-induced transformation may help us gain a better understanding of tumor initiation and promotion. Here, we demonstrate that SOX2+ foregut epithelial cells are prone to oncogenic transformation upon mutagenic insults, such as KrasG12D and p53 deletion. GFP-based lineage-tracing experiments indicate that SOX2+ cells are the cells-of-origin of esophagus and stomach hyperplasia. Our observations indicate distinct roles for oncogenic KRAS mutation and P53 deletion. p53 homozygous deletion is required for the acquisition of an invasive potential, and KrasG12D expression, but not p53 deletion, suffices for tumor formation. Global gene expression analysis reveals secreting factors upregulated in the hyperplasia induced by oncogenic KRAS and highlights a crucial role for the CXCR2 pathway in driving hyperplasia. Collectively, the array of genetic models presented here demonstrate that stratified epithelial cells are susceptible to oncogenic insults, which may lead to a better understanding of tumor initiation and aid in the design of new cancer therapeutics.


Asunto(s)
Neoplasias Esofágicas/metabolismo , Mutación , Receptores de Interleucina-8B/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Proliferación Celular , Neoplasias Esofágicas/patología , Femenino , Masculino , Ratones , Ratones Mutantes , Transducción de Señal , Células Tumorales Cultivadas
19.
Science ; 356(6337): 503-508, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28473583

RESUMEN

CpG islands (CGIs) are primarily promoter-associated genomic regions and are mostly unmethylated within highly methylated mammalian genomes. The mechanisms by which CGIs are protected from de novo methylation remain elusive. Here we show that insertion of CpG-free DNA into targeted CGIs induces de novo methylation of the entire CGI in human pluripotent stem cells (PSCs). The methylation status is stably maintained even after CpG-free DNA removal, extensive passaging, and differentiation. By targeting the DNA mismatch repair gene MLH1 CGI, we could generate a PSC model of a cancer-related epimutation. Furthermore, we successfully corrected aberrant imprinting in induced PSCs derived from an Angelman syndrome patient. Our results provide insights into how CpG-free DNA induces de novo CGI methylation and broaden the application of targeted epigenome editing for a better understanding of human development and disease.


Asunto(s)
Islas de CpG , Metilación de ADN , Epigénesis Genética , Células Madre Pluripotentes/metabolismo , ADN/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Reparación del ADN/genética , Humanos , Homólogo 1 de la Proteína MutL/genética , Mutagénesis Insercional , Neuronas/metabolismo , Ubiquitina-Proteína Ligasas/genética
20.
Cell Cycle ; 15(18): 2393-7, 2016 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-27314153

RESUMEN

Cancer comprises heterogeneous cells, ranging from highly proliferative immature precursors to more differentiated cell lineages. The emergence of the "cancer stem cell" (CSC) hypothesis that they are the cells responsible for resistance, metastasis and secondary tumor appearance identifies these populations as novel obligatory targets for the treatment of cancer. CSCs, like their normal tissue-specific stem cell counterparts, are multipotent, partially differentiated, self-sustaining, yet transformed cells. To date, most studies on CSC biology have relied on the use of murine models and primary human material. In spite of much progress, the use of primary material presents several limitations that limit our understanding of the mechanisms underlying CSC formation, the similarities between normal stem cells and CSCs and ultimately, the possibility for developing targeted therapies. Recently, different strategies for controlling cell fate have been applied to the modeling of human cancer initiation and for the generation of human CSC models. Here we will summarize recent developments in the establishment and application of reprogramming strategies for the modeling of human cancer initiation and CSC formation.


Asunto(s)
Reprogramación Celular , Modelos Biológicos , Neoplasias/patología , Células Madre Neoplásicas/patología , Animales , Carcinogénesis/patología , Humanos , Mutación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA