Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
Curr Top Dev Biol ; 158: 407-431, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670714

RESUMEN

Skeletal muscle stem cells (MuSCs), also known as satellite cells, are essential for muscle growth and injury induced regeneration. In healthy adult muscle, MuSCs remain in a quiescent state located in a specialized niche beneath the basal lamina. Upon injury, these dormant MuSCs can quickly activate to re-enter the cell cycle and differentiate into new myofibers, while a subset undergoes self-renewal and returns to quiescence to restore the stem cell pool. The myogenic lineage progression is intricately controlled by complex intrinsic and extrinsic cues and coupled with dynamic transcriptional programs. In transcriptional regulation, enhancers are key regulatory elements controlling spatiotemporal gene expression through physical contacting promoters of target genes. The three-dimensional (3D) chromatin architecture is known to orchestrate the establishment of proper enhancer-promoter interactions throughout development and aging. However, studies dissecting the 3D organization of enhancers in MuSCs are just emerging. Here, we provide an overview of the general properties of enhancers and newly developed methods for assessing their activity. In particular, we summarize recent discoveries regarding the 3D rewiring of enhancers during MuSC specification, lineage progression as well as aging.


Asunto(s)
Elementos de Facilitación Genéticos , Animales , Humanos , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Desarrollo de Músculos/genética , Diferenciación Celular , Linaje de la Célula , Cromatina/metabolismo , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica
2.
Bioorg Chem ; 146: 107309, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38537338

RESUMEN

Prostate Cancer (PCa) easily progress to metastatic Castration-Resistant Prostate Cancer (mCRPC) that remains a significant cause of cancer-related death. Androgen receptor (AR)-dependent transcription is a major driver of prostate tumor cell proliferation. Proteolysis-targeting chimaera (PROTAC) technology based on Hydrophobic Tagging (HyT) represents an intriguing strategy to regulate the function of therapeutically androgen receptor proteins. In the present study, we have designed, synthesized, and evaluated a series of PROTAC-HyT AR degraders using AR antagonists, RU59063, which were connected with adamantane-based hydrophobic moieties by different alkyl chains. Compound D-4-6 exhibited significant AR protein degradation activity, with a degradation rate of 57 % at 5 µM and nearly 90 % at 20 µM in 24 h, and inhibited the proliferation of LNCaP cells significantly with an IC50 value of 4.77 ± 0.26 µM in a time-concentration-dependent manner. In conclusion, the present study lays the foundation for the development of a completely new class of therapeutic agents for the treatment of mCRPC, and further design and synthesis of AR-targeting degraders are currently in progress for better degradation rate.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Receptores Androgénicos , Masculino , Humanos , Receptores Androgénicos/química , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Línea Celular Tumoral , Antagonistas de Receptores Androgénicos/química , Antagonistas de Receptores Androgénicos/farmacología , Antagonistas de Receptores Androgénicos/uso terapéutico , Proteolisis
3.
J Med Chem ; 67(1): 138-151, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38153295

RESUMEN

Androgen receptor (AR) is the primary target for treating prostate cancer (PCa), which inevitably progresses due to drug-resistant mutations. Bromodomain-containing protein 4 (BRD4) has been a new potential drug target for PCa treatment. Herein, we report the rational design and discovery of novel BRD4 inhibitors through computer-aided drug design (CADD), and a hit compound SQ-1 (IC50 = 676 nM) was identified by structure-based virtual screening (SBVS) with the conserved water network. To optimize the structure of SQ-1, the free energy landscape was constructed, and the binding mechanism was explored by characterizing the water profile and the dissociation mechanism. Finally, the compound SQ-17 with improved inhibitory activity (IC50 < 100 nM) was discovered, which showed potent antiproliferative activity against LNCaP. These data highlighted a successful attempt to identify and optimize a small molecule by comprehensive CADD application and provided essential clues for developing novel therapeutics for PCa treatment.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata , Masculino , Humanos , Factores de Transcripción , Proteínas Nucleares , Agua/química , Detección Precoz del Cáncer , Diseño de Fármacos , Proteínas de Ciclo Celular/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Relación Estructura-Actividad , Antineoplásicos/química , Proteínas que Contienen Bromodominio
5.
Skelet Muscle ; 13(1): 15, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37705115

RESUMEN

Transcription factors (TFs) play key roles in regulating differentiation and function of stem cells, including muscle satellite cells (MuSCs), a resident stem cell population responsible for postnatal regeneration of the skeletal muscle. Sox11 belongs to the Sry-related HMG-box (SOX) family of TFs that play diverse roles in stem cell behavior and tissue specification. Analysis of single-cell RNA-sequencing (scRNA-seq) datasets identify a specific enrichment of Sox11 mRNA in differentiating but not quiescent MuSCs. Consistent with the scRNA-seq data, Sox11 levels increase during differentiation of murine primary myoblasts in vitro. scRNA-seq data comparing muscle regeneration in young and old mice further demonstrate that Sox11 expression is reduced in aged MuSCs. Age-related decline of Sox11 expression is associated with reduced chromatin contacts within the topologically associating domains. Unexpectedly, Myod1Cre-driven deletion of Sox11 in embryonic myoblasts has no effects on muscle development and growth, resulting in apparently healthy muscles that regenerate normally. Pax7CreER- or Rosa26CreER- driven (MuSC-specific or global) deletion of Sox11 in adult mice similarly has no effects on MuSC differentiation or muscle regeneration. These results identify Sox11 as a novel myogenic differentiation marker with reduced expression in quiescent and aged MuSCs, but the specific function of Sox11 in myogenesis remains to be elucidated.


Asunto(s)
Músculo Esquelético , Células Madre , Animales , Ratones , Diferenciación Celular , Desarrollo de Músculos , Regeneración , Factores de Transcripción SOXC/genética
6.
Nat Commun ; 14(1): 4978, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37591871

RESUMEN

Skeletal muscle stem cells (also called satellite cells, SCs) are important for maintaining muscle tissue homeostasis and damage-induced regeneration. However, it remains poorly understood how SCs enter cell cycle to become activated upon injury. Here we report that AP-1 family member ATF3 (Activating Transcription Factor 3) prevents SC premature activation. Atf3 is rapidly and transiently induced in SCs upon activation. Short-term deletion of Atf3 in SCs accelerates acute injury-induced regeneration, however, its long-term deletion exhausts the SC pool and thus impairs muscle regeneration. The Atf3 loss also provokes SC activation during voluntary exercise and enhances the activation during endurance exercise. Mechanistically, ATF3 directly activates the transcription of Histone 2B genes, whose reduction accelerates nucleosome displacement and gene transcription required for SC activation. Finally, the ATF3-dependent H2B expression also prevents genome instability and replicative senescence in SCs. Therefore, this study has revealed a previously unknown mechanism for preserving the SC population by actively suppressing precocious activation, in which ATF3 is a key regulator.


Asunto(s)
Factor de Transcripción Activador 3 , Fibras Musculares Esqueléticas , Factor de Transcripción Activador 3/genética , Ciclo Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Células Madre
7.
Environ Pollut ; 334: 122207, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37467914

RESUMEN

Molybdenum (Mo) is an essential trace element that exists in all tissues of the human body, but excessive Mo intake has a toxic effect. Cadmium (Cd) is a widely known and harmful heavy metal that exists in the environment. Although studies on Mo and Cd are available, it is still unknown how the combination of Mo and Cd causes pulmonary injury. Forty-eight sheep that were 2 months old were chosen and randomly separated into four groups as follows: Control group, Mo group, Cd group, and Mo + Cd group. The experiment lasted 50 days. The results showed that Mo and/or Cd caused significant pathological damage and oxidative stress in the lungs of sheep. Moreover, Mo and/or Cd exposure could downregulate the expression levels of xCT (SLC7A11 and SLC3A2), GPX4 and FTH-1 and upregulate the expression levels of PTGS2 and NCOA4, which led to iron overload and ferroptosis. Ferroptosis induced Wnt/ß-catenin-mediated fibrosis by elevating the expression levels of Caveolin-1 (CAV-1), Wnt 1, Wnt3a, ß-catenin (CTNNB1), TCF4, Cyclin D1, mmp7, α-SMA (ACTA2), Collagen 1 (COL1A1) and Vimentin. These changes were particularly noticeable in the Mo and Cd combination group. In conclusion, these data demonstrated that Mo and/or Cd exposure led to lung ferroptosis by inhibiting the SLC7A11/GSH/GPX4 axis, which in turn increases CAV-1 expression and subsequently activates the Wnt/ß-catenin pathway, leading to fibrosis in sheep lungs.


Asunto(s)
Ferroptosis , Molibdeno , Humanos , Animales , Ovinos , Lactante , Molibdeno/toxicidad , Cadmio/toxicidad , beta Catenina , Caveolina 1 , Fibrosis , Pulmón
8.
bioRxiv ; 2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37034612

RESUMEN

Transcription factors (TFs) play key roles in regulating the differentiation and function of stem cells, including muscle satellite cells (MuSCs), a resident stem cell population responsible for postnatal regeneration of the skeletal muscle. Sox11 belongs to the Sry-related HMG-box (SOX) family of TFs that play diverse roles in stem cell behavior and tissue specification. Analysis of single-cell RNA-sequencing (scRNA-seq) datasets identify a specific enrichment of Sox11 mRNA in differentiating but not quiescent MuSCs. Consistent with the scRNA-seq data, Sox11 levels increase during differentiation of murine primary myoblasts in vitro. scRNA-seq data comparing muscle regeneration in young and old mice further demonstrate that Sox11 expression is reduced in aged MuSCs. Age-related decline of Sox11 expression is associated with reduced chromatin contacts within the topologically associated domains. Unexpectedly, Myod1 Cre -driven deletion of Sox11 in embryonic myoblasts has no effects on muscle development and growth, resulting in apparently healthy muscles that regenerate normally. Pax7 CreER or Rosa26 CreER driven (MuSC-specific or global) deletion of Sox11 in adult mice similarly has no effects on MuSC differentiation or muscle regeneration. These results identify Sox11 as a novel myogenic differentiation marker with reduced expression in quiescent and aged MuSCs, but the specific function of Sox11 in myogenesis remain to be elucidated.

9.
Methods Mol Biol ; 2640: 287-311, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995603

RESUMEN

Skeletal muscle satellite cells (SCs) are adult stem cells responsible for muscle development and injury-induced muscle regeneration. Functional elucidation of intrinsic regulatory factors governing SC activity is constrained partially by the technological limitations in editing SCs in vivo. Although the power of CRISPR/Cas9 in genome manipulation has been widely documented, its application in endogenous SCs remains largely untested. Our recent study generates a muscle-specific genome editing system leveraging the Cre-dependent Cas9 knockin mice and AAV9-mediated sgRNAs delivery, which allows gene disruption in SCs in vivo. Here, we illustrate the step-by-step procedure for achieving efficient editing using the above system.


Asunto(s)
Edición Génica , Células Satélite del Músculo Esquelético , Ratones , Animales , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Músculos
10.
Elife ; 122023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36892464

RESUMEN

Skeletal muscle stem cells (also known as satellite cells [SCs]) are essential for muscle regeneration and the regenerative activities of SCs are intrinsically governed by gene regulatory mechanisms, but the post-transcriptional regulation in SCs remains largely unknown. N(6)-methyladenosine (m6A) modification of RNAs is the most pervasive and highly conserved RNA modification in eukaryotic cells; it exerts powerful impact on almost all aspects of mRNA processing that is mainly endowed by its binding with m6A reader proteins. In this study, we investigate the previously uncharacterized regulatory roles of YTHDC1, an m6A reader in mouse SCs. Our results demonstrate that YTHDC1 is an essential regulator of SC activation and proliferation upon acute injury-induced muscle regeneration. The induction of YTHDC1 is indispensable for SC activation and proliferation; thus, inducible YTHDC1 depletion almost abolishes SC regenerative capacity. Mechanistically, transcriptome-wide profiling using LACE-seq in both SCs and mouse C2C12 myoblasts identifies m6A-mediated binding targets of YTHDC1. Next, splicing analysis defines splicing mRNA targets of m6A-YTHDC1. Furthermore, nuclear export analysis also leads to the identification of potential mRNA export targets of m6A-YTHDC1 in SCs and C2C12 myoblasts;interestingly, some mRNAs can be regulated at both splicing and export levels. Lastly, we map YTHDC1 interacting protein partners in myoblasts and unveil a myriad of factors governing mRNA splicing, nuclear export, and transcription, among which hnRNPG appears to be a bona fide interacting partner of YTHDC1. Altogether, our findings uncover YTHDC1 as an essential factor controlling SC regenerative ability through multifaceted gene regulatory mechanisms in mouse myoblast cells.


Asunto(s)
Fibras Musculares Esqueléticas , Células Madre , Animales , Ratones , Transporte Activo de Núcleo Celular , Proliferación Celular , Fibras Musculares Esqueléticas/metabolismo , ARN Mensajero/metabolismo , Células Madre/metabolismo
11.
Sci Adv ; 9(7): eabo1360, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36800432

RESUMEN

Little is known about three-dimensional (3D) genome organization in skeletal muscle stem cells [also called satellite cells (SCs)]. Here, we comprehensively map the 3D genome topology reorganization during mouse SC lineage progression. Specifically, rewiring at the compartment level is most pronounced when SCs become activated. Marked loss in topologically associating domain (TAD) border insulation and chromatin looping also occurs during early activation process. Meanwhile, TADs can form TAD clusters and super-enhancer-containing TAD clusters orchestrate stage-specific gene expression. Furthermore, we uncover that transcription factor PAX7 is pivotal in enhancer-promoter (E-P) loop formation. We also identify cis-regulatory elements that are crucial for local chromatin organization at Pax7 locus and Pax7 expression. Lastly, we unveil that geriatric SC displays a prominent gain in long-range contacts and loss of TAD border insulation. Together, our results uncover that 3D chromatin extensively reorganizes at multiple architectural levels and underpins the transcriptome remodeling during SC lineage development and SC aging.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Animales , Ratones , Linaje de la Célula/genética , Cromatina/genética , Cromosomas , Músculo Esquelético
12.
Acta Pharmacol Sin ; 44(7): 1500-1518, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36639570

RESUMEN

As a major class of medicine for treating the lethal type of castration-resistant prostate cancer (PCa), long-term use of androgen receptor (AR) antagonists commonly leads to antiandrogen resistance. When AR signaling pathway is blocked by AR-targeted therapy, glucocorticoid receptor (GR) could compensate for AR function especially at the late stage of PCa. AR-GR dual antagonist is expected to be a good solution for this situation. Nevertheless, no effective non-steroidal AR-GR dual antagonist has been reported so far. In this study, an AR-GR dual binder H18 was first discovered by combining structure-based virtual screening and biological evaluation. Then with the aid of computationally guided design, the AR-GR dual antagonist HD57 was finally identified with antagonistic activity towards both AR (IC50 = 0.394 µM) and GR (IC50 = 17.81 µM). Moreover, HD57 could effectively antagonize various clinically relevant AR mutants. Further molecular dynamics simulation provided more atomic insights into the mode of action of HD57. Our research presents an efficient and rational strategy for discovering novel AR-GR dual antagonists, and the new scaffold provides important clues for the development of novel therapeutics for castration-resistant PCa.


Asunto(s)
Antagonistas de Andrógenos , Neoplasias de la Próstata , Masculino , Humanos , Antagonistas de Andrógenos/farmacología , Receptores de Glucocorticoides/metabolismo , Receptores Androgénicos/metabolismo , Antagonistas de Receptores Androgénicos/farmacología , Neoplasias de la Próstata/metabolismo , Línea Celular Tumoral
13.
Cell Mol Life Sci ; 80(1): 3, 2022 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36477660

RESUMEN

The pathological proliferation of cells in vascular smooth muscle underlies neointimal hyperplasia (NIH) development during atherosclerosis. Circular RNAs (circRNAs), which represent novel functional biomarkers and RNA-binding proteins, contribute to multiple cardiovascular diseases; however, their roles in regulating the vascular smooth muscle cell cycle remain unknown. Thus, we aimed to identify the roles of circRNAs in vascular smooth muscle during coronary heart disease (CHD). Through circRNA sequencing of CHD samples and human antigen R (ELAVL1) immunoprecipitation, we identified circRNAs that are associated with CHD and interact with ELAVL1. Our results suggested that the hsa_circ_0000280 associated with CHD inhibits cell proliferation and induces ELAVL1-dependent cell cycle arrest. Gain/loss-of-function experiments and assays in vivo indicated that hsa_circ_0000280 facilitates interactions between ELAVL1 and cyclin-dependent kinase suppressor 1 (CDKN1A) mRNA and stabilization of this complex and leads to cell cycle arrest at the G1/S checkpoint, inhibiting cell proliferation of vascular smooth muscle cells in vitro and NIH in vivo. Importantly, hsa_circ_0000280 reduced neointimal thickness and smooth muscle cell proliferation in vivo. Taken together, these findings reveal a novel pathway in which hsa_circ_0000280 facilitates the regulation of ELAVL1 on CDKN1A mRNA to inhibit NIH. Therefore, measuring and modulating their expression might represent a potential diagnostic or therapeutic strategy for CHD.


Asunto(s)
Músculo Liso Vascular , Miocitos del Músculo Liso , Humanos , Hiperplasia/genética , Proteína 1 Similar a ELAV/genética
14.
Aging Cell ; 21(8): e13673, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35851988

RESUMEN

Muscle satellite cells (SCs) are responsible for muscle homeostasis and regeneration and lncRNAs play important roles in regulating SC activities. Here, in this study, we identify PAM (Pax7 Associated Muscle lncRNA) that is induced in activated/proliferating SCs upon injury to promote SC proliferation as myoblast cells. PAM is generated from a myoblast-specific super-enhancer (SE); as a seRNA it binds with a number of target genomic loci predominantly in trans. Further studies demonstrate that it interacts with Ddx5 to tether PAM SE to its inter-chromosomal targets Timp2 and Vim to activate the gene expression. Lastly, we show that PAM expression is increased in aging SCs, which leads to enhanced inter-chromosomal interaction and target genes upregulation. Altogether, our findings identify PAM as a previously unknown lncRNA that regulates both SC proliferation and aging through its trans gene regulatory activity.


Asunto(s)
ARN Largo no Codificante , Células Satélite del Músculo Esquelético , Diferenciación Celular/genética , Proliferación Celular/genética , Músculo Esquelético/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células Satélite del Músculo Esquelético/metabolismo
15.
Cell Rep ; 39(9): 110884, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35649375

RESUMEN

Muscle regeneration is known to be defective under diabetic conditions. However, the underlying mechanisms remain less clear. Adult quiescent muscle satellite cells (MuSCs) from leptin-receptor-deficient (i.e., db/db) diabetic mice are defective in early activation in vivo, but not in culture, suggesting the involvement of pathogenic niche factors. Elevated extracellular adenosine (eAdo) and AMP (eAMP) are detected under diabetic conditions. eAdo and eAMP potently inhibit cell cycle re-entry of quiescent MuSCs and injury-induced muscle regeneration. Mechanistically, eAdo and eAMP engage the equilibrative Ado transporters (ENTs)-Ado kinase (ADK)-AMPK signaling axis in MuSCs to inhibit the mTORC1-dependent cell growth checkpoint. eAdo and eAMP also inhibit early activation of quiescent fibroadipogenic progenitors and human MuSCs by the same mechanism. Treatment of db/db diabetic mice with an ADK inhibitor partially rescues the activation defects of MuSCs in vivo. Thus, both ADK and ENTs represent potential therapeutic targets for restoring the regenerative functions of tissue stem cells in patients with diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Adenosina , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos , Músculos
16.
Cell Rep ; 39(10): 110927, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35675771

RESUMEN

Adult muscle stem cells, also known as satellite cells (SCs), play pivotal roles in muscle regeneration, and long non-coding RNA (lncRNA) functions in SCs remain largely unknown. Here, we identify a lncRNA, Lockd, which is induced in activated SCs upon acute muscle injury. We demonstrate that Lockd promotes SC proliferation; deletion of Lockd leads to cell-cycle arrest, and in vivo repression of Lockd in mouse muscles hinders regeneration process. Mechanistically, we show that Lockd directly interacts with RNA helicase DHX36 and the 5'end of Lockd possesses the strongest binding with DHX36. Furthermore, we demonstrate that Lockd stabilizes the interaction between DHX36 and EIF3B proteins; synergistically, this complex unwinds the RNA G-quadruplex (rG4) structure formed at Anp32e mRNA 5' UTR and promotes the translation of ANP32E protein, which is required for myoblast proliferation. Altogether, our findings identify a regulatory Lockd/DHX36/Anp32e axis that promotes myoblast proliferation and acute-injury-induced muscle regeneration.


Asunto(s)
ARN Helicasas DEAD-box , G-Cuádruplex , Chaperonas Moleculares , Desarrollo de Músculos , Mioblastos , ARN Largo no Codificante , Regiones no Traducidas 5' , Animales , Proliferación Celular , ARN Helicasas DEAD-box/metabolismo , Ratones , Chaperonas Moleculares/metabolismo , Músculos/metabolismo , Mioblastos/citología , ARN Largo no Codificante/metabolismo , Regeneración
17.
Nucleic Acids Res ; 50(6): 3323-3347, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35286396

RESUMEN

Aging in humans is intricately linked with alterations in circadian rhythms concomitant with physiological decline and stem cell exhaustion. However, whether the circadian machinery directly regulates stem cell aging, especially in primates, remains poorly understood. In this study, we found that deficiency of BMAL1, the only non-redundant circadian clock component, results in an accelerated aging phenotype in both human and cynomolgus monkey mesenchymal progenitor cells (MPCs). Unexpectedly, this phenotype was mainly attributed to a transcription-independent role of BMAL1 in stabilizing heterochromatin and thus preventing activation of the LINE1-cGAS-STING pathway. In senescent primate MPCs, we observed decreased capacity of BMAL1 to bind to LINE1 and synergistic activation of LINE1 expression. Likewise, in the skin and muscle tissues from the BMAL1-deficient cynomolgus monkey, we observed destabilized heterochromatin and aberrant LINE1 transcription. Altogether, these findings uncovered a noncanonical role of BMAL1 in stabilizing heterochromatin to inactivate LINE1 that drives aging in primate cells.


Asunto(s)
Factores de Transcripción ARNTL , Senescencia Celular , Relojes Circadianos , Macaca fascicularis/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Relojes Circadianos/genética , Ritmo Circadiano , Heterocromatina , Macaca fascicularis/genética
18.
Diabetes ; 71(5): 961-977, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35113157

RESUMEN

To date, identification of nonislet-specific transcriptional factors in the regulation of insulin gene expression has been little studied. Here, we report that the expression level of the transcription factor YY1 is increased dramatically in both human and mouse pancreatic ß-cells after birth. Nevertheless, the physiological role of YY1 during ß-cell development and its regulatory mechanism in ß-cell function remain largely unknown. After ß-cell ablation of Yy1, we observed rapid onset of hyperglycemia, impaired glucose tolerance, and reduced ß-cell mass in neonatal and adult mice. These mice also had hypoinsulinemia with normal insulin sensitivity compared with their wild-type littermates, manifesting as a type 1 diabetic phenotype. Mechanistically, genome-wide RNA sequencing has defined dysregulated insulin signaling and defective glucose responsiveness in ß-cells devoid of YY1. Integrative analyses coupled with chromatin immunoprecipitation assays targeting YY1, and histone modifications, including H3K4me1, H3K27ac, and H3K27me3, have further identified Ins1 and Ins2 as direct gene targets of YY1. Luciferase reporter assays and loss- and gain-of-function experiments also demonstrated that YY1 binds to the enhancer regions in exon 2 of Ins1 and Ins2, activating insulin transcription and, therefore, proinsulin and insulin production in pancreatic ß-cells. YY1 also directly interacts with RNA polymerase II, potentially stabilizing the enhancer-promoter interaction in the multiprotein-DNA complex during transcription initiation. Taken together, our findings suggest a role for YY1 as a transcriptional activator of insulin gene expression, assisting ß-cell maturation and function after birth. These analyses may advance our understanding of ß-cell biology and provide clinically relevant insights targeting the pathophysiological origins of diabetes.


Asunto(s)
Diabetes Mellitus , Células Secretoras de Insulina , Animales , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Insulina Regular Humana , Células Secretoras de Insulina/metabolismo , Ratones , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
19.
Stem Cell Reports ; 16(10): 2442-2458, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34534448

RESUMEN

Skeletal muscle satellite cells (SCs) are stem cells responsible for muscle development and regeneration. Although CRISPR/Cas9 has been widely used, its application in endogenous SCs remains elusive. Here, we generate mice expressing Cas9 in SCs and achieve robust editing in juvenile SCs at the postnatal stage through AAV9-mediated short guide RNA (sgRNA) delivery. Additionally, we reveal that quiescent SCs are resistant to CRISPR/Cas9-mediated editing. As a proof of concept, we demonstrate efficient editing of master transcription factor (TF) Myod1 locus using the CRISPR/Cas9/AAV9-sgRNA system in juvenile SCs. Application on two key TFs, MYC and BCL6, unveils distinct functions in SC activation and muscle regeneration. Particularly, we reveal that MYC orchestrates SC activation through regulating 3D genome architecture. Its depletion results in strengthening of the topologically associating domain boundaries thus may affect gene expression. Altogether, our study establishes a platform for editing endogenous SCs that can be harnessed to elucidate the functionality of key regulators governing SC activities.


Asunto(s)
Cromatina/metabolismo , Genes myc , Genoma , Proteína MioD/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Animales , Sistemas CRISPR-Cas , Edición Génica/métodos , Regulación de la Expresión Génica , Ratones , Proteína MioD/genética , Conformación de Ácido Nucleico , Proteínas Proto-Oncogénicas c-bcl-6/genética , ARN Guía de Kinetoplastida/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
J Cell Mol Med ; 25(20): 9660-9673, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34514716

RESUMEN

This study aimed to characterize the cells and gene expression landscape in atrial septal defect (ASD). We performed single-cell RNA sequencing of cells derived from cardiac tissue of an ASD patient. Unsupervised clustering analysis was performed to identify different cell populations, followed by the investigation of the cellular crosstalk by analysing ligand-receptor interactions across cell types. Finally, differences between ASD and normal samples for all cell types were further investigated. An expression matrix of 18,411 genes in 6487 cells was obtained and used in this analysis. Five cell types, including cardiomyocytes, endothelial cells, smooth muscle cells, fibroblasts and macrophages were identified. ASD showed a decreased proportion of cardiomyocytes and an increased proportion of fibroblasts. There was more cellular crosstalk among cardiomyocytes, fibroblasts and macrophages, especially between fibroblast and macrophage. For all cell types, the majority of the DEGs were downregulated in ASD samples. For cardiomyocytes, there were 199 DEGs (42 upregulated and 157 downregulated) between ASD and normal samples. PPI analysis showed that cardiomyocyte marker gene FABP4 interacted with FOS, while FOS showed interaction with NPPA. Cell trajectory analysis showed that FABP4, FOS, and NPPA showed different expression changes along the pseudotime trajectory. Our results showed that single-cell RNA sequencing provides a powerful tool to study DEG profiles in the cell subpopulations of interest at the single-cell level. These findings enhance the understanding of the underlying mechanisms of ASD at both the cellular and molecular level and highlight potential targets for the treatment of ASD.


Asunto(s)
Perfilación de la Expresión Génica , Defectos del Tabique Interatrial/genética , RNA-Seq , Análisis de la Célula Individual , Transcriptoma , Cadáver , Comunicación Celular , Línea Celular , Biología Computacional/métodos , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Lactante , Masculino , Anotación de Secuencia Molecular , Miocitos Cardíacos/metabolismo , Especificidad de Órganos , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , RNA-Seq/métodos , Análisis de la Célula Individual/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA