Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 59(4): 496-516.e6, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38228141

RESUMEN

The details of how macrophages control different healing trajectories (regeneration vs. scar formation) remain poorly defined. Spiny mice (Acomys spp.) can regenerate external ear pinnae tissue, whereas lab mice (Mus musculus) form scar tissue in response to an identical injury. Here, we used this dual species system to dissect macrophage phenotypes between healing modes. We identified secreted factors from activated Acomys macrophages that induce a pro-regenerative phenotype in fibroblasts from both species. Transcriptional profiling of Acomys macrophages and subsequent in vitro tests identified VEGFC, PDGFA, and Lactotransferrin (LTF) as potential pro-regenerative modulators. Examining macrophages in vivo, we found that Acomys-resident macrophages secreted VEGFC and LTF, whereas Mus macrophages do not. Lastly, we demonstrate the requirement for VEGFC during regeneration and find that interrupting lymphangiogenesis delays blastema and new tissue formation. Together, our results demonstrate that cell-autonomous mechanisms govern how macrophages react to the same stimuli to differentially produce factors that facilitate regeneration.


Asunto(s)
Cicatriz , Pabellón Auricular , Animales , Cicatriz/patología , Lactoferrina , Pabellón Auricular/patología , Macrófagos/patología , Murinae/fisiología
2.
Science ; 381(6658): eabq5693, 2023 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-37561875

RESUMEN

Using DNA methylation profiles (n = 15,456) from 348 mammalian species, we constructed phyloepigenetic trees that bear marked similarities to traditional phylogenetic ones. Using unsupervised clustering across all samples, we identified 55 distinct cytosine modules, of which 30 are related to traits such as maximum life span, adult weight, age, sex, and human mortality risk. Maximum life span is associated with methylation levels in HOXL subclass homeobox genes and developmental processes and is potentially regulated by pluripotency transcription factors. The methylation state of some modules responds to perturbations such as caloric restriction, ablation of growth hormone receptors, consumption of high-fat diets, and expression of Yamanaka factors. This study reveals an intertwined evolution of the genome and epigenome that mediates the biological characteristics and traits of different mammalian species.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Mamíferos , Adulto , Animales , Humanos , Epigenoma , Genoma , Mamíferos/genética , Filogenia
3.
J Cell Sci ; 135(15)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35781573

RESUMEN

Adult stem cells persist in mammalian tissues by entering a state of reversible quiescence, referred to as G0, which is associated with low levels of transcription. Using cultured myoblasts and muscle stem cells, we report that in G0, global RNA content and synthesis are substantially repressed, correlating with decreased RNA polymerase II (RNAPII) expression and activation. Integrating RNAPII occupancy and transcriptome profiling, we identify repressed networks and a role for promoter-proximal RNAPII pausing in G0. Strikingly, RNAPII shows enhanced pausing in G0 on repressed genes encoding regulators of RNA biogenesis (such as Ncl, Rps24, Ctdp1), and release of pausing is associated with increased expression of these genes in G1. Knockdown of these transcripts in proliferating cells leads to induction of G0 markers, confirming the importance of their repression in establishment of G0. A targeted screen of RNAPII regulators revealed that knockdown of Aff4 (a positive regulator of elongation) unexpectedly enhances expression of G0-stalled genes and hastens S phase; however, the negative elongation factor (NELF) complex, a regulator of pausing, appears to be dispensable. We propose that RNAPII pausing contributes to transcriptional control of a subset of G0-repressed genes to maintain quiescence and impacts the timing of the G0-G1 transition. This article has an associated First Person interview with the first authors of the paper.


Asunto(s)
Regulación de la Expresión Génica , ARN Polimerasa II , Animales , Ciclo Celular/genética , Mamíferos/metabolismo , Regiones Promotoras Genéticas/genética , ARN , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Transcripción Genética , Factores de Elongación Transcripcional/genética
4.
Skelet Muscle ; 11(1): 18, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34238354

RESUMEN

BACKGROUND: During skeletal muscle regeneration, satellite stem cells use distinct pathways to repair damaged myofibers or to self-renew by returning to quiescence. Cellular/mitotic quiescence employs mechanisms that promote a poised or primed state, including altered RNA turnover and translational repression. Here, we investigate the role of mRNP granule proteins Fragile X Mental Retardation Protein (Fmrp) and Decapping protein 1a (Dcp1a) in muscle stem cell quiescence and differentiation. METHODS: Using isolated single muscle fibers from adult mice, we established differential enrichment of mRNP granule proteins including Fmrp and Dcp1a in muscle stem cells vs. myofibers. We investigated muscle tissue homeostasis in adult Fmr1-/- mice, analyzing myofiber cross-sectional area in vivo and satellite cell proliferation ex vivo. We explored the molecular mechanisms of Dcp1a and Fmrp function in quiescence, proliferation and differentiation in a C2C12 culture model. Here, we used polysome profiling, imaging and RNA/protein expression analysis to establish the abundance and assembly status of mRNP granule proteins in different cellular states, and the phenotype of knockdown cells. RESULTS: Quiescent muscle satellite cells are enriched for puncta containing the translational repressor Fmrp, but not the mRNA decay factor Dcp1a. MuSC isolated from Fmr1-/- mice exhibit defective proliferation, and mature myofibers show reduced cross-sectional area, suggesting a role for Fmrp in muscle homeostasis. Expression and organization of Fmrp and Dcp1a varies during primary MuSC activation on myofibers, with Fmrp puncta prominent in quiescence, but Dcp1a puncta appearing during activation/proliferation. This reciprocal expression of Fmrp and Dcp1a puncta is recapitulated in a C2C12 culture model of quiescence and activation: consistent with its role as a translational repressor, Fmrp is enriched in non-translating mRNP complexes abundant in quiescent myoblasts; Dcp1a puncta are lost in quiescence, suggesting stabilized and repressed transcripts. The function of each protein differs during proliferation; whereas Fmrp knockdown led to decreased proliferation and lower cyclin expression, Dcp1a knockdown led to increased cell proliferation and higher cyclin expression. However, knockdown of either Fmrp or Dcp1a led to compromised differentiation. We also observed cross-regulation of decay versus storage mRNP granules; knockdown of Fmrp enhances accumulation of Dcp1a puncta, whereas knockdown of Dcp1a leads to increased Fmrp in puncta. CONCLUSIONS: Taken together, our results provide evidence that the balance of mRNA turnover versus utilization is specific for distinct cellular states.


Asunto(s)
Endorribonucleasas/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Mioblastos , Ribonucleoproteínas , Células Madre/citología , Transactivadores/genética , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas , Mioblastos/citología
5.
J Mol Cell Cardiol ; 154: 60-69, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33556393

RESUMEN

Sinoatrial node cardiomyocytes (SANcm) possess automatic, rhythmic electrical activity. SAN rate is influenced by autonomic nervous system input, including sympathetic nerve increases of heart rate (HR) via activation of ß-adrenergic receptor signaling cascade (ß-AR). L-type calcium channel (LTCC) activity contributes to membrane depolarization and is a central target of ß-AR signaling. Recent studies revealed that the small G-protein Rad plays a central role in ß-adrenergic receptor directed modulation of LTCC. These studies have identified a conserved mechanism in which ß-AR stimulation results in PKA-dependent Rad phosphorylation: depletion of Rad from the LTCC complex, which is proposed to relieve the constitutive inhibition of CaV1.2 imposed by Rad association. Here, using a transgenic mouse model permitting conditional cardiomyocyte selective Rad ablation, we examine the contribution of Rad to the control of SANcm LTCC current (ICa,L) and sinus rhythm. Single cell analysis from a recent published database indicates that Rad is expressed in SANcm, and we show that SANcm ICa,L was significantly increased in dispersed SANcm following Rad silencing compared to those from CTRL hearts. Moreover, cRadKO SANcm ICa,L was not further increased with ß-AR agonists. We also evaluated heart rhythm in vivo using radiotelemetered ECG recordings in ambulating mice. In vivo, intrinsic HR is significantly elevated in cRadKO. During the sleep phase cRadKO also show elevated HR, and during the active phase there is no significant difference. Rad-deletion had no significant effect on heart rate variability. These results are consistent with Rad governing LTCC function under relatively low sympathetic drive conditions to contribute to slower HR during the diurnal sleep phase HR. In the absence of Rad, the tonic modulated SANcm ICa,L promotes elevated sinus HR. Future novel therapeutics for bradycardia targeting Rad - LTCC can thus elevate HR while retaining ßAR responsiveness.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Frecuencia Cardíaca , Activación del Canal Iónico , Proteínas de Unión al GTP Monoméricas/metabolismo , Miocardio/metabolismo , Animales , Canales de Calcio Tipo L/genética , Ratones , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/genética , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo
6.
Curr Opin Immunol ; 68: 72-82, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33171376

RESUMEN

Metabolism regulates an array of cellular processes from embryonic development through adulthood. These include proliferation, differentiation and the effector functions of adult cells to maintain homeostasis and repair. It is becoming clear that bioenergetic shifts can control how cells respond to environmental disruptions during tissue injury to initiate a healing response. Specifically, innate immune cells shift their phenotypes to initiate and resolve inflammation, and there is intense interest to understand how these responses might regulate healing outcomes. Here, we review recent literature describing how cellular metabolism and metabolic byproducts regulate phenotype conversions among innate immune cells. Although most studies of this kind do not focus on tissue damage, we discuss how metabolic regulation of these phenotypes promotes tissue repair. In particular, we provide a framework for considering the extent to which altering the innate immune response might shift fibrotic repair towards regenerative healing.


Asunto(s)
Inmunidad Innata/inmunología , Regeneración/inmunología , Cicatrización de Heridas/inmunología , Animales , Humanos , Fenotipo
7.
BMC Mol Cell Biol ; 21(1): 25, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32293249

RESUMEN

BACKGROUND: Reversible cell cycle arrest (quiescence/G0) is characteristic of adult stem cells and is actively controlled at multiple levels. Quiescent cells also extend a primary cilium, which functions as a signaling hub. Primary cilia have been shown to be important in multiple developmental processes, and are implicated in numerous developmental disorders. Although the association of the cilium with G0 is established, the role of the cilium in the control of the quiescence program is still poorly understood. RESULTS: Primary cilia are dynamically regulated across different states of cell cycle exit in skeletal muscle myoblasts: quiescent myoblasts elaborate a primary cilium in vivo and in vitro, but terminally differentiated myofibers do not. Myoblasts where ciliogenesis is ablated using RNAi against a key ciliary assembly protein (IFT88) can exit the cell cycle but display an altered quiescence program and impaired self-renewal. Specifically, the G0 transcriptome in IFT88 knockdown cells is aberrantly enriched for G2/M regulators, suggesting a focused repression of this network by the cilium. Cilium-ablated cells also exhibit features of activation including enhanced activity of Wnt and mitogen signaling and elevated protein synthesis via inactivation of the translational repressor 4E-BP1. CONCLUSIONS: Taken together, our results show that the primary cilium integrates and dampens proliferative signaling, represses translation and G2/M genes, and is integral to the establishment of the quiescence program.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Cilios/metabolismo , Redes Reguladoras de Genes , Mioblastos Esqueléticos/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Ciclo Celular/fisiología , Proteínas de Ciclo Celular , Diferenciación Celular , Línea Celular , Proliferación Celular , Centrosoma/metabolismo , Ratones , Transducción de Señal , Factores de Transcripción , Proteínas Supresoras de Tumor/metabolismo
8.
Sci Signal ; 11(540)2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30042129

RESUMEN

Skeletal muscle stem cells (MuSCs), also known as satellite cells, persist in adult mammals by entering a state of quiescence (G0) during the early postnatal period. Quiescence is reversed during damage-induced regeneration and re-established after regeneration. Entry of cultured myoblasts into G0 is associated with a specific, reversible induction of Wnt target genes, thus implicating members of the Tcf and Lef1 (Tcf/Lef) transcription factor family, which mediate transcriptional responses to Wnt signaling, in the initiation of quiescence. We found that the canonical Wnt effector ß-catenin, which cooperates with Tcf/Lef, was dispensable for myoblasts to enter quiescence. Using pharmacological and genetic approaches in cultured C2C12 myoblasts and in MuSCs, we demonstrated that Tcf/Lef activity during quiescence depended not on ß-catenin but on the transforming growth factor-ß (TGF-ß) effector and transcriptional coactivator Smad3, which colocalized with Lef1 at canonical Wnt-responsive elements and directly interacted with Lef1 specifically in G0 Depletion of Smad3, but not ß-catenin, reduced Lef1 occupancy at target promoters, Tcf/Lef target gene expression, and self-renewal of myoblasts. In vivo, MuSCs underwent a switch from ß-catenin-Lef1 to Smad3-Lef1 interactions during the postnatal switch from proliferation to quiescence, with ß-catenin-Lef1 interactions recurring during damage-induced reactivation. Our findings suggest that the interplay of Wnt-Tcf/Lef and TGF-ß-Smad3 signaling activates canonical Wnt target promoters in a manner that depends on ß-catenin during myoblast proliferation but is independent of ß-catenin during MuSC quiescence.


Asunto(s)
Factor de Unión 1 al Potenciador Linfoide/metabolismo , Células Satélite del Músculo Esquelético/citología , Proteína smad3/metabolismo , beta Catenina/metabolismo , Animales , Proliferación Celular , Autorrenovación de las Células , Células Cultivadas , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal , Proteína smad3/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...