Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 82(21): 4116-4130.e6, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36283412

RESUMEN

Pyruvate carboxylase (PC) catalyzes the two-step carboxylation of pyruvate to produce oxaloacetate, playing a key role in the maintenance of metabolic homeostasis in cells. Given its involvement in multiple diseases, PC has been regarded as a potential therapeutic target for obesity, diabetes, and cancer. Albeit acetyl-CoA has been recognized as the allosteric regulator of PC for over 60 years, the underlying mechanism of how acetyl-CoA induces PC activation remains enigmatic. Herein, by using time-resolved cryo-electron microscopy, we have captured the snapshots of PC transitional states during its catalytic cycle. These structures and the biochemical studies reveal that acetyl-CoA stabilizes PC in a catalytically competent conformation, which triggers a cascade of events, including ATP hydrolysis and the long-distance communication between the two reactive centers. These findings provide an integrated picture for PC catalysis and unveil the unique allosteric mechanism of acetyl-CoA in an essential biochemical reaction in all kingdoms of life.


Asunto(s)
Acetil-CoA Carboxilasa , Piruvato Carboxilasa , Humanos , Piruvato Carboxilasa/genética , Piruvato Carboxilasa/metabolismo , Acetilcoenzima A/metabolismo , Regulación Alostérica , Microscopía por Crioelectrón , Conformación Molecular , Acetil-CoA Carboxilasa/metabolismo
2.
Nucleic Acids Res ; 52(5): 2273-2289, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38118002

RESUMEN

Albeit N1-Methyladenosine (m1A) RNA modification represents an important regulator of RNA metabolism, the role of m1A modification in carcinogenesis remains enigmatic. Herein, we found that histone lactylation enhances ALKBH3 expression and simultaneously attenuates the formation of tumor-suppressive promyelocytic leukemia protein (PML) condensates by removing the m1A methylation of SP100A, promoting the malignant transformation of cancers. First, ALKBH3 is specifically upregulated in high-risk ocular melanoma due to excessive histone lactylation levels, referring to m1A hypomethylation status. Moreover, the multiomics analysis subsequently identified that SP100A, a core component for PML bodies, serves as a downstream candidate target for ALKBH3. Therapeutically, the silencing of ALKBH3 exhibits efficient therapeutic efficacy in melanoma both in vitro and in vivo, which could be reversed by the depletion of SP100A. Mechanistically, we found that YTHDF1 is responsible for recognition of the m1A methylated SP100A transcript, which increases its RNA stability and translational efficacy. Conclusively, we initially demonstrated that m1A modification is necessary for tumor suppressor gene expression, expanding the current understandings of dynamic m1A function during tumor progression. In addition, our results indicate that lactylation-driven ALKBH3 is essential for the formation of PML nuclear condensates, which bridges our knowledge of m1A modification, metabolic reprogramming, and phase-separation events.


Asunto(s)
Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 3 de AlkB , Antígenos Nucleares , Autoantígenos , Neoplasias del Ojo , Histonas , Melanoma , Humanos , Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 3 de AlkB/metabolismo , Desmetilación , Metilación de ADN , Histonas/genética , Histonas/metabolismo , Proteína de la Leucemia Promielocítica/genética , Proteína de la Leucemia Promielocítica/metabolismo , ARN/metabolismo , Factores de Transcripción/metabolismo , Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Neoplasias del Ojo/metabolismo
3.
J Transl Med ; 22(1): 605, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951874

RESUMEN

BACKGROUND: Uveal melanoma (UM), the most common adult intraocular tumor, is characterized by high malignancy and poor prognosis in advanced stages. Angiogenesis is critical for UM development, however, not only the role of vascular endothelial dysfunction in UM remains unknown, but also their analysis at the single-cell level has been lacking. A comprehensive analysis is essential to clarify the role of the endothelium in the development of UM. METHODS: By using single-cell RNA transcriptomics data of 11 cases of primary and liver metastasis UM, we analyzed the endothelial cell status. In addition, we analyzed and validated ECs in the in vitro model and collected clinical specimens. Subsequently, we explored the impact of endothelial dysfunction on UM cell migration and explored the mechanisms responsible for the endothelial cell abnormalities and the reasons for their peripheral effects. RESULTS: UM metastasis has a significantly higher percentage of vascular endothelial cells compared to in situ tumors, and endothelial cells in metastasis show significant senescence. Senescent endothelial cells in metastatic tumors showed significant Krüppel-like factor 4 (KLF4) upregulation, overexpression of KLF4 in normal endothelial cells induced senescence, and knockdown of KLF4 in senescent endothelium inhibited senescence, suggesting that KLF4 is a driver gene for endothelial senescence. KLF4-induced endothelial senescence drove tumor cell migration through a senescence-associated secretory phenotype (SASP), of which the most important component of the effector was CXCL12 (C-X-C motif chemokine ligand 12), and participated in the composition of the immunosuppressive microenvironment. CONCLUSION: This study provides an undesirable insight of senescent endothelial cells in promoting UM metastasis.


Asunto(s)
Movimiento Celular , Senescencia Celular , Células Endoteliales , Factor 4 Similar a Kruppel , Neoplasias Hepáticas , Melanoma , Análisis de la Célula Individual , Neoplasias de la Úvea , Humanos , Neoplasias de la Úvea/patología , Neoplasias de la Úvea/genética , Melanoma/patología , Melanoma/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética , Regulación Neoplásica de la Expresión Génica , Femenino , Masculino
4.
J Transl Med ; 21(1): 403, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37344841

RESUMEN

Metformin is a well-known anti-diabetic drug that has been repurposed for several emerging applications, including as an anti-cancer agent. It boasts the distinct advantages of an excellent safety and tolerability profile and high cost-effectiveness at less than one US dollar per daily dose. Epidemiological evidence reveals that metformin reduces the risk of cancer and decreases cancer-related mortality in patients with diabetes; however, the exact mechanisms are not well understood. Energy metabolism may be central to the mechanism of action. Based on altering whole-body energy metabolism or cellular state, metformin's modes of action can be divided into two broad, non-mutually exclusive categories: "direct effects", which induce a direct effect on cancer cells, independent of blood glucose and insulin levels, and "indirect effects" that arise from systemic metabolic changes depending on blood glucose and insulin levels. In this review, we summarize an updated account of the current knowledge on metformin antitumor action, elaborate on the underlying mechanisms in terms of the hallmarks of cancer, and propose potential applications for repurposing metformin for cancer therapeutics.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insulinas , Metformina , Neoplasias , Humanos , Metformina/farmacología , Metformina/uso terapéutico , Glucemia , Reposicionamiento de Medicamentos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Insulinas/uso terapéutico , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico
5.
J Craniofac Surg ; 34(8): 2379-2383, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37639658

RESUMEN

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Our study attempted to analyze the research trends in HNSCC and compare contributions from different countries, institutions, journals, and authors. MATERIALS AND METHODS: The authors extracted publications in this field from 2002 to 2022 from the Web of Science database. Microsoft Excel and VOSviewer were performed to collect data on publication numbers, analyze publication trends, and visualize relevant results. RESULTS: A total of 1903 publications were screened. In the past 20 years, the United States contributed the most publications and citations in the HNSCC research. China ranked second in the number of publications. The Ophthalmic Plastic and Reconstructive Surgery was the most productive journal concerning HNSCC. ESMAELIB of the University of Texas System and ROSENTHAL EL of Stanford University had published the most publications in this field. Keywords were categorized into 3 clusters: basic study, clinical feature study, and treatment-related study. The keywords "reflectance confocal microscopy", "raman-spectroscopy", and "confocal laser endomicroscopy" were most frequently emerged in the recent years. Management-related research has been recognized as a potential focus in the HNSCC.


Asunto(s)
Bibliometría , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , China , Bases de Datos Factuales , Neoplasias de Cabeza y Cuello/cirugía
6.
J Mol Cell Cardiol ; 162: 130-143, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34536440

RESUMEN

Venous malformation (VM) and cavernous venous malformation (CVM) are two types of vascular malformations. Even if the two diseases are similar in appearance and imaging, the distinct cellular components and signaling pathways between them might help distinguish the two from a molecular perspective. Here, we performed single-cell profiling of 35,245 cells from two VM samples and three CVM samples, with a focus on endothelial cells (ECs), smooth muscle cells (SMCs) and immune microenvironment (IME). Clustering analysis based on differential gene expression unveiled 11 specific cell types, and determined CVM had more SMCs. Re-clustering of ECs and SMCs indicated CVM was dominated by arterial components, while VM is dominated by venous components. Gene set variation analysis suggested the activation of inflammation-related pathways in VM ECs, and upregulation of myogenesis pathway in CVM SMCs. In IME analysis, immune cells were identified to accounted for nearly 30% of the total cell number, including macrophages, monocytes, NK cells, T cells and B cells. Notably, more macrophages and monocytes were discovered in VM, indicating innate immune responses might be more closely related to VM pathogenesis. In addition, angiogenesis pathway was highlighted among the significant pathways of macrophages & monocytes between CVM and VM. In VM, VEGFA was highly expressed in macrophages & monocytes, while its receptors were all abundantly present in ECs. The close interaction of VEGFA on macrophages with its receptors on ECs was also predicted by CellPhoneDB analysis. Our results document cellular composition, significant pathways, and critical IME in CVM and VM development.


Asunto(s)
Transcriptoma , Malformaciones Vasculares , Células Endoteliales/metabolismo , Humanos , Miocitos del Músculo Liso/metabolismo , Malformaciones Vasculares/genética , Malformaciones Vasculares/metabolismo , Malformaciones Vasculares/patología , Venas/anomalías
7.
Ophthalmology ; 129(7): 771-780, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35245602

RESUMEN

PURPOSE: To assess the predictive value of the tumor staging system in the AJCC Cancer Staging Manual, Eighth Edition, and histologic features for outcomes and metastasis patterns in conjunctival melanoma (CM). DESIGN: Retrospective, single-center cohort study. PARTICIPANTS: Eighty-three patients with CM were treated at Shanghai Ninth People's Hospital between 2000 and 2021. METHODS: We reviewed the clinical and histologic parameters and used Kaplan-Meier survival curves and Cox proportional hazards regression models for risk factor analyses. MAIN OUTCOME MEASURES: Time to nodal/distant metastasis, disease-specific survival, metastatic pattern, and metastatic site. RESULTS: At presentation, 5 patients (6%) had clinical tumor (cT)1 disease, 34 patients (41%) had cT2 disease, and 44 patients (53%) had cT3 disease. Four patients (5%) had nodal metastasis (N1), and none had distant metastasis (M1). During follow-up, 12 patients (14%) developed nodal metastasis, 29 patients (35%) developed distant metastasis, and 26 patients (31%) died of disease. The brain, liver, and lung were common distant metastasis sites. Higher cT category was associated with increased risks of distant metastasis (P < 0.001) and disease-specific death (P = 0.002). The separate analysis of primary and recurrent tumors at presentation showed that the patients with cT3 tumors had a higher risk of distant metastasis than those with cT2 tumors. Greater tumor thickness, ulceration, and the presence of regression were correlated with distant metastasis. Previously unreported mutations were detected in the tumor suppressor genes FAT atypical cadherin 4 (FAT4) and spleen associated tyrosine kinase (SYK). Among the 29 patients who developed distant metastasis, we analyzed 2 patterns of metastasis: Eleven patients (38%) developed nodal metastasis before distant metastasis, and 18 patients (62%) developed distant metastasis without previously known nodal metastasis. The patients with cT3 tumors were more likely to follow the latter metastasis pattern (P = 0.02). CONCLUSIONS: Conjunctival melanoma presented with mostly advanced stages and high rates of distant metastasis in the current Chinese cohort. This study confirmed the prognostic value of the tumor staging system in the AJCC Cancer Staging Manual, Eighth Edition, for Chinese patients. Histologic features, such as tumor thickness and ulceration, should be emphasized when assessing prognosis and guiding the treatment of CM.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Mama , Neoplasias de la Conjuntiva , Melanoma , Neoplasias de la Mama/patología , China/epidemiología , Estudios de Cohortes , Neoplasias de la Conjuntiva/patología , Femenino , Humanos , Metástasis Linfática , Melanoma/patología , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Estudios Retrospectivos , Úlcera , Estados Unidos
8.
Nucleic Acids Res ; 48(21): 12135-12150, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33196849

RESUMEN

Chromatin remodeling impacts the structural neighborhoods and regulates gene expression. However, the role of enhancer-guided chromatin remodeling in the gene regulation remains unclear. Here, using RNA-seq and ChIP-seq, we identified for the first time that neurotensin (NTS) serves as a key oncogene in uveal melanoma and that CTCF interacts with the upstream enhancer of NTS and orchestrates an 800 kb chromosomal loop between the promoter and enhancer. Intriguingly, this novel CTCF-guided chromatin loop was ubiquitous in a cohort of tumor patients. In addition, a disruption in this chromosomal interaction prevented the histone acetyltransferase EP300 from embedding in the promoter of NTS and resulted in NTS silencing. Most importantly, in vitro and in vivo experiments showed that the ability of tumor formation was significantly suppressed via deletion of the enhancer by CRISPR-Cas9. These studies delineate a novel onco-enhancer guided epigenetic mechanism and provide a promising therapeutic concept for disease therapy.


Asunto(s)
Factor de Unión a CCCTC/genética , Carcinogénesis/genética , Proteína p300 Asociada a E1A/genética , Melanoma/genética , Neurotensina/genética , Neoplasias de la Úvea/genética , Animales , Factor de Unión a CCCTC/metabolismo , Sistemas CRISPR-Cas , Carcinogénesis/metabolismo , Carcinogénesis/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteína p300 Asociada a E1A/metabolismo , Elementos de Facilitación Genéticos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Melanoma/mortalidad , Melanoma/patología , Ratones , Ratones Desnudos , Neurotensina/antagonistas & inhibidores , Neurotensina/metabolismo , Regiones Promotoras Genéticas , Eliminación de Secuencia , Análisis de Supervivencia , Carga Tumoral , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/mortalidad , Neoplasias de la Úvea/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Craniofac Surg ; 33(3): e238-e240, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34374675

RESUMEN

ABSTRACT: The aim of the study was to report a novel forkhead box L2 (FOXL2) missense mutation in a Chinese blepharophimosis/ ptosis/epicanthus inversus syndrome family. Three generations of the Chinese family with blepharophimosis/ptosis/epicanthus inversus syndrome were enrolled in this study. Blood samples from patients of this family were collected and then analyzed by whole-exome sequencing. Confocal microscopy was performed to detect the subcellular location of FOXL2. Transactivation studies were performed and verified with real time polymerase chain reaction. A novel mutation (c.1068G>C) located in the downstream of deoxyribonucleic acid-binding forkhead domain was identified. Confocal photos showed the novel mutation did not disturb FOXL2 function, and the mutant protein could still transactivate steroidogenic acute regulatory protein, a key regulator of primary ovarian failure (POF). Our study revealed a novel missense mutation (c.1068G>C) and expanded the spectrum of FOXL2 gene mutations.


Asunto(s)
Blefarofimosis , Proteína Forkhead Box L2 , Anomalías Cutáneas , Anomalías Urogenitales , Blefarofimosis/diagnóstico , Blefarofimosis/genética , China , Proteína Forkhead Box L2/genética , Humanos , Mutación Missense , Linaje , Anomalías Cutáneas/diagnóstico , Anomalías Cutáneas/genética , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/genética
10.
J Mol Cell Cardiol ; 147: 92-107, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32730768

RESUMEN

Venous malformation (VM) is a type of vascular morphogenic defect in humans with an incidence of 1%. Although gene mutation is considered as the most common cause of VM, the pathogenesis of those without gene mutation remains to be elucidated. Here, we aimed to explore the relation of bone morphogenetic protein 9 (BMP9) and development of VM. At first, we found serum and tissue BMP9 expression in VM patients was significantly lower than that in healthy subjects, detected via enzyme-linked immunosorbent assay. Next, with wound healing assay, transwell assay and tube formation assay, we discovered BMP9 could inhibit migration and enhance tube formation activity of human umbilical vein endothelial cells (HUVECs) via receptor activin receptor-like kinase 1 (ALK1). Besides, BMP9 improved the expression of structural proteins alpha-smooth muscle actin (α-SMA) and Desmin in human umbilical vein smooth muscle cells (HUVSMCs) via activation of the SMAD1/5-ID1 pathway, determined by RNA-based next-generation sequencing, qPCR, immunofluorescence and western blotting. Intriguingly, this effect could be blocked by receptor ALK1 inhibitor, SMAD1/5 inhibitor and siRNAs targeting ID1, verifying the BMP9/ALK1/SMAD1/5/ID1/α-SMA pathway. Meanwhile, knocking out BMP9 in C57BL/6 mice embryo led to α-SMA scarcity in walls of lung and mesenteric vessels, as well as walls of small trachea. BMP9-/- zebrafish also exhibited abnormal vascular maturity, indicating a critical role of BMP9 in vascular maturity and remodeling. Finally, a VM mice model revealed that BMP9 might have therapeutic effect in VM progression. Our study discovered that BMP9 might inhibit the occurrence of VM by strengthening the vessel wall and maintaining endothelium quiescence. These findings provide promising evidences of new therapeutic targets that might be used for the management of VM.


Asunto(s)
Actinas/metabolismo , Factor 2 de Diferenciación de Crecimiento/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Venas/anomalías , Adolescente , Adulto , Anciano , Animales , Movimiento Celular , Proliferación Celular , Niño , Preescolar , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , Factor de Crecimiento Transformador beta/metabolismo , Venas/patología , Adulto Joven
11.
Mol Cancer ; 19(1): 115, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669100

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) have been identified as important epigenetic regulators that play critical roles in human cancers. However, the regulatory functions of lncRNAs in tumorigenesis remains to be elucidated. Here, we aimed to investigate the molecular mechanisms and potential clinical application of a novel lncRNA, retinoblastoma associated transcript-1 (RBAT1), in tumorigenesis. METHODS: RBAT1 expression was determined by real-time PCR in both retinoblastoma (Rb) and bladder cancer (BCa) cell lines and clinical tissues. Chromatin isolation using RNA purification (ChIRP) assays were performed to identify RBAT1-interacting proteins. Patient-derived xenograft (PDX) retinoblastoma models were established to test the therapeutic potential of RBAT1-targeting GapmeRs. RESULTS: Here, we found that RBAT1 expression was significantly higher in Rb and BCa tissues than that in adjacent tissues. Functional assays revealed that RBAT1 accelerated tumorigenesis both in vitro and in vivo. Mechanistically, RBAT1 recruited HNRNPL protein to E2F3 promoter, thereby activating E2F3 transcription. Therapeutically, GapmeR-mediated RBAT1 silencing significantly inhibited tumorigenesis in orthotopic xenograft retinoblastoma models derived from Rb cell lines and Rb primary cells. CONCLUSIONS: RBAT1 overexpression upregulates a known oncogene, E2F3, via directly recruiting HNPNPL to its promoter and cis-activating its expression. Our finding provides a novel mechanism of lncRNA biology and provides potential targets for diagnosis and treatment of Rb and BCa.


Asunto(s)
Transformación Celular Neoplásica/genética , Factor de Transcripción E2F3/genética , Regulación de la Expresión Génica , ARN Largo no Codificante/genética , Ribonucleoproteínas/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Factor de Transcripción E2F3/metabolismo , Silenciador del Gen , Humanos , Inmunohistoquímica , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , Retinoblastoma/genética , Retinoblastoma/metabolismo , Ribonucleoproteínas/metabolismo , Transducción de Señal , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Hum Genet ; 139(12): 1499-1511, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32533362

RESUMEN

Vascular anomalies (VAs), comprising wide subtypes of tumors and malformations, are often caused by variants in multiple tyrosine kinase (TK) receptor signaling pathways including TIE2, PIK3CA and GNAQ/11. Yet, a portion of individuals with clinical features of VA do not have variants in these genes, suggesting that there are undiscovered pathogenic factors underlying these patients and possibly with overlapping phenotypes. Here, we identified one rare non-synonymous variant (c.968A > G) in the seventh exon of GPAA1 (Glycosylphosphatidylinositol Anchor Attachment Protein 1), shared by the four affected members of a large pedigree with multiple types of VA using whole-exome sequencing. GPAA1 encodes a glycosylphosphatidylinositol (GPI) transamidase complex protein. This complex orchestrates the attachment of the GPI anchor to the C terminus of precursor proteins in the endoplasmic reticulum (ER). We showed such variant led to scarce expression of GPAA1 protein in vascular endothelium and induced a localization change from ER membrane to cytoplasm and nucleus. In addition, expressing wild-type GPAA1 in endothelial cells had an effect to inhibit cell proliferation and migration, while expressing variant GPAA1 led to overgrowth and overmigration, indicating a loss of the quiescent status. Finally, a gpaa1-deficient zebrafish model displayed several types of developmental defects as well as vascular dysplasia, demonstrating that GPAA1 is involved in angiogenesis and vascular remodeling. Altogether, our results indicate that the rare coding variant in GPAA1 (c.968A > G) is causally related to familial forms of VAs.


Asunto(s)
Exoftalmia/genética , Glicoproteínas de Membrana/genética , Complejos Multiproteicos/genética , Malformaciones Vasculares/genética , Aciltransferasas/genética , Adulto , Animales , Sistemas CRISPR-Cas/genética , Movimiento Celular/genética , Proliferación Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Exones/genética , Exoftalmia/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Malformaciones Vasculares/patología , Secuenciación del Exoma , Pez Cebra/genética
13.
Mol Ther ; 27(12): 2182-2194, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31451355

RESUMEN

Long noncoding RNAs (lncRNAs) are an important class of pervasive noncoding RNA involved in a variety of biological functions. Numerous studies have demonstrated their important regulatory role in human disease, especially cancer. However, the mechanism underlying the transcription of lncRNAs is not fully elucidated. Here, a comparison of local chromatin structure of the ROR lncRNA locus revealed a cohesin-complex-mediated intrachromosomal loop that is juxtaposed with an upstream enhancer to the ROR promoter, enabling activation of endogenous ROR lncRNA in tumor cells. This chromosomal interaction was not observed in normal control cells. Knockdown of SMC1 by RNAi or deletion of the enhancer DNA by CRISPR/Cas9 abolished the intrachromosomal interaction, resulting in ROR lncRNA silencing and inhibition of the tumor progression in animals carrying tumor xenografts. Our results reveal a novel mechanism by which the cohesin-orchestrated intrachromosomal looping may serve as a critical epigenetic driver to activate transcription of ROR lncRNA, subsequently inducing tumorigenesis. Our data represent a novel chromosomal folding pattern of lncRNA regulation, thereby providing a novel alternative concept of chromosomal interaction in lncRNA-triggered tumorigenesis.


Asunto(s)
Carcinogénesis/patología , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos , Complejo Mediador/genética , Neoplasias/patología , ARN Largo no Codificante/genética , ARN Interferente Pequeño/genética , Animales , Apoptosis , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proliferación Celular , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Complejo Mediador/antagonistas & inhibidores , Ratones , Ratones Desnudos , Neoplasias/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , ARN Largo no Codificante/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nucleic Acids Res ; 46(12): 6041-6056, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29741668

RESUMEN

Aberrant chromatin transformation dysregulates gene expression and may be an important driver of tumorigenesis. However, the functional role of chromosomal dynamics in tumorigenesis remains to be elucidated. Here, using in vitro and in vivo experiments, we reveal a novel long noncoding (lncing) driver at chr12p13.3, in which a novel lncRNA GALNT8 Antisense Upstream 1 (GAU1) is initially activated by an open chromatin status, triggering recruitment of the transcription elongation factor TCEA1 at the oncogene GALNT8 promoter and cis-activates the expression of GALNT8. Analysis of The Cancer Genome Atlas (TCGA) clinical database revealed that the GAU1/GALNT8 driver serves as an important indicative biomarker, and targeted silencing of GAU1 via the HKP-encapsulated method exhibited therapeutic efficacy in orthotopic xenografts. Our study presents a novel oncogenetic mechanism in which aberrant tuning of the chromatin state at specific chromosomal loci exposes factor-binding sites, leading to recruitment of trans-factor and activation of oncogenetic driver, thereby provide a novel alternative concept of chromatin dynamics in tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Cromosomas Humanos Par 12 , Regulación Neoplásica de la Expresión Génica , N-Acetilgalactosaminiltransferasas/genética , ARN Largo no Codificante/metabolismo , Adulto , Animales , Biomarcadores de Tumor , Línea Celular , Células Cultivadas , Cromatina/metabolismo , Progresión de la Enfermedad , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , N-Acetilgalactosaminiltransferasas/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Regiones Promotoras Genéticas , ARN Largo no Codificante/genética , Neoplasias de la Retina/genética , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patología , Factores de Elongación Transcripcional/metabolismo , Polipéptido N-Acetilgalactosaminiltransferasa
15.
J Biol Chem ; 293(52): 20099-20111, 2018 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-30389786

RESUMEN

The CD4+CD25+FOXP3+ regulatory T (Treg) cells are critical for maintaining immune tolerance in healthy individuals and are reported to restrict anti-inflammatory responses and thereby promote tumor progression, suggesting them as a target in the development of antitumor immunotherapy. Forkhead box P3 (FOXP3) is a key transcription factor governing Treg lineage differentiation and their immune-suppressive function. Here, using Treg cells, as well as HEK-293T and Jurkat T cells, we report that the stability of FOXP3 is directly and positively regulated by the E3 ubiquitin ligase ring finger protein 31 (RNF31), which catalyzes the conjugation of atypical ubiquitin chains to the FOXP3 protein. We observed that shRNA-mediated RNF31 knockdown in human Treg cells decreases FOXP3 protein levels and increases levels of interferon-γ, resulting in a Th1 helper cell-like phenotype. Human Treg cells that ectopically expressed RNF31 displayed stronger immune-suppressive capacity, suggesting that RNF31 positively regulates both FOXP3 stability and Treg cell function. Moreover, we found that RNF31 is up-regulated in Treg cells that infiltrate human gastric tumor tissues compared with their counterparts residing in peripheral and normal tissue. We also found that elevated RNF31 expression in intratumoral Treg cells is associated with poor survival of gastric cancer patients, suggesting that RNF31 supports the immune-suppressive functions of Treg cells. Our results suggest that RNF31 could be a potential therapeutic target in immunity-based interventions against human gastric cancer.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Linfocitos T Reguladores/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación/inmunología , Regulación hacia Arriba/inmunología , Supervivencia sin Enfermedad , Células HEK293 , Humanos , Células Jurkat , Estabilidad Proteica , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Tasa de Supervivencia , Linfocitos T Reguladores/patología
16.
J Cell Biochem ; 120(4): 5766-5776, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30320917

RESUMEN

BACKGROUND: Uveal melanoma (UM) is a severe human malignancy with a high mortality rate, as well as high metastasis and recurrence potential. The active mutation of G protein subunit alpha q (GNAQ) or G protein subunit alpha 11 (GNA11) is a major trigger for UM. Oncolytic adenovirus H101 (H101) is the first oncolytic virus approved for clinical applications in cancer therapy by the China Food and Drug Administration. We investigated whether combining H101 with the downregulation of GNAQ expression would act synergistically in UM therapy. METHODS: Three UM cell lines OMM2.3 and 92.1, harboring GNAQ mutation, and OCM1, harboring B-Raf proto-oncogene mutation, were chosen for our research. The cellular toxicity of adenoviral infection and the cell growth rate were measured with a Cell Counting Kit-8. Western blot analysis was used to detect GNAQ, p-MEK1/2, YAP, and p-YAP expression. The apoptosis and cell-cycle distribution of cells were evaluated with annexin-V and propidium iodide staining. RESULTS: Our results revealed that OMM2.3 and 92.1 cells were more sensitive to H101 infection than OCM1 cells. GNAQ expression was markedly reduced by small interfering RNA, siGNAQ. Combined treatment of siGNAQ and H101 inhibited the proliferation and activated the apoptosis of OMM2.3 and 92.1 cells by blocking the phosphorylation of MEK1/2 and increasing the phosphorylation of YAP. CONCLUSIONS: In summary, a therapy combining H101 and siGNAQ is feasible, with potential utility as a novel targeted molecular therapy for UM, especially those carrying a GNAQ mutation.


Asunto(s)
Apoptosis , Proliferación Celular , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Melanoma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , ARN Interferente Pequeño/genética , Neoplasias de la Úvea/terapia , Ciclo Celular , Terapia Combinada , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Humanos , Melanoma/genética , Melanoma/patología , Fosforilación , Proto-Oncogenes Mas , Transducción de Señal , Células Tumorales Cultivadas , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/patología
17.
Mol Cancer ; 18(1): 6, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626395

RESUMEN

Covalently closed single-stranded circular RNAs (circRNAs) consist of introns or exons and are widely present in eukaryotic cells. CircRNAs generally have low expression levels and relatively stable structures compared with messenger RNAs (mRNAs), most of which are located in the cytoplasm and often act in cell type and tissue-specific manners, indicating that they may serve as novel biomarkers. In recent years, circRNAs have gradually become a hotspot in the field of RNA and cancer research, but the functions of most circRNAs have not yet been discovered. Known circRNAs can affect the biogenesis of cancers in diverse ways, such as functioning as a microRNA (miRNA) sponges, combining with RNA binding proteins (RBPs), working as a transcription factor and translation of proteins. In this review, we summarize the characteristics and types of circRNAs, introduce the biogenesis of circRNAs, discuss the emerging functions and databases on circRNAs and present the current challenges of circRNAs studies.


Asunto(s)
Neoplasias/genética , ARN/genética , Animales , Biomarcadores/metabolismo , Exones/genética , Humanos , Intrones/genética , MicroARNs/genética , Neoplasias/metabolismo , ARN Circular , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética
18.
Mol Cancer ; 18(1): 161, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31722709

RESUMEN

BACKGROUND: Dynamic N6-methyladenosine (m6A) RNA modification generated and erased by N6-methyltransferases and demethylases regulates gene expression, alternative splicing and cell fate. Ocular melanoma, comprising uveal melanoma (UM) and conjunctival melanoma (CM), is the most common primary eye tumor in adults and the 2nd most common melanoma. However, the functional role of m6A modification in ocular melanoma remains unclear. METHODS: m6A assays and survival analysis were used to explore decreased global m6A levels, indicating a late stage of ocular melanoma and a poor prognosis. Multiomic analysis of miCLIP-seq, RNA-seq and Label-free MS data revealed that m6A RNA modification posttranscriptionally promoted HINT2 expression. RNA immunoprecipitation (RIP)-qPCR and dual luciferase assays revealed that HINT2 mRNA specifically interacted with YTHDF1. Furthermore, polysome profiling analysis indicated a greater amount of HINT2 mRNA in the translation pool in ocular melanoma cells with higher m6A methylation. RESULTS: Here, we show that RNA methylation significantly inhibits the progression of UM and CM. Ocular melanoma samples showed decreased m6A levels, indicating a poor prognosis. Changes in global m6A modification were highly associated with tumor progression in vitro and in vivo. Mechanistically, YTHDF1 promoted the translation of methylated HINT2 mRNA, a tumor suppressor in ocular melanoma. CONCLUSIONS: Our work uncovers a critical function for m6A methylation in ocular melanoma and provides additional insight into the understanding of m6A modification.


Asunto(s)
Adenosina/análogos & derivados , Neoplasias del Ojo/genética , Melanoma/genética , Proteínas Mitocondriales/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Adenosina/metabolismo , Apoptosis , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias del Ojo/metabolismo , Neoplasias del Ojo/mortalidad , Neoplasias del Ojo/patología , Perfilación de la Expresión Génica , Humanos , Melanoma/metabolismo , Melanoma/mortalidad , Melanoma/patología , Metilación , Proteínas Mitocondriales/metabolismo , Pronóstico , ARN Mensajero/metabolismo
19.
Exp Eye Res ; 184: 64-71, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30898556

RESUMEN

Neovascularization during ocular tissue repair can cause severe visual loss in the optical axis and is therefore an issue of considerable concern to ophthalmologists. Here, we introduced a cholesterol-modified siRNA delivery system targeting stromal cell-derived factor 1 (SDF-1) to treat ocular angiogenesis in vivo. SDF-1 expression was analyzed in rat endothelial progenitor cells (EPCs) and bone marrow mesenchymal stem cells (BMSCs) using quantitative PCR (qPCR). Migration ability of BMSC and HUVEC were assessed through transwell assay. The proliferation effect of chol-siSDF1 on HUVEC was measured by colony formation assay. In vivo anti-angiogenic effects of chol-siSDF1 were tested in a cornea alkali burn model and the area of cornea neovascularization was measured using computer-imaging analysis system. Then phosphorylated Akt and total Akt protein levels were measured through western blot. Results turned out that rat EPCs and BMSCs showed high SDF-1 mRNA expression, which can be down-regulated by using chol-siSDF-1. Chol-siSDF-1 could significantly inhibit migration of BMSC and HUVEC. In addition, chol-siSDF1 also could inhibit HUVEC proliferation and exert a significant anti-angiogenic effect in corneal alkali burn model. As for the mechanism, chol-siSDF1 may inhibit the neovascularization, proliferation and metastasis through inhibiting the Akt signaling pathway. Thus, cholesterol modification of siRNA targeting SDF-1 displays an effective inhibition of migration and angiogenesis, with a much longer duration of inhibition effect.


Asunto(s)
Quemaduras Químicas/terapia , Quimiocina CXCL12/genética , Colesterol/química , Neovascularización de la Córnea/terapia , Quemaduras Oculares/inducido químicamente , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , ARN Interferente Pequeño/uso terapéutico , Inhibidores de la Angiogénesis , Animales , Western Blotting , Movimiento Celular/fisiología , Células Cultivadas , Neovascularización de la Córnea/metabolismo , Células Progenitoras Endoteliales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Hidróxido de Sodio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA