Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 112, 2024 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-38433139

RESUMEN

Down syndrome (DS) arises from a genetic anomaly characterized by an extra copy of chromosome 21 (exCh21). Despite high incidence of congenital diseases among DS patients, direct impacts of exCh21 remain elusive. Here, we established a robust DS model harnessing human-induced pluripotent stem cells (hiPSCs) from mosaic DS patient. These hiPSC lines encompassed both those with standard karyotype and those carrying an extra copy of exCh21, allowing to generate isogenic cell lines with a consistent genetic background. We unraveled that exCh21 inflicted disruption upon the cellular transcriptome, ushering in alterations in metabolic processes and triggering DNA damage. The impact of exCh21 was also manifested in profound modifications in chromatin accessibility patterns. Moreover, we identified two signature metabolites, 5-oxo-ETE and Calcitriol, whose biosynthesis is affected by exCh21. Notably, supplementation with 5-oxo-ETE promoted DNA damage, in stark contrast to the protective effect elicited by Calcitriol against such damage. We also found that exCh21 disrupted cardiogenesis, and that this impairment could be mitigated through supplementation with Calcitriol. Specifically, the deleterious effects of 5-oxo-ETE unfolded in the form of DNA damage induction and the repression of cardiogenesis. On the other hand, Calcitriol emerged as a potent activator of its nuclear receptor VDR, fostering amplified binding to chromatin and subsequent facilitation of gene transcription. Our findings provide a comprehensive understanding of exCh21's metabolic implications within the context of Down syndrome, offering potential avenues for therapeutic interventions for Down syndrome treatment.


Asunto(s)
Síndrome de Down , Humanos , Síndrome de Down/genética , Calcitriol/farmacología , Cromatina , Línea Celular , Daño del ADN
2.
Development ; 148(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34027990

RESUMEN

Polycomb repressive complex 2 (PRC2) deposits H3K27me3 on chromatin to silence transcription. PRC2 broadly interacts with RNAs. Currently, the role of the RNA-PRC2 interaction in human cardiogenesis remains elusive. Here, we found that human-specific heart brake lncRNA 1 (HBL1) interacted with two PRC2 subunits, JARID2 and EED, in human pluripotent stem cells (hPSCs). Loss of JARID2, EED or HBL1 significantly enhanced cardiac differentiation from hPSCs. HBL1 depletion disrupted genome-wide PRC2 occupancy and H3K27me3 chromatin modification on essential cardiogenic genes, and broadly enhanced cardiogenic gene transcription in undifferentiated hPSCs and later-on differentiation. In addition, ChIP-seq revealed reduced EED occupancy on 62 overlapped cardiogenic genes in HBL1-/- and JARID2-/- hPSCs, indicating that the epigenetic state of cardiogenic genes was determined by HBL1 and JARID2 at pluripotency stage. Furthermore, after cardiac development occurs, the cytosolic and nuclear fractions of HBL1 could crosstalk via a conserved 'microRNA-1-JARID2' axis to modulate cardiogenic gene transcription. Overall, our findings delineate the indispensable role of HBL1 in guiding PRC2 function during early human cardiogenesis, and expand the mechanistic scope of lncRNA(s) that cytosolic and nuclear portions of HBL1 could coordinate to orchestrate human cardiogenesis.


Asunto(s)
Genoma , Organogénesis , Células Madre Pluripotentes/metabolismo , Complejo Represivo Polycomb 2/genética , ARN Largo no Codificante/metabolismo , Diferenciación Celular , Cromatina , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Histonas/genética , Humanos , MicroARNs
3.
Circulation ; 145(16): 1238-1253, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35384713

RESUMEN

BACKGROUND: Familial hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disease and is typically caused by mutations in genes encoding sarcomeric proteins that regulate cardiac contractility. HCM manifestations include left ventricular hypertrophy and heart failure, arrythmias, and sudden cardiac death. How dysregulated sarcomeric force production is sensed and leads to pathological remodeling remains poorly understood in HCM, thereby inhibiting the efficient development of new therapeutics. METHODS: Our discovery was based on insights from a severe phenotype of an individual with HCM and a second genetic alteration in a sarcomeric mechanosensing protein. We derived cardiomyocytes from patient-specific induced pluripotent stem cells and developed robust engineered heart tissues by seeding induced pluripotent stem cell-derived cardiomyocytes into a laser-cut scaffold possessing native cardiac fiber alignment to study human cardiac mechanobiology at both the cellular and tissue levels. Coupled with computational modeling for muscle contraction and rescue of disease phenotype by gene editing and pharmacological interventions, we have identified a new mechanotransduction pathway in HCM, shown to be essential in modulating the phenotypic expression of HCM in 5 families bearing distinct sarcomeric mutations. RESULTS: Enhanced actomyosin crossbridge formation caused by sarcomeric mutations in cardiac myosin heavy chain (MYH7) led to increased force generation, which, when coupled with slower twitch relaxation, destabilized the MLP (muscle LIM protein) stretch-sensing complex at the Z-disc. Subsequent reduction in the sarcomeric muscle LIM protein level caused disinhibition of calcineurin-nuclear factor of activated T-cells signaling, which promoted cardiac hypertrophy. We demonstrate that the common muscle LIM protein-W4R variant is an important modifier, exacerbating the phenotypic expression of HCM, but alone may not be a disease-causing mutation. By mitigating enhanced actomyosin crossbridge formation through either genetic or pharmacological means, we alleviated stress at the Z-disc, preventing the development of hypertrophy associated with sarcomeric mutations. CONCLUSIONS: Our studies have uncovered a novel biomechanical mechanism through which dysregulated sarcomeric force production is sensed and leads to pathological signaling, remodeling, and hypertrophic responses. Together, these establish the foundation for developing innovative mechanism-based treatments for HCM that stabilize the Z-disc MLP-mechanosensory complex.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar , Cardiomiopatía Hipertrófica , Actomiosina/genética , Humanos , Proteínas con Dominio LIM , Mecanotransducción Celular , Proteínas Musculares , Mutación , Miocitos Cardíacos
4.
Nucleic Acids Res ; 49(D1): D706-D714, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33045727

RESUMEN

The COVID-19 outbreak has become a global emergency since December 2019. Analysis of SARS-CoV-2 sequences can uncover single nucleotide variants (SNVs) and corresponding evolution patterns. The Global Evaluation of SARS-CoV-2/hCoV-19 Sequences (GESS, https://wan-bioinfo.shinyapps.io/GESS/) is a resource to provide comprehensive analysis results based on tens of thousands of high-coverage and high-quality SARS-CoV-2 complete genomes. The database allows user to browse, search and download SNVs at any individual or multiple SARS-CoV-2 genomic positions, or within a chosen genomic region or protein, or in certain country/area of interest. GESS reveals geographical distributions of SNVs around the world and across the states of USA, while exhibiting time-dependent patterns for SNV occurrences which reflect development of SARS-CoV-2 genomes. For each month, the top 100 SNVs that were firstly identified world-widely can be retrieved. GESS also explores SNVs occurring simultaneously with specific SNVs of user's interests. Furthermore, the database can be of great help to calibrate mutation rates and identify conserved genome regions. Taken together, GESS is a powerful resource and tool to monitor SARS-CoV-2 migration and evolution according to featured genomic variations. It provides potential directive information for prevalence prediction, related public health policy making, and vaccine designs.


Asunto(s)
COVID-19/prevención & control , Biología Computacional/métodos , Bases de Datos Genéticas , Genoma Viral/genética , Genómica/métodos , SARS-CoV-2/genética , Algoritmos , COVID-19/epidemiología , COVID-19/virología , Brotes de Enfermedades , Salud Global , Humanos , Internet , Tasa de Mutación , Polimorfismo de Nucleótido Simple , Dinámica Poblacional , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo
5.
J Med Virol ; 93(12): 6525-6534, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34245452

RESUMEN

By analyzing newly collected SARS-CoV-2 genomes and comparing them with our previous study about SARS-CoV-2 single nucleotide variants (SNVs) before June 2020, we found that the SNV clustering had changed remarkably since June 2020. Apart from that the group of SNVs became dominant, which is represented by two nonsynonymous mutations A23403G (S:D614G) and C14408T (ORF1ab:P4715L), a few emerging groups of SNVs were recognized with sharply increased monthly incidence ratios of up to 70% in November 2020. Further investigation revealed sets of SNVs specific to patients' ages and/or gender, or strongly associated with mortality. Our logistic regression model explored features contributing to mortality status, including three critical SNVs, G25088T(S:V1176F), T27484C (ORF7a:L31L), and T25A (upstream of ORF1ab), ages above 40 years old, and the male gender. The protein structure analysis indicated that the emerging subgroups of nonsynonymous SNVs and the mortality-related ones were located on the protein surface area. The clashes in protein structure introduced by these mutations might in turn affect the viral pathogenesis through the alteration of protein conformation, leading to a difference in transmission and virulence. Particularly, we explored the fact that nonsynonymous SNVs tended to occur in intrinsic disordered regions of Spike and ORF1ab to significantly increase hydrophobicity, suggesting a potential role in the change of protein folding related to immune evasion.


Asunto(s)
COVID-19/mortalidad , Genoma Viral/genética , Polimorfismo de Nucleótido Simple/genética , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Poliproteínas/genética , Glicoproteína de la Espiga del Coronavirus/genética , Proteínas Virales/genética , Virulencia/genética , Adulto Joven
6.
Biochem Biophys Res Commun ; 502(4): 442-449, 2018 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-29807013

RESUMEN

Neural conversion from human pluripotent cells (hPSCs) is a potential therapy to neurological disease in the future. However, this is still limited by efficiency and stability of existed protocols used for neural induction from hPSCs. To overcome this obstacle, we developed a reporter system to screen PAX6+ neural progenitor/stem cells using transcription activator like effector nuclease (TALEN). We found that knock-in 2 A-EGFP cassette into PAX6 exon of human embryonic stem cells H1 with TALEN-based homology recombination could establish PAX6WT/EGFP H1 reporter cell line fast and efficiently. This reporter cell line could differentiate into PAX6 and EGFP double positive neural progenitor/stem cells (NPCs/NSCs) after neural induction. Those PAX6WT/EGFP NPCs could be purified, expanded and specified to post-mitotic neurons in vitro efficiently. With this reporter cell line, we also screened out 1 NPC-specific microRNA, hsa-miR-99a-5p, and 3 ESCs-enriched miRNAs, hsa-miR-302c-5p, hsa-miR-512-3p and hsa-miR-518 b. In conclusion, the TALEN-based neural stem cell screening system is safe and efficient and could help researcher to acquire adequate and pure neural progenitor cells for further application.


Asunto(s)
Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factor de Transcripción PAX6/metabolismo , Animales , Diferenciación Celular , Técnicas de Sustitución del Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , MicroARNs/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Factor de Transcripción PAX6/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo
7.
FASEB J ; 28(11): 4642-56, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25114173

RESUMEN

Differentiation of neural lineages from human pluripotent stem cells (hPSCs) raises the hope of generating functional cells for the treatment of neural diseases. However, current protocols for differentiating hPSCs into neural lineages remain inefficient and largely variable between different hPSC lines. We report that microRNA 376c (miR-376c) significantly enhanced neural differentiation of hPSCs in a defined condition by suppressing SMAD4, the co-SMAD for TGF-ß signaling. Downstream, SMAD4 directly bound and suppressed PAX6, the critical neural lineage specification factor. Interestingly, we also found that SMAD4 binds and suppresses miR-376c clusters in undifferentiated hESCs. In summary, our findings revealed a reciprocal antagonism between miR-376c and SMAD signaling that regulates cell fate during human neural differentiation.


Asunto(s)
Diferenciación Celular , MicroARNs/metabolismo , Células Madre Pluripotentes/citología , Transducción de Señal , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Silenciamiento del Gen/métodos , Humanos , Células Madre Pluripotentes/metabolismo , Transducción de Señal/fisiología
8.
Cell Death Dis ; 15(8): 607, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39168969

RESUMEN

Monkeypox virus (MPV) is known to inflict injuries and, in some cases, lead to fatalities in humans. However, the underlying mechanisms responsible for its pathogenicity remain poorly understood. We investigated functions of MPV core proteins, H3L, A35R, A29L, and I1L, and discovered that H3L induced transcriptional perturbations and injuries. We substantiated that H3L upregulated IL1A expression. IL1A, in consequence, caused cellular injuries, and this detrimental effect was mitigated when countered with IL1A blockage. We also observed that H3L significantly perturbed the transcriptions of genes in cardiac system. Mechanistically, H3L occupied the promoters of genes governing cellular injury, leading to alterations in the binding patterns of H3K27me3 and H3K4me3 histone marks, ultimately resulting in expression perturbations. In vivo and in vitro models confirmed that H3L induced transcriptional disturbances and cardiac dysfunction, which were ameliorated when IL1A was blocked or repressed. Our study provides valuable insights into comprehensive understanding of MPV pathogenicity, highlights the significant roles of H3L in inducing injuries, and potentially paves the way for the development of therapeutic strategies targeting IL1A.


Asunto(s)
Proteínas Virales , Animales , Humanos , Ratones , Proteínas Virales/metabolismo , Proteínas Virales/genética , Histonas/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1alfa/genética , Ratones Endogámicos C57BL
9.
Theranostics ; 14(12): 4894-4915, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39239522

RESUMEN

Rationale: Regulatory processes of transcription factors (TFs) shape heart development and influence the adult heart's response to stress, contributing to cardiac disorders. Despite their significance, the precise mechanisms underpinning TF-mediated regulation remain elusive. Here, we identify that EBF1, as a TF, is highly expressed in human heart tissues. EBF1 is reported to be associated with human cardiovascular disease, but its roles are unclear in heart. In this study, we investigated EBF1 function in cardiac system. Methods: RNA-seq was utilized to profile EBF1 expression patterns. CRISPR/Cas9 was utilized to knock out EBF1 to investigate its effects. Human pluripotent stem cells (hPSCs) differentiated into cardiac lineages were used to mimic cardiac development. Cardiac function was evaluated on mouse model with Ebf1 knockout by using techniques such as echocardiography. RNA-seq was conducted to analyze transcriptional perturbations. ChIP-seq was employed to elucidate EBF1-bound genes and the underlying regulatory mechanisms. Results: EBF1 was expressed in some human and mouse cardiomyocyte. Knockout of EBF1 inhibited cardiac development. ChIP-seq indicated EBF1's binding on promoters of cardiogenic TFs pivotal to cardiac development, facilitating their transcriptional expression and promoting cardiac development. In mouse, Ebf1 depletion triggered transcriptional perturbations of genes, resulting in cardiac remodeling. Mechanistically, we found that EBF1 directly bound to upstream chromatin regions of cardiac hypertrophy-inducing genes, contributing to cardiac hypertrophy. Conclusions: We uncover the mechanisms underlying EBF1-mediated regulatory processes, shedding light on cardiac development, and the pathogenesis of cardiac remodeling. These findings emphasize EBF1's critical role in orchestrating diverse aspects of cardiac processes and provide a promising therapeutic intervention for cardiomyopathy.


Asunto(s)
Perfilación de la Expresión Génica , Miocitos Cardíacos , Transactivadores , Animales , Humanos , Ratones , Transactivadores/genética , Transactivadores/metabolismo , Miocitos Cardíacos/metabolismo , Diferenciación Celular/genética , Corazón/fisiopatología , Ratones Noqueados , Células Madre Pluripotentes/metabolismo , Transcriptoma/genética , Sistemas CRISPR-Cas/genética
10.
Bioact Mater ; 34: 17-36, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38173843

RESUMEN

Abdominal Aortic Aneurysm (AAA) is a life-threatening vascular disease characterized by the weakening and ballooning of the abdominal aorta, which has no effective therapeutic approaches due to unclear molecular mechanisms. Using single-cell RNA sequencing, we analyzed the molecular profile of individual cells within control and AAA abdominal aortas. We found cellular heterogeneity, with increased plasmacytoid dendritic cells and reduced endothelial cells and vascular smooth muscle cells (VSMCs) in AAA. Up-regulated genes in AAA were associated with muscle tissue development and apoptosis. Genes controlling VSMCs aberrant switch from contractile to synthetic phenotype were significantly enriched in AAA. Additionally, VSMCs in AAA exhibited cell senescence and impaired oxidative phosphorylation. Similar observations were made in a mouse model of AAA induced by Angiotensin II, further affirming the relevance of our findings to human AAA. The concurrence of gene expression changes between human and mouse highlighted the impairment of oxidative phosphorylation as a potential target for intervention. Nicotinamide phosphoribosyltransferase (NAMPT, also named VISFATIN) signaling emerged as a signature event in AAA. NAMPT was significantly downregulated in AAA. NAMPT-extracellular vesicles (EVs) derived from mesenchymal stem cells restored NAMPT levels, and offered protection against AAA. Furthermore, NAMPT-EVs not only repressed injuries, such as cell senescence and DNA damage, but also rescued impairments of oxidative phosphorylation in both mouse and human AAA models, suggesting NAMPT supplementation as a potential therapeutic approach for AAA treatment. These findings shed light on the cellular heterogeneity and injuries in AAA, and offered promising therapeutic intervention for AAA treatment.

11.
Stem Cell Res Ther ; 14(1): 249, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37705046

RESUMEN

BACKGROUND: Cardiovascular complications significantly augment the overall COVID-19 mortality, largely due to the susceptibility of human cardiomyocytes (CMs) to SARS-CoV-2 virus. SARS-CoV-2 virus encodes 27 genes, whose specific impacts on CM health are not fully understood. This study elucidates the deleterious effects of SARS-CoV-2 genes Nsp6, M, and Nsp8 on human CMs. METHODS: CMs were derived from human pluripotent stem cells (hPSCs), including human embryonic stem cells and induced pluripotent stem cells, using 2D and 3D differentiation methods. We overexpressed Nsp6, M, or Nsp8 in hPSCs and then applied whole mRNA-seq and mass spectrometry for multi-omics analysis. Co-immunoprecipitation mass spectrometry was utilized to map the protein interaction networks of Nsp6, M, and Nsp8 within host hiPSC-CMs. RESULTS: Nsp6, Nsp8, and M globally perturb the transcriptome and proteome of hPSC-CMs. SARS-CoV-2 infection and the overexpression of Nsp6, Nsp8, or M coherently upregulated genes associated with apoptosis and immune/inflammation pathways, whereas downregulated genes linked to heart contraction and functions. Global interactome analysis revealed interactions between Nsp6, Nsp8, and M with ATPase subunits. Overexpression of Nsp6, Nsp8, or M significantly reduced cellular ATP levels, markedly increased apoptosis, and compromised Ca2+ handling in hPSC-CMs. Importantly, administration of FDA-approved drugs, ivermectin and meclizine, could restore ATP levels, thereby mitigating apoptosis and dysfunction in hPSC-CMs overexpressing Nsp6, Nsp8, or M. CONCLUSION: Overall, our findings uncover the extensive damaging effects of Nsp6, Nsp8, and M on hPSC-CMs, underlining the crucial role of ATP homeostasis in CM death and functional abnormalities induced by these SARS-CoV-2 genes, and reveal the potential therapeutic strategies to alleviate these detrimental effects with FDA-approved drugs.


Asunto(s)
COVID-19 , Células Madre Pluripotentes , Humanos , Miocitos Cardíacos , SARS-CoV-2 , Genes Virales , Adenosina Trifosfato
12.
Stem Cell Reports ; 17(3): 522-537, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35180394

RESUMEN

Patients with coronavirus disease 2019 (COVID-19) commonly have manifestations of heart disease. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome encodes 27 proteins. Currently, SARS-CoV-2 gene-induced abnormalities of human heart muscle cells remain elusive. Here, we comprehensively characterized the detrimental effects of a SARS-CoV-2 gene, Orf9c, on human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) by preforming multi-omic analyses. Transcriptomic analyses of hPSC-CMs infected by SARS-CoV-2 with Orf9c overexpression (Orf9cOE) identified concordantly up-regulated genes enriched into stress-related apoptosis and inflammation signaling pathways, and down-regulated CM functional genes. Proteomic analysis revealed enhanced expressions of apoptotic factors, whereas reduced protein factors for ATP synthesis by Orf9cOE. Orf9cOE significantly reduced cellular ATP level, induced apoptosis, and caused electrical dysfunctions of hPSC-CMs. Finally, drugs approved by the U.S. Food and Drug Administration, namely, ivermectin and meclizine, restored ATP levels and ameliorated CM death and functional abnormalities of Orf9cOE hPSC-CMs. Overall, we defined the molecular mechanisms underlying the detrimental impacts of Orf9c on hPSC-CMs and explored potentially therapeutic approaches to ameliorate Orf9c-induced cardiac injury and abnormalities.


Asunto(s)
COVID-19/patología , Proteínas de la Nucleocápside de Coronavirus/genética , Estudio de Asociación del Genoma Completo/métodos , SARS-CoV-2/genética , Potenciales de Acción/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , COVID-19/virología , Regulación hacia Abajo , Humanos , Ivermectina/farmacología , Meclizina/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosfoproteínas/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Mapas de Interacción de Proteínas/genética , ARN Mensajero/química , ARN Mensajero/metabolismo , SARS-CoV-2/aislamiento & purificación , Transducción de Señal/genética , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba
13.
Sci Immunol ; 7(72): eabo5407, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35749515

RESUMEN

Differing from the mouse Foxp3 gene that encodes only one protein product, human FOXP3 encodes two major isoforms through alternative splicing-a longer isoform (FOXP3 FL) containing all the coding exons and a shorter isoform lacking the amino acids encoded by exon 2 (FOXP3 ΔE2). The two isoforms are naturally expressed in humans, yet their differences in controlling regulatory T cell phenotype and functionality remain unclear. In this study, we show that patients expressing only the shorter isoform fail to maintain self-tolerance and develop immunodeficiency, polyendocrinopathy, and enteropathy X-linked (IPEX) syndrome. Mice with Foxp3 exon 2 deletion have excessive follicular helper T (TFH) and germinal center B (GC B) cell responses, and develop systemic autoimmune disease with anti-dsDNA and antinuclear autoantibody production, as well as immune complex glomerulonephritis. Despite having normal suppressive function in in vitro assays, regulatory T cells expressing FOXP3 ΔE2 are unstable and sufficient to induce autoimmunity when transferred into Tcrb-deficient mice. Mechanistically, the FOXP3 ΔE2 isoform allows increased expression of selected cytokines, but decreased expression of a set of positive regulators of Foxp3 without altered binding to these gene loci. These findings uncover indispensable functions of the FOXP3 exon 2 region, highlighting a role in regulating a transcriptional program that maintains Treg stability and immune homeostasis.


Asunto(s)
Autoinmunidad , Linfocitos T Reguladores , Animales , Autoinmunidad/genética , Exones/genética , Factores de Transcripción Forkhead , Humanos , Ratones , Isoformas de Proteínas/metabolismo
14.
Cytokine ; 53(1): 35-41, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21030269

RESUMEN

Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine associated with the atherosclerotic process and atherosclerotic plaque stability. MIF was shown to be highly expressed in advanced atherosclerotic lesions. Neutralizing MIF with a blocking antibody induced a regression of established atherosclerotic lesions. In this study, we investigated the mechanism underlying the proangiogenic effect of MIF in human umbilical vein endothelial cells (HUVECs). We showed that MIF induced the expression of angiogenesis-related genes in HUVECs. We also showed that MIF induced tube formation of HUVECs in vitro and in vivo. Angiotensin II (Ang II) could specifically up-regulate MIF expression in HUVECs. Using a luciferase reporter assay, we demonstrated that the AP-1 response element in the 5'-UTR of the MIF gene played a role in Ang II-induced MIF expression. Small hairpin RNA (shRNA) targeting c-Jun, a component of AP-1, and the AP-1 inhibitor CHX both efficiently inhibited MIF expression. The consistent result of electrophoretic mobility shift assay (EMSA) showed that Ang II specifically increased AP-1 activation in HUVECs. Our results suggest that AP-1 mediates Ang II-induced MIF expression which contributes to atherosclerotic plaque destabilization in human endothelial cells.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Angiotensina II/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Animales , Secuencia de Bases , Extractos Celulares , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Humanos , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/farmacología , Datos de Secuencia Molecular , Ratas , Venas Umbilicales/citología
15.
J Ethnopharmacol ; 268: 113639, 2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33301914

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Panax notoginseng saponins (PNS), the main active ingredients of Panax notoginseng (Burkill) F.H.Chen, have been clinically used for cardiovascular diseases treatment in China as the Traditional Chinese Medicine (TCM) (Duan et al., 2017). Evidence demonstrated that PNS protected cardiomyocytes from myocardial ischemia, but the more underlying molecular mechanisms of the protective effect are still unclear. The aims of this study are to systematically know the function of PNS and discover new roles of PNS in ischemic cardiomyocytes. MATERIALS AND METHODS: To confirm PNS function on ischemic cardiomyopathy, we established in vitro myocardial ischemia model on H9C2 cardiomyocyte line, which was induced by oxygen-glucose depletion (OGD). Then RNA-seq was carried out to systematically analyze global gene expression. This study was aimed to systematically investigate the protective effect and more potential molecular mechanisms of PNS on H9C2 cardiomyocytes in vitro through whole-transcriptome analysis with total RNA sequencing (RNA-Seq). RESULTS: PNS exhibited anti-apoptotic effect in H9C2 cardiomyocytes in OGD-induced myocardial ischemia model. Through RNA-seq, we found that OGD affected expression profiling of many genes, including upregulated and downregulated genes. PNS inhibited cardiomyocyte apoptosis and death through rescuing cell cycle arrest, the DNA double-strand breakage repair process and chromosome segregation. Interestingly, for the canonical signaling pathways regulation, RNA-seq showed PNS could inhibit cardiac hypertrophy, MAPK signaling pathway, and re-activate PI3K/AKT and AMPK signaling pathways. Experimental data also confirmed the PNS could protect cardiomyocytes from OGD-induced apoptosis through activating PI3K/AKT and AMPK signaling pathways. Moreover, RNA-seq demonstrated that the expression levels of many non-coding RNAs, such as miRNAs and lncRNAs, were significantly affected after PNS treatment, suggesting that PNS could protect cardiomyocytes through regulating non-coding RNAs. CONCLUSION: RNA-seq systematically revealed different novel roles of Panax Notoginseng Saponins (PNS) in protecting cardiomyocytes from apoptosis, induced by myocardial ischemia, through rescuing cell cycle arrest and cardiac hypertrophy, re-activating the DNA double-strand breakage repair process, chromosome segregation, PI3K/Akt and AMPK signaling pathways and regulating non-coding RNAs.


Asunto(s)
Isquemia Miocárdica/genética , Miocitos Cardíacos/efectos de los fármacos , Panax notoginseng , Extractos Vegetales/farmacología , RNA-Seq/métodos , Saponinas/farmacología , Animales , Línea Celular , Expresión Génica , Perfilación de la Expresión Génica/métodos , Isquemia Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/fisiología , Extractos Vegetales/uso terapéutico , Ratas , Saponinas/uso terapéutico
16.
medRxiv ; 2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32588000

RESUMEN

Four signature groups of frequently occurred single-nucleotide variants (SNVs) were identified in over twenty-eight thousand high-quality and high-coverage SARS-CoV-2 complete genome sequences, representing different viral strains. Some SNVs predominated but were mutually exclusively presented in patients from different countries and areas. These major SNV signatures exhibited distinguishable evolution patterns over time. A few hundred patients were detected with multiple viral strain-representing mutations simultaneously, which may stand for possible co-infection or potential homogenous recombination of SARS-CoV-2 in environment or within the viral host. Interestingly nucleotide substitutions among SARS-CoV-2 genomes tended to switch between bat RaTG13 coronavirus sequence and Wuhan-Hu-1 genome, indicating the higher genetic instability or tolerance of mutations on those sites or suggesting that major viral strains might exist between Wuhan-Hu-1 and RaTG13 coronavirus.

17.
Front Microbiol ; 11: 593548, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33101264

RESUMEN

Four signature groups of frequently occurred single-nucleotide variants (SNVs) were identified in over twenty-eight thousand high-quality and high-coverage SARS-CoV-2 complete genome sequences, representing different viral strains. Some SNVs predominated but were mutually exclusively presented in patients from different countries and areas. These major SNV signatures exhibited distinguishable evolution patterns over time. A few hundred patients were detected with multiple viral strain-representing mutations simultaneously, which may stand for possible co-infection or potential homogenous recombination of SARS-CoV-2 in environment or within the viral host. Interestingly nucleotide substitutions among SARS-CoV-2 genomes tended to switch between bat RaTG13 coronavirus sequence and Wuhan-Hu-1 genome, indicating the higher genetic instability or tolerance of mutations on those sites or suggesting that major viral strains might exist between Wuhan-Hu-1 and RaTG13 coronavirus.

18.
Genome Biol ; 21(1): 169, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32646524

RESUMEN

BACKGROUND: Early human heart and brain development simultaneously occur during embryogenesis. Notably, in human newborns, congenital heart defects strongly associate with neurodevelopmental abnormalities, suggesting a common gene or complex underlying both cardiogenesis and neurogenesis. However, due to lack of in vivo studies, the molecular mechanisms that govern both early human heart and brain development remain elusive. RESULTS: Here, we report ARID1A, a DNA-binding subunit of the SWI/SNF epigenetic complex, controls both neurogenesis and cardiogenesis from human embryonic stem cells (hESCs) through distinct mechanisms. Knockout-of-ARID1A (ARID1A-/-) leads to spontaneous differentiation of neural cells together with globally enhanced expression of neurogenic genes in undifferentiated hESCs. Additionally, when compared with WT hESCs, cardiac differentiation from ARID1A -/- hESCs is prominently suppressed, whereas neural differentiation is significantly promoted. Whole genome-wide scRNA-seq, ATAC-seq, and ChIP-seq analyses reveal that ARID1A is required to open chromatin accessibility on promoters of essential cardiogenic genes, and temporally associated with key cardiogenic transcriptional factors T and MEF2C during early cardiac development. However, during early neural development, transcription of most essential neurogenic genes is dependent on ARID1A, which can interact with a known neural restrictive silencer factor REST/NRSF. CONCLUSIONS: We uncover the opposite roles by ARID1A to govern both early cardiac and neural development from pluripotent stem cells. Global chromatin accessibility on cardiogenic genes is dependent on ARID1A, whereas transcriptional activity of neurogenic genes is under control by ARID1A, possibly through ARID1A-REST/NRSF interaction.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Corazón/embriología , Células Madre Embrionarias Humanas/fisiología , Neurogénesis , Factores de Transcripción/fisiología , Línea Celular , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Humanos , RNA-Seq , Análisis de Secuencia de ARN , Análisis de la Célula Individual
19.
Dev Cell ; 42(4): 333-348.e5, 2017 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-28829943

RESUMEN

Cardiogenesis processes in human and animals have differential dynamics, suggesting the existence of species-specific regulators during heart development. However, it remains a challenge to discover the human-specific cardiac regulatory genes, given that most coding genes are conserved. Here, we report the identification of a human-specific long noncoding RNA, Heart Brake LncRNA 1 (HBL1), which regulates cardiomyocyte development from human induced pluripotent stem cells (hiPSCs). Overexpression of HBL1 repressed, whereas knockdown and knockout of HBL1 increased, cardiomyocyte differentiation from hiPSCs. HBL1 physically interacted with MIR1 in an AGO2 complex. Disruption of MIR1 binding sites in HBL1 showed an effect similar to that of HBL1 knockout. SOX2 bound to HBL1 promoter and activated its transcription. Knockdown of SOX2 in hiPSCs led to decreased HBL1 expression and increased cardiomyocyte differentiation efficiency. Thus, HBL1 plays a modulatory role in fine-tuning human-specific cardiomyocyte development by forming a regulatory network with SOX2 and MIR1.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción SOXB1/metabolismo , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sitios de Unión , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Proteínas del Tejido Nervioso/genética , Unión Proteica , ARN Largo no Codificante/metabolismo , Factores de Transcripción SOXB1/genética
20.
Artículo en Zh | MEDLINE | ID: mdl-17094627

RESUMEN

OBJECTIVE: To clone and characterize a RRas-like gene from Trichomonas vaginalis for studying cellular signal transduction pathways in the organism. METHODS: A cDNA clone, which showed homology with RRas proteins of human being, was isolated and sequenced from a cDNA expression library of T. vaginalis. The genomic DNA corresponding to the cDNA sequence was amplified using PCR technique and sequenced. Sequence analysis was performed using BLASTP, RPS-BLAST and Clustal W programs. Phylogenetic tree was constructed and bootstrapped with 1050 replicates using the software MEGA3. RESULTS: The cDNA sequence showed a length of 705 bp with an open reading frame of 615 bp. The deduced amino acid sequence from the open reading frame possesses 205 residuals. Sequencing of the PCR product of genomic DNA revealed that the genomic DNA sequence encompassing the putative 5'-ATG and 3'-stop codons was identical to the cDNA sequence. Sequence analysis demonstrated that this gene was most homologous to the RRas members of Homo sapiens and Mus musculus (both having 51% identity and 70% similarity), and the amino acid sequence contains highly conserved GTP-binding domains and a fully conserved effector domain of human RRas member. Phylogenetic analysis showed that TvRRas clustered with RAS oncoprotein branch and RRAS branch of human. CONCLUSION: The encoding protein probably belongs to a RRas family of T. vaginalis.


Asunto(s)
ADN Complementario/genética , Proteínas de la Membrana/genética , Proteínas de Unión al GTP Monoméricas/genética , Trichomonas vaginalis/genética , Animales , Clonación Molecular , Biblioteca de Genes , Humanos , Reacción en Cadena de la Polimerasa , Análisis de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA