Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24.012
Filtrar
Mais filtros

Coleção CLAP
Intervalo de ano de publicação
1.
Cell ; 187(9): 2143-2157.e15, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38670072

RESUMO

A central question for regenerative neuroscience is whether synthetic neural circuits, such as those built from two species, can function in an intact brain. Here, we apply blastocyst complementation to selectively build and test interspecies neural circuits. Despite approximately 10-20 million years of evolution, and prominent species differences in brain size, rat pluripotent stem cells injected into mouse blastocysts develop and persist throughout the mouse brain. Unexpectedly, the mouse niche reprograms the birth dates of rat neurons in the cortex and hippocampus, supporting rat-mouse synaptic activity. When mouse olfactory neurons are genetically silenced or killed, rat neurons restore information flow to odor processing circuits. Moreover, they rescue the primal behavior of food seeking, although less well than mouse neurons. By revealing that a mouse can sense the world using neurons from another species, we establish neural blastocyst complementation as a powerful tool to identify conserved mechanisms of brain development, plasticity, and repair.


Assuntos
Neurônios , Animais , Camundongos , Ratos , Neurônios/metabolismo , Neurônios/citologia , Neurônios/fisiologia , Blastocisto/metabolismo , Blastocisto/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Encéfalo/citologia , Encéfalo/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Especificidade da Espécie , Camundongos Endogâmicos C57BL , Masculino
2.
Cell ; 186(4): 715-731.e19, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36754048

RESUMO

Transgenerational epigenetic inheritance in mammals remains a debated subject. Here, we demonstrate that DNA methylation of promoter-associated CpG islands (CGIs) can be transmitted from parents to their offspring in mice. We generated DNA methylation-edited mouse embryonic stem cells (ESCs), in which CGIs of two metabolism-related genes, the Ankyrin repeat domain 26 and the low-density lipoprotein receptor, were specifically methylated and silenced. DNA methylation-edited mice generated by microinjection of the methylated ESCs exhibited abnormal metabolic phenotypes. Acquired methylation of the targeted CGI and the phenotypic traits were maintained and transmitted across multiple generations. The heritable CGI methylation was subjected to reprogramming in parental PGCs and subsequently reestablished in the next generation at post-implantation stages. These observations provide a concrete step toward demonstrating transgenerational epigenetic inheritance in mammals, which may have implications in our understanding of evolutionary biology as well as the etiology, diagnosis, and prevention of non-genetically inherited human diseases.


Assuntos
Metilação de DNA , Epigênese Genética , Camundongos , Humanos , Animais , Ilhas de CpG , Padrões de Herança , Mamíferos/genética
3.
Cell ; 186(2): 287-304.e26, 2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36610399

RESUMO

Whether and how certain transposable elements with viral origins, such as endogenous retroviruses (ERVs) dormant in our genomes, can become awakened and contribute to the aging process is largely unknown. In human senescent cells, we found that HERVK (HML-2), the most recently integrated human ERVs, are unlocked to transcribe viral genes and produce retrovirus-like particles (RVLPs). These HERVK RVLPs constitute a transmissible message to elicit senescence phenotypes in young cells, which can be blocked by neutralizing antibodies. The activation of ERVs was also observed in organs of aged primates and mice as well as in human tissues and serum from the elderly. Their repression alleviates cellular senescence and tissue degeneration and, to some extent, organismal aging. These findings indicate that the resurrection of ERVs is a hallmark and driving force of cellular senescence and tissue aging.


Assuntos
Envelhecimento , Retrovirus Endógenos , Idoso , Animais , Humanos , Camundongos , Envelhecimento/genética , Envelhecimento/patologia , Senescência Celular , Retrovirus Endógenos/genética , Primatas
4.
Cell ; 185(4): 581-584, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35180387

RESUMO

In a recent issue of Nature, Kagawa et al. reported a highly efficient and robust protocol for generating human blastoids from naive human pluripotent stem cells. The blastoids resemble human blastocysts, follow the sequential lineage specification of blastocyst development, and can attach to endometrial cells with the polar trophectoderm to model implantation.


Assuntos
Desenvolvimento Embrionário , Células-Tronco Pluripotentes , Blastocisto , Implantação do Embrião , Humanos
5.
Cell ; 184(8): 2020-2032.e14, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33861963

RESUMO

Interspecies chimera formation with human pluripotent stem cells (hPSCs) represents a necessary alternative to evaluate hPSC pluripotency in vivo and might constitute a promising strategy for various regenerative medicine applications, including the generation of organs and tissues for transplantation. Studies using mouse and pig embryos suggest that hPSCs do not robustly contribute to chimera formation in species evolutionarily distant to humans. We studied the chimeric competency of human extended pluripotent stem cells (hEPSCs) in cynomolgus monkey (Macaca fascicularis) embryos cultured ex vivo. We demonstrate that hEPSCs survived, proliferated, and generated several peri- and early post-implantation cell lineages inside monkey embryos. We also uncovered signaling events underlying interspecific crosstalk that may help shape the unique developmental trajectories of human and monkey cells within chimeric embryos. These results may help to better understand early human development and primate evolution and develop strategies to improve human chimerism in evolutionarily distant species.


Assuntos
Quimerismo , Embrião de Mamíferos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Macaca fascicularis , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante , RNA-Seq , Análise de Célula Única , Transcriptoma
6.
Cell ; 180(5): 984-1001.e22, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32109414

RESUMO

Aging causes a functional decline in tissues throughout the body that may be delayed by caloric restriction (CR). However, the cellular profiles and signatures of aging, as well as those ameliorated by CR, remain unclear. Here, we built comprehensive single-cell and single-nucleus transcriptomic atlases across various rat tissues undergoing aging and CR. CR attenuated aging-related changes in cell type composition, gene expression, and core transcriptional regulatory networks. Immune cells were increased during aging, and CR favorably reversed the aging-disturbed immune ecosystem. Computational prediction revealed that the abnormal cell-cell communication patterns observed during aging, including the excessive proinflammatory ligand-receptor interplay, were reversed by CR. Our work provides multi-tissue single-cell transcriptional landscapes associated with aging and CR in a mammal, enhances our understanding of the robustness of CR as a geroprotective intervention, and uncovers how metabolic intervention can act upon the immune system to modify the process of aging.


Assuntos
Envelhecimento/genética , Restrição Calórica , Sistema Imunitário/metabolismo , Transcriptoma/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Reprogramação Celular/genética , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Humanos , Ratos , Análise de Célula Única
7.
Cell ; 180(3): 585-600.e19, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32004457

RESUMO

Molecular mechanisms of ovarian aging and female age-related fertility decline remain unclear. We surveyed the single-cell transcriptomic landscape of ovaries from young and aged non-human primates (NHPs) and identified seven ovarian cell types with distinct gene-expression signatures, including oocyte and six types of ovarian somatic cells. In-depth dissection of gene-expression dynamics of oocytes revealed four subtypes at sequential and stepwise developmental stages. Further analysis of cell-type-specific aging-associated transcriptional changes uncovered the disturbance of antioxidant signaling specific to early-stage oocytes and granulosa cells, indicative of oxidative damage as a crucial factor in ovarian functional decline with age. Additionally, inactivated antioxidative pathways, increased reactive oxygen species, and apoptosis were observed in granulosa cells from aged women. This study provides a comprehensive understanding of the cell-type-specific mechanisms underlying primate ovarian aging at single-cell resolution, revealing new diagnostic biomarkers and potential therapeutic targets for age-related human ovarian disorders.


Assuntos
Envelhecimento/genética , Ovário/fisiologia , Análise de Célula Única/métodos , Transcriptoma , Idoso , Animais , Antioxidantes/metabolismo , Apoptose/fisiologia , Atlas como Assunto , Biomarcadores , Linhagem Celular Tumoral , Feminino , Células da Granulosa/metabolismo , Humanos , Macaca fascicularis , Oócitos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia
8.
Cell ; 179(3): 687-702.e18, 2019 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-31626770

RESUMO

A single mouse blastomere from an embryo until the 8-cell stage can generate an entire blastocyst. Whether laboratory-cultured cells retain a similar generative capacity remains unknown. Starting from a single stem cell type, extended pluripotent stem (EPS) cells, we established a 3D differentiation system that enabled the generation of blastocyst-like structures (EPS-blastoids) through lineage segregation and self-organization. EPS-blastoids resembled blastocysts in morphology and cell-lineage allocation and recapitulated key morphogenetic events during preimplantation and early postimplantation development in vitro. Upon transfer, some EPS-blastoids underwent implantation, induced decidualization, and generated live, albeit disorganized, tissues in utero. Single-cell and bulk RNA-sequencing analysis revealed that EPS-blastoids contained all three blastocyst cell lineages and shared transcriptional similarity with natural blastocysts. We also provide proof of concept that EPS-blastoids can be generated from adult cells via cellular reprogramming. EPS-blastoids provide a unique platform for studying early embryogenesis and pave the way to creating viable synthetic embryos by using cultured cells.


Assuntos
Blastocisto/citologia , Linhagem da Célula , Implantação do Embrião , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Embrionárias Murinas/citologia , Criação de Embriões para Pesquisa/métodos , Animais , Blastocisto/metabolismo , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Técnicas de Reprogramação Celular/métodos , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Células-Tronco Embrionárias Murinas/metabolismo , Transcriptoma
9.
Cell ; 176(4): 816-830.e18, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30595451

RESUMO

The temporal order of DNA replication (replication timing [RT]) is highly coupled with genome architecture, but cis-elements regulating either remain elusive. We created a series of CRISPR-mediated deletions and inversions of a pluripotency-associated topologically associating domain (TAD) in mouse ESCs. CTCF-associated domain boundaries were dispensable for RT. CTCF protein depletion weakened most TAD boundaries but had no effect on RT or A/B compartmentalization genome-wide. By contrast, deletion of three intra-TAD CTCF-independent 3D contact sites caused a domain-wide early-to-late RT shift, an A-to-B compartment switch, weakening of TAD architecture, and loss of transcription. The dispensability of TAD boundaries and the necessity of these "early replication control elements" (ERCEs) was validated by deletions and inversions at additional domains. Our results demonstrate that discrete cis-regulatory elements orchestrate domain-wide RT, A/B compartmentalization, TAD architecture, and transcription, revealing fundamental principles linking genome structure and function.


Assuntos
Período de Replicação do DNA/fisiologia , Replicação do DNA/genética , Replicação do DNA/fisiologia , Animais , Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Cromatina , DNA/genética , Período de Replicação do DNA/genética , Células-Tronco Embrionárias , Elementos Facilitadores Genéticos/genética , Mamíferos/genética , Mamíferos/metabolismo , Camundongos , Proteínas Repressoras/metabolismo , Análise Espaço-Temporal
10.
Nat Rev Mol Cell Biol ; 21(3): 137-150, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32020082

RESUMO

Ageing is characterized by the functional decline of tissues and organs and the increased risk of ageing-associated disorders. Several 'rejuvenating' interventions have been proposed to delay ageing and the onset of age-associated decline and disease to extend healthspan and lifespan. These interventions include metabolic manipulation, partial reprogramming, heterochronic parabiosis, pharmaceutical administration and senescent cell ablation. As the ageing process is associated with altered epigenetic mechanisms of gene regulation, such as DNA methylation, histone modification and chromatin remodelling, and non-coding RNAs, the manipulation of these mechanisms is central to the effectiveness of age-delaying interventions. This Review discusses the epigenetic changes that occur during ageing and the rapidly increasing knowledge of how these epigenetic mechanisms have an effect on healthspan and lifespan extension, and outlines questions to guide future research on interventions to rejuvenate the epigenome and delay ageing processes.


Assuntos
Envelhecimento/genética , Epigênese Genética/genética , Rejuvenescimento/fisiologia , Animais , Montagem e Desmontagem da Cromatina/genética , Metilação de DNA/genética , Epigenoma/genética , Epigenômica/métodos , Regulação da Expressão Gênica/genética , Código das Histonas/genética , Humanos , Longevidade/genética
11.
Cell ; 170(4): 599-600, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28802034

RESUMO

Aging and circadian rhythms have been linked for decades, but their molecular interplay has remained obscure. Sato et al. and Solanas et al. now reveal that, while core clock components remain nearly unaltered, aging is associated with tissue-specific rewiring, which can be prevented by calorie restriction.


Assuntos
Envelhecimento , Ritmo Circadiano , Restrição Calórica
12.
Cell ; 171(7): 1495-1507.e15, 2017 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-29224783

RESUMO

Current genome-editing systems generally rely on inducing DNA double-strand breaks (DSBs). This may limit their utility in clinical therapies, as unwanted mutations caused by DSBs can have deleterious effects. CRISPR/Cas9 system has recently been repurposed to enable target gene activation, allowing regulation of endogenous gene expression without creating DSBs. However, in vivo implementation of this gain-of-function system has proven difficult. Here, we report a robust system for in vivo activation of endogenous target genes through trans-epigenetic remodeling. The system relies on recruitment of Cas9 and transcriptional activation complexes to target loci by modified single guide RNAs. As proof-of-concept, we used this technology to treat mouse models of diabetes, muscular dystrophy, and acute kidney disease. Results demonstrate that CRISPR/Cas9-mediated target gene activation can be achieved in vivo, leading to measurable phenotypes and amelioration of disease symptoms. This establishes new avenues for developing targeted epigenetic therapies against human diseases. VIDEO ABSTRACT.


Assuntos
Sistemas CRISPR-Cas , Epigênese Genética , Marcação de Genes/métodos , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Utrofina/genética , Animais , Sequência de Bases , Modelos Animais de Doenças , Distrofina/genética , Interleucina-10/genética , Proteínas Klotho , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Ativação Transcricional
13.
Cell ; 169(2): 243-257.e25, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28388409

RESUMO

Of all known cultured stem cell types, pluripotent stem cells (PSCs) sit atop the landscape of developmental potency and are characterized by their ability to generate all cell types of an adult organism. However, PSCs show limited contribution to the extraembryonic placental tissues in vivo. Here, we show that a chemical cocktail enables the derivation of stem cells with unique functional and molecular features from mice and humans, designated as extended pluripotent stem (EPS) cells, which are capable of chimerizing both embryonic and extraembryonic tissues. Notably, a single mouse EPS cell shows widespread chimeric contribution to both embryonic and extraembryonic lineages in vivo and permits generating single-EPS-cell-derived mice by tetraploid complementation. Furthermore, human EPS cells exhibit interspecies chimeric competency in mouse conceptuses. Our findings constitute a first step toward capturing pluripotent stem cells with extraembryonic developmental potentials in culture and open new avenues for basic and translational research. VIDEO ABSTRACT.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes/citologia , Animais , Blastocisto/citologia , Linhagem Celular , Quimera/metabolismo , Dimetideno/farmacologia , Humanos , Indicadores e Reagentes/química , Camundongos , Minociclina/química , Minociclina/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo
14.
Cell ; 168(3): 473-486.e15, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28129541

RESUMO

Interspecies blastocyst complementation enables organ-specific enrichment of xenogenic pluripotent stem cell (PSC) derivatives. Here, we establish a versatile blastocyst complementation platform based on CRISPR-Cas9-mediated zygote genome editing and show enrichment of rat PSC-derivatives in several tissues of gene-edited organogenesis-disabled mice. Besides gaining insights into species evolution, embryogenesis, and human disease, interspecies blastocyst complementation might allow human organ generation in animals whose organ size, anatomy, and physiology are closer to humans. To date, however, whether human PSCs (hPSCs) can contribute to chimera formation in non-rodent species remains unknown. We systematically evaluate the chimeric competency of several types of hPSCs using a more diversified clade of mammals, the ungulates. We find that naïve hPSCs robustly engraft in both pig and cattle pre-implantation blastocysts but show limited contribution to post-implantation pig embryos. Instead, an intermediate hPSC type exhibits higher degree of chimerism and is able to generate differentiated progenies in post-implantation pig embryos.


Assuntos
Quimerismo , Edição de Genes , Mamíferos/embriologia , Animais , Blastocisto , Sistemas CRISPR-Cas , Bovinos , Embrião de Mamíferos/citologia , Feminino , Humanos , Masculino , Mamíferos/classificação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Células-Tronco Pluripotentes , Ratos , Ratos Sprague-Dawley , Sus scrofa
15.
Nat Immunol ; 20(11): 1456-1468, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31636466

RESUMO

T cell specification and commitment require Notch signaling. Although the requirement for Notch signaling during intrathymic T cell development is known, it is still unclear whether the onset of T cell priming can occur in a prethymic niche and whether RBPJ-dependent Notch signaling has a role during this event. Here, we established an Rbpj-inducible system that allowed temporal and tissue-specific control of the responsiveness to Notch in all hematopoietic cells. Using this system, we found that Notch signaling was required before the early T cell progenitor stage in the thymus. Lymphoid-primed multipotent progenitors in the bone marrow underwent Notch signaling with Rbpj induction, which inhibited development towards the myeloid lineage in thymus-seeding progenitors. Thus, our results indicated that the onset of T cell differentiation occurred in a prethymic setting, and that Notch played an important role during this event.


Assuntos
Diferenciação Celular/imunologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Células Precursoras de Linfócitos T/fisiologia , Receptores Notch/metabolismo , Subpopulações de Linfócitos T/imunologia , Animais , Linhagem da Célula/imunologia , Separação Celular , Feminino , Citometria de Fluxo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Masculino , Camundongos , Camundongos Transgênicos , Cultura Primária de Células , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/metabolismo , Timo/citologia , Timo/imunologia
16.
Nat Immunol ; 20(10): 1381-1392, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31451788

RESUMO

Proliferation is tightly regulated during T cell development, and is limited to immature CD4-CD8- thymocytes. The major proliferative event is initiated at the 'ß-selection' stage following successful rearrangement of Tcrß, and is triggered by and dependent on concurrent signaling by Notch and the pre-T cell receptor (TCR); however, it is unclear how these signals cooperate to promote cell proliferation. Here, we found that ß-selection-associated proliferation required the combined activity of two Skp-cullin-F-box (SCF) ubiquitin ligase complexes that included as substrate recognition subunits the F-box proteins Fbxl1 or Fbxl12. Both SCF complexes targeted the cyclin-dependent kinase inhibitor Cdkn1b for polyubiquitination and proteasomal degradation. We found that Notch signals induced the transcription of Fbxl1, whereas pre-TCR signals induced the transcription of Fbxl12. Thus, concurrent Notch and pre-TCR signaling induced the expression of two genes, Fbxl1 and Fbxl12, whose products functioned identically but additively to promote degradation of Cdkn1b, cell cycle progression, and proliferation of ß-selected thymocytes.


Assuntos
Proteínas F-Box/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores Notch/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Linfócitos T/fisiologia , Timócitos/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Seleção Clonal Mediada por Antígeno , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas F-Box/genética , Regulação da Expressão Gênica , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Camundongos , Camundongos Endogâmicos C57BL , Receptor Cross-Talk , Transdução de Sinais
17.
Cell ; 165(7): 1572-1585, 2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-27315475

RESUMO

The understanding of human biology and how it relates to that of other species represents an ancient quest. Limited access to human material, particularly during early development, has restricted researchers to only scratching the surface of this inherently challenging subject. Recent technological innovations, such as single cell "omics" and human stem cell derivation, have now greatly accelerated our ability to gain insights into uniquely human biology. The opportunities afforded to delve molecularly into scarce material and to model human embryogenesis and pathophysiological processes are leading to new insights of human development and are changing our understanding of disease and choice of therapy options.


Assuntos
Técnicas Citológicas , Desenvolvimento Embrionário , Pesquisa com Células-Tronco , Animais , Implantação do Embrião , Embrião de Mamíferos/metabolismo , Humanos , Células-Tronco Pluripotentes/metabolismo
18.
Cell ; 165(1): 13-15, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27015301

RESUMO

Random or not, the mechanisms at play during the first cell-fate determination in mammalian embryos have been debated for years. Studies by Goolam et al. and White et al. shed new light on the molecular mechanisms underlying the intrinsic biases that lead to non-random lineage segregation in early mammalian development.


Assuntos
Diferenciação Celular , Embrião de Mamíferos , Animais , Padronização Corporal , Linhagem da Célula
19.
Cell ; 166(6): 1371-1385, 2016 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-27610564

RESUMO

The discovery of induced pluripotent stem cells (iPSCs) a decade ago, which we are celebrating in this issue of Cell, represents a landmark discovery in biomedical research. Together with somatic cell nuclear transfer, iPSC generation reveals the remarkable plasticity associated with differentiated cells and provides an unprecedented means for modeling diseases using patient samples. In addition to transcriptional and epigenetic remodeling, cellular reprogramming to pluripotency is also accompanied by a rewiring of metabolic pathways, which ultimately leads to changes in cell identities.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Redes e Vias Metabólicas , Animais , Reprogramação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Doenças Metabólicas/metabolismo , Modelos Biológicos
20.
Cell ; 167(7): 1719-1733.e12, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984723

RESUMO

Aging is the major risk factor for many human diseases. In vitro studies have demonstrated that cellular reprogramming to pluripotency reverses cellular age, but alteration of the aging process through reprogramming has not been directly demonstrated in vivo. Here, we report that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in a mouse model of premature aging. Similarly, expression of OSKM in vivo improves recovery from metabolic disease and muscle injury in older wild-type mice. The amelioration of age-associated phenotypes by epigenetic remodeling during cellular reprogramming highlights the role of epigenetic dysregulation as a driver of mammalian aging. Establishing in vivo platforms to modulate age-associated epigenetic marks may provide further insights into the biology of aging.


Assuntos
Envelhecimento/genética , Reprogramação Celular , Epigênese Genética , Doenças Metabólicas/genética , Fatores de Transcrição/metabolismo , Senilidade Prematura/genética , Senilidade Prematura/metabolismo , Animais , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Lamina Tipo A/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/prevenção & controle , Camundongos , Modelos Animais , Pâncreas/metabolismo , Sarcopenia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA