Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 36(6): e22366, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35608889

RESUMO

AGGF1 is an angiogenic factor with G-Patch and FHA domains 1 described by our group. Gain-of-function mutations in AGGF1 cause Klippel-Trenaunay syndrome, whereas somatic loss-of-function mutations cause cancer. Paraspeckles are small membraneless subnuclear structures with a diameter of 0.5-1 µm, and composed of lncRNA NEAT1 as the scaffold and three core RNA-binding proteins NONO, PSPC1, and PSF. Here, we show that AGGF1 is a key regulatory and structural component of paraspeckles that induces paraspeckle formation, forms an outside rim of paraspeckles, wraps around the NONO/PSF/PSPC1/NEAT1 core, and regulates the size and number of paraspeckles. AGGF1-paraspeckles are larger (>1 µm) than conventional paraspeckles. RNA-FISH in combination with immunostaining shows that AGGF1, NONO, and NEAT1_2 co-localize in 20.58% of NEAT1_2-positive paraspeckles. Mechanistically, AGGF1 interacts with NONO, PSF, and HNRNPK, and upregulates NEAT1_2, a longer, 23 kb NEAT1 transcript with a key role in regulation of paraspeckle size and number. RNA-immunoprecipitation shows that AGGF1 interacts with NEAT1, which may be another possible mechanism underlying the formation of AGGF1-paraspeckles. NEAT1_2 knockdown reduces the number and size of AGGF1-paraspeckles. Functionally, AGGF1 regulates alternative RNA splicing as it decreases the exon skipping/inclusion ratio in a CD44 model. AGGF1 is also localized in some nuclear foci without NEAT1 or NONO, suggesting that AGGF1 is an important liquid-liquid phase separation (LLPS) driver for other types of AGGF1-positive nuclear condensates (referred to as AGGF1-bodies). Our results identify a special type of AGGF1-coated paraspeckles and provide important insights into the formation, structure, and function of paraspeckles.


Assuntos
Paraspeckles , RNA Longo não Codificante , Núcleo Celular/metabolismo , Domínios Proteicos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
2.
Cell ; 135(6): 1017-27, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-19070573

RESUMO

Atrial fibrillation (AF) is the most common form of sustained clinical arrhythmia. We previously mapped an AF locus to chromosome 5p13 in an AF family with sudden death in early childhood. Here we show that the specific AF gene underlying this linkage is NUP155, which encodes a member of the nucleoporins, the components of the nuclear pore complex (NPC). We have identified a homozygous mutation, R391H, in NUP155 that cosegregates with AF, affects nuclear localization of NUP155, and reduces nuclear envelope permeability. Homozygous NUP155(-/-) knockout mice die before E8.5, but heterozygous NUP155(+/-) mice show the AF phenotype. The R391H mutation and reduction of NUP155 are associated with inhibition of both export of Hsp70 mRNA and nuclear import of Hsp70 protein. These human and mouse studies indicate that loss of NUP155 function causes AF by altering mRNA and protein transport and link the NPC to cardiovascular disease.


Assuntos
Fibrilação Atrial/genética , Morte Súbita Cardíaca , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Sequência de Aminoácidos , Animais , Feminino , Proteínas de Choque Térmico HSP72/genética , Proteínas de Choque Térmico HSP72/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Membrana Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Linhagem , Alinhamento de Sequência
3.
Support Care Cancer ; 31(10): 583, 2023 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-37728783

RESUMO

PURPOSE: To explore the correlation between financial toxicity, social support, and social functioning in post-chemotherapy breast cancer patients, as well as any possible interaction of financial toxicity and social support on social functioning. METHODS: Post-chemotherapy breast cancer patients admitted to the thyroid and breast surgery departments of three first-class general hospitals in East China from December 2020 to January 2022 were recruited by convenience sampling for a cross-sectional survey. The survey instruments included the general information form, the comprehensive scores for financial toxicity based on the patient-reported outcome measures (COST-PROM), the social roles and activity participation subscale from the patient-reported outcomes measurement system-breast-chemotherapy (PROMS-B-C) (score range: 8-40), and the social support subscale from PROMS-B-C (score range: 16-80). RESULTS: The results showed that low social functioning (low score) in post-chemotherapy breast cancer patients was positively correlated with high financial toxicity (low score) as well as poor economic resources (low score) and poor psychosocial responses (low score) (P<0.01) and negatively correlated with low economic expenditures (low score) (P<0.01); high social functioning (high score) was positively correlated with high social support (high score) (P<0.01). The interaction analysis results showed an additive interaction between financial toxicity and social support in social functioning. CONCLUSION: There was an additive interaction of financial toxicity and social support in the social functioning of post-chemotherapy breast cancer patients. Those patients with high financial toxicity and low social support are the most likely to benefit from relevant intervention measures compared to other breast cancer populations.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Estudos Transversais , Estresse Financeiro , Interação Social , Apoio Social
4.
FASEB J ; 35(5): e21465, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33788967

RESUMO

N6 -methyladenosine (m6A) methylation is the most prevalent RNA modification, and it emerges as an important regulatory mechanism of gene expression involved in many cellular and biological processes. However, the role of m6 A methylation in vascular development is not clear. The m6 A RNA methylation is regulated by dynamic interplay among methyltransferases, binding proteins, and demethylases. Mettl3 is a member of the mettl3-mettl14 methyltransferase complex, referred to as writers that catalyze m6A RNA methylation. Here, we used CRISPR-Cas9 genome editing to develop two lines of knockout (KO) zebrafish for mettl3. Heterozygous mettl3+/- KO embryos show defective vascular development, which is directly visible in fli-EGFP and flk-EGFP zebrafish. Alkaline phosphatase staining and whole mount in situ hybridization with cdh5, and flk markers demonstrated defective development of intersegmental vessels (ISVs), subintestinal vessels (SIVs), interconnecting vessels (ICVs) and dorsal longitudinal anastomotic vessels (DLAV) in both heterozygous mettl3+/- and homozygous mettl3-/- KO zebrafish embryos. Similar phenotypes were observed in zebrafish embryos with morpholino knockdown (KD) of mettl3; however, the vascular defects were rescued fully by overexpression of constitutively active AKT1. KD of METTL3 in human endothelial cells inhibited cell proliferation, migration, and capillary tube formation. Mechanistically, mettl3 KO and KD significantly reduced the levels of m6 A RNA methylation, and AKT phosphorylation (S473) by an increase in the expression of phosphatase enzyme PHLPP2 and reduction in the phosphorylation of mTOR (S2481), a member of the phosphatidylinositol 3-kinase-related kinase family of protein kinases. These data suggest that m6 A RNA methylation regulates vascular development via PHLPP2/mTOR-AKT signaling.


Assuntos
Adenosina/análogos & derivados , Embrião não Mamífero/citologia , Metiltransferases/metabolismo , Neovascularização Fisiológica , Fosfoproteínas Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Adenosina/química , Animais , Embrião não Mamífero/metabolismo , Metilação , Metiltransferases/genética , Fosfoproteínas Fosfatases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Serina-Treonina Quinases TOR/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
Langmuir ; 38(34): 10385-10391, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35980392

RESUMO

Microbe-catalyzed surface modification is a promising method for the production of special targeting nanomaterials. A bacterium-selective material can be obtained by investigating the microbe-catalyzed mineralization of proteins. Herein, a novel method was fabricated for the biosynthesis of FeS-decorated porphyrin-protein clusters (P-CA@BE) via E. coli (Escherichia coli)-catalyzed bio-Fe(III) reduction and bio-sulfidation of porphyrin (P), caffeic acid (CA), and protein [bovine serum albumin (BSA)] assemblies. The assembly (P-CA@BSA) was identified by spectroscopic methods. Next, the P-CA@BSA assembly was transferred into FeS-decorated porphyrin-protein clusters (P-CA@BE) catalyzed by E. coli. There are partial ß-folding proteins in P-CA@BE, which selectively recognize S. aureus (Staphylococcus aureus) and show different antibacterial properties against E. coli and S. aureus. Results demonstrate that the E. coli-catalyzed mineralization of the porphyrin-protein assembly is an effective method for the biosynthesis of S. aureus-sensitive metal-protein clusters.


Assuntos
Porfirinas , Staphylococcus aureus , Proteínas de Bactérias , Escherichia coli , Compostos Férricos/química , Compostos Ferrosos , Porfirinas/farmacologia , Soroalbumina Bovina/química
6.
Arterioscler Thromb Vasc Biol ; 41(11): 2756-2769, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34551592

RESUMO

Objective: Angiogenic factor AGGF1 (angiogenic factor with G-patch and FHA [Forkhead-associated] domain 1) promotes angiogenesis as potently as VEGFA (vascular endothelial growth factor A) and regulates endothelial cell (EC) proliferation, migration, specification of multipotent hemangioblasts and venous ECs, hematopoiesis, and vascular development and causes vascular disease Klippel-Trenaunay syndrome when mutated. However, the receptor for AGGF1 and the underlying molecular mechanisms remain to be defined. Approach and Results: Using functional blocking studies with neutralizing antibodies, we identified [alpha]5[beta]1 as the receptor for AGGF1 on ECs. AGGF1 interacts with [alpha]5[beta]1 and activates FAK (focal adhesion kinase), Src (proto-oncogene tyrosine-protein kinase), and AKT (protein kinase B). Functional analysis of 12 serial N-terminal deletions and 13 C-terminal deletions by every 50 amino acids mapped the angiogenic domain of AGGF1 to a domain between amino acids 604-613 (FQRDDAPAS). The angiogenic domain is required for EC adhesion and migration, capillary tube formation, and AKT activation. The deletion of the angiogenic domain eliminated the effects of AGGF1 on therapeutic angiogenesis and increased blood flow in a mouse model for peripheral artery disease. A 40-mer or 15-mer peptide containing the angiogenic domain blocks AGGF1 function, however, a 15-mer peptide containing a single amino acid mutation from -RDD- to -RGD- (a classical RGD integrin-binding motif) failed to block AGGF1 function. Conclusions: We have identified integrin [alpha]5[beta]1 as an EC receptor for AGGF1 and a novel AGGF1-mediated signaling pathway of [alpha]5[beta]1-FAK-Src-AKT for angiogenesis. Our results identify an FQRDDAPAS angiogenic domain of AGGF1 crucial for its interaction with [alpha]5[beta]1 and signaling.


Assuntos
Proteínas Angiogênicas/metabolismo , Células Endoteliais/metabolismo , Membro Posterior/irrigação sanguínea , Integrina alfa5beta1/metabolismo , Isquemia/metabolismo , Neovascularização Fisiológica , Células 3T3-L1 , Indutores da Angiogênese/farmacologia , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/farmacologia , Animais , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Feminino , Quinase 1 de Adesão Focal/metabolismo , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Integrina alfa5beta1/genética , Isquemia/tratamento farmacológico , Isquemia/genética , Isquemia/fisiopatologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , Quinases da Família src/metabolismo
7.
J Wound Ostomy Continence Nurs ; 49(3): 286-289, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35255076

RESUMO

BACKGROUND: Acetic acid is bacteriostatic or bactericidal to many gram-negative and gram-positive microorganisms, especially Pseudomonas. Nevertheless, it has also been found to possess cytotoxic effects in concentrations as low as 0.25% inhibiting the epithelialization process during wound healing. CASES: In this multiple case series, we present 2 cases of chronic traumatic leg wounds treated with gauze moistened with acetic acid (0.25%), which were covered with a securing dressing and compression stockinet. Both patients were told to apply gauze moistened with acetic acid (0.25%) twice daily. In both cases, the wound progressed to blue-green drainage and wet yellow slough tissue to near-complete beefy granulation tissue. At this point, acetic acid was replaced with collagen or petrolatum dressing until complete wound closure was achieved. The treatment of these wounds illustrated successful use of acetic acid for chronic wound care. CONCLUSION: Our experience with these cases suggests that appearance of blue-green wound drainage and wet yellow slough tissue is a reasonable indication for the use of gauze moistened with acetic acid (0.25%). Further research is needed to test the efficacy of these principles in guiding acetic acid use in wound care.


Assuntos
Ácido Acético , Cicatrização , Ácido Acético/farmacologia , Ácido Acético/uso terapêutico , Bandagens , Colágeno , Humanos
8.
Hum Mol Genet ; 28(17): 2937-2951, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31152168

RESUMO

KCNMA1 encodes the large-conductance Ca2+- and voltage-activated K+ (BK) potassium channel α-subunit, and pathogenic gain-of-function variants in this gene have been associated with a dominant form of generalized epilepsy and paroxysmal dyskinesia. Here, we genetically and functionally characterize eight novel loss-of-function (LoF) variants of KCNMA1. Genome or exome sequencing and the participation in the international Matchmaker Exchange effort allowed for the identification of novel KCNMA1 variants. Patch clamping was used to assess functionality of mutant BK channels. The KCNMA1 variants p.(Ser351Tyr), p.(Gly356Arg), p.(Gly375Arg), p.(Asn449fs) and p.(Ile663Val) abolished the BK current, whereas p.(Cys413Tyr) and p.(Pro805Leu) reduced the BK current amplitude and shifted the activation curves toward positive potentials. The p.(Asp984Asn) variant reduced the current amplitude without affecting kinetics. A phenotypic analysis of the patients carrying the recurrent p.(Gly375Arg) de novo missense LoF variant revealed a novel syndromic neurodevelopmental disorder associated with severe developmental delay, visceral and cardiac malformations, connective tissue presentations with arterial involvement, bone dysplasia and characteristic dysmorphic features. Patients with other LoF variants presented with neurological and developmental symptoms including developmental delay, intellectual disability, ataxia, axial hypotonia, cerebral atrophy and speech delay/apraxia/dysarthria. Therefore, LoF KCNMA1 variants are associated with a new syndrome characterized by a broad spectrum of neurological phenotypes and developmental disorders. LoF variants of KCNMA1 cause a new syndrome distinctly different from gain-of-function variants in the same gene.


Assuntos
Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Mutação com Perda de Função , Fenótipo , Alelos , Substituição de Aminoácidos , Fenômenos Eletrofisiológicos , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Humanos , Recém-Nascido , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/química , Masculino , Mutação de Sentido Incorreto , Linhagem , Domínios Proteicos , Domínios e Motivos de Interação entre Proteínas
9.
Nanotechnology ; 32(42)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34256355

RESUMO

The mysterious properties of individual U atoms on transition metal surfaces play indispensable parts in supplementing our understanding of uranium-transition metal systems, which are important subjects for both nuclear energy applications and fundamental scientific studies. By using scanning tunneling microscopy and density functional theory calculations, the adsorptions, reactions and electronic properties of individual U atoms on Cu(111), Ag(111), Au(111) and Ru(0001) surfaces were comparatively studied for the first time in this work. Upon the deposition of a small amount of U onto Cu(111) or Ag(111) at 8 K, individual U atoms show relatively high activity and can either be adsorbed on intact substrate surfaces or induce various surface vacancies surrounded by clusters of substrate atoms. By contrast, the majority of U atoms tend to dispersedly adsorb on intact surfaces of Au(111) and Ru(0001) rather than producing surface vacancies at the same temperature. In all cases, Kondo resonance manifested as asymmetric dip feature around Fermi energy is only observed in the differential tunneling conductance spectra of single U adatoms on Ag(111).

10.
J Fluoresc ; 31(2): 501-507, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33449251

RESUMO

Increasing acidity is an effective method for bacterial inactivation by inhibiting the synthesis of intracellular proteins at low pH. Photo-driven proton release probe can be used for the measurement of proton in hydrophobic condition. To develop fluorescent proton donor, two boron dipyrromethene derivatives (BDP-S and BDP-S2) were characterized by spectroscopic methods. Irradiation of BDP-S by white LED light resulted in efficient generation of acidic species with changes of fluorescence emission. The linear relationship between the pH value and the fluorescence intensity of BDP-S was obtained, indicating that BDP-S is a fluorescent visual proton donor. Light-induced antibacterial results indicate that BDP-S can significantly inhibit the growth of E. coli. The results prove that BDP-S is a very promising photoacid sterilant.


Assuntos
Antibacterianos/farmacologia , Compostos de Boro/farmacologia , Escherichia coli/efeitos dos fármacos , Corantes Fluorescentes/farmacologia , Prótons , Ácidos Sulfônicos/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Compostos de Boro/química , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Concentração de Íons de Hidrogênio , Testes de Sensibilidade Microbiana , Estrutura Molecular , Espectrometria de Fluorescência , Ácidos Sulfônicos/química
11.
Biochem J ; 477(9): 1683-1700, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32315024

RESUMO

Cardiac sodium channel Nav1.5 is associated with cardiac arrhythmias and heart failure. Protein ubiquitination is catalyzed by an E1-E2-E3 cascade of enzymes. However, the E1 enzyme catalyzing Nav1.5 ubiquitination is unknown. Here, we show that UBE1 and UBA6 are two E1 enzymes regulating Nav1.5 ubiquitination and expression. Western blot analysis and patch-clamping recordings showed that overexpression of UBE1 or UBA6 increased the ubiquitination of Nav1.5 and significantly reduced Nav1.5 expression and sodium current density, and knockdown of UBE1 or UBA6 expression significantly increased Nav1.5 expression and sodium current density in HEK293/Nav1.5 cells. Similar results were obtained in neonatal cardiomyocytes. Bioinformatic analysis predicted two ubiquitination sites at K590 and K591. Mutations of K590 and K591 to K590A and K591A abolished the effects of overexpression or knockdown of UBE1 or UBA6 on Nav1.5 expression and sodium current density. Western blot analysis showed that the effects of UBE1 or UBA6 overexpression on the ubiquitination and expression of Nav1.5 were abolished by knockdown of UBC9, a putative E2 enzyme reported for Nav1.5 ubiquitination by us. Interestingly, real-time RT-PCR analysis showed that the expression level of UBE1, but not UBA6, was significantly up-regulated in ventricular tissues from heart failure patients. These data establish UBE1 and UBA6 as the E1 enzymes involved in Nav1.5 ubiquitination, and suggest that UBE1 and UBA6 regulate ubiquitination of Nav1.5 through UBC9. Our study is the first to reveal the regulatory role of the UBE1 or UBA6 E1 enzyme in the ubiquitination of an ion channel and links UBE1 up-regulation to heart failure.


Assuntos
Canais de Sódio/metabolismo , Enzimas Ativadoras de Ubiquitina , Ubiquitinação/fisiologia , Arritmias Cardíacas/metabolismo , Células HEK293 , Humanos , Mutação , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Sódio/metabolismo , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo
12.
BMC Health Serv Res ; 21(1): 742, 2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-34315453

RESUMO

BACKGROUND: The incidence of Sudden Sensorineural Hearing Loss (SSNHL) is increasing and tends to occur at a young age. The patient's disease experience during treatment is related to their physical and mental health. Effective coping styles such as proactively solving problems and asking for help will alleviate the patients' psychological symptoms and improve their quality of life. AIMS AND OBJECTIVES: To explore the illness experience and coping styles of young and middle-aged patients with SSNHL (age: 18-64 years), clarify the relationship between the two, and understand the psychological state and needs of the patients. METHODS: The purposive and maximum difference sampling method was used to conduct semi-structured interviews with 23 young and middle-aged SSNHL patients. The interview data were analyzed by Colaizzi's seven-step analysis. RESULTS: The illness experience of young and middle-aged SSNHL patients was complex, including symptoms during the onset of deafness and emotional experience before and after diagnosis. The coping styles of young and middle-aged SSNHL patients were active and diverse, including active acquisition of information, change in living habits, and seeking the care and attention of medical staff. Illness experience and coping style influence each other: good illness experience leads to active coping styles; active coping style results in good illness experience. CONCLUSIONS: The illness experience of young and middle-aged SSNHL patients includes not only physical symptoms, but also changes in psychological and emotional reactions. Good illness experience can lead patients to adopt active coping style. Active and effective coping styles, such as positive acquisition of information, change in living habits and seeking care and help, can improve patients' illness experience.


Assuntos
Perda Auditiva Neurossensorial , Perda Auditiva Súbita , Adaptação Psicológica , Adolescente , Adulto , Perda Auditiva Neurossensorial/terapia , Perda Auditiva Súbita/terapia , Humanos , Saúde Mental , Pessoa de Meia-Idade , Qualidade de Vida , Adulto Jovem
13.
J Biol Chem ; 294(11): 3881-3898, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30655286

RESUMO

Coronary artery disease (CAD) is the leading cause of death worldwide. Long noncoding RNAs (lncRNAs) are a class of noncoding transcripts of > 200 nucleotides and are increasingly recognized as playing functional roles in physiology and disease. ANRIL is an lncRNA gene mapped to the chromosome 9p21 genetic locus for CAD identified by the first series of genome-wide association studies (GWAS). However, ANRIL's role in CAD and the underlying molecular mechanism are unknown. Here, we show that the major ANRIL transcript in endothelial cells (ECs) is DQ485454 with a much higher expression level in ECs than in THP-1 monocytes. Of note, DQ485454 expression was down-regulated in CAD coronary arteries compared with non-CAD arteries. DQ485454 overexpression significantly reduced monocyte adhesion to ECs, transendothelial monocyte migration (TEM), and EC migration, which are critical cellular processes involved in CAD initiation, whereas siRNA-mediated ANRIL knockdown (KD) had the opposite effect. Microarray and follow-up quantitative RT-PCR analyses revealed that the ANRIL KD down-regulated expression of AHNAK2, CLIP1, CXCL11, ENC1, EZR, LYVE1, WASL, and TNFSF10 genes and up-regulated TMEM100 and TMEM106B genes. Mechanistic studies disclosed that overexpression of CLIP1, EZR, and LYVE1 reversed the effects of ANRIL KD on monocyte adhesion to ECs, TEM, and EC migration. These findings indicate that ANRIL regulates EC functions directly related to CAD, supporting the hypothesis that ANRIL is involved in CAD pathogenesis at the 9p21 genetic locus and identifying a molecular mechanism underlying lncRNA-mediated regulation of EC function and CAD development.


Assuntos
Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , RNA Longo não Codificante/metabolismo , Regulação para Cima , Proteínas de Transporte Vesicular/metabolismo , Movimento Celular , Células Cultivadas , Proteínas do Citoesqueleto/genética , Humanos , Proteínas Associadas aos Microtúbulos/genética , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas de Transporte Vesicular/genética
14.
FASEB J ; 33(7): 8555-8564, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30991833

RESUMO

Vascular hyperpermeability caused by distorted endothelial cell-cell junctions is associated with the no-reflow phenomenon after opening of the occluded vessels in patients with coronary artery disease (CAD), the leading cause of death worldwide. Coronary no-reflow is observed in ∼30% of CAD patients after percutaneous coronary stenting and is associated with a worse prognosis at follow-up and a higher incidence of death. However, limited tools are available to control vascular hyperpermeability and no-reflow. Losartan, an angiotensin II (Ang II) receptor blocker acting on the Ang II type-1 receptor (AT1R) subtype, is a prescription drug for treating hypertension. Here we show that in a murine model of ischemia and reperfusion (I/R), losartan blocked vascular hyperpermeability and decreased infarct size, hemorrhages, edema, and inflammation. Mechanistically, losartan-mediated inhibition of vascular hyperpermeability is mediated by the inhibition of phosphorylation of Src and vascular endothelial cadherin (VE-cadherin), which increases VEGF receptor 2 (VEGFR2)-Src-VE-cadherin complex formation, resulting in increased cell surface VE-cadherin and inhibition of vascular hyperpermeability. On the other hand, hypoxia and reoxygenation increased the phosphorylation levels of Src and VE-cadherin and reduced the formation of the VEGFR2-Src-VE-cadherin complex, which led to reduced cell surface VE-cadherin and increased vascular hyperpermeability; all were inhibited by losartan. These data suggest that losartan may be used for blocking vascular hyperpermeability associated with I/R.-Li, Y., Yao, Y., Li, J., Chen, Q., Zhang, L., Wang, Q. K. Losartan protects against myocardial ischemia and reperfusion injury via vascular integrity preservation.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Losartan/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Substâncias Protetoras/farmacologia , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/metabolismo , Coração/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
FASEB J ; 33(8): 8878-8891, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31034774

RESUMO

Atrial fibrillation (AF) affects >30 million individuals worldwide. However, no genetic mutation from human patients with AF has been linked to inflammation. Here, we show that AF-associated human variant p.Ile138Thr in natriuretic peptide A (NPPA) encoding the atrial natriuretic peptide (ANP) causes inflammation, fibroblast activation, atrial fibrosis, and AF in knock-in (KI) rats. Variant p.Ile138Thr inhibits the interaction between ANP and its receptor natriuretic peptide receptor A and reduces intracellular cGMP levels. RNA sequencing and follow-up analyses showed that mutant ANP (mANP) activates multiple innate immunity pathways, including TNF-α, NF-κB, and IL-1ß signaling. mANP induces differentiation of cardiac fibroblasts (CFs) to myofibroblasts and promotes CF proliferation and fibrosis. These results suggest that NPPA variant p.Ile138Thr causes AF by activating TNF-α, NF-κB, and IL-1ß signaling, inflammation, and fibrosis. Multiple computational programs suggest that p.Ile138Thr is damaging or deleterious. Based on the 2015 American College of Medical Genetics and Genomics Standards and Guidelines, p.Ile138Thr can be classified as a likely pathogenic variant. Variant p.Ile138Thr was found only in Asian people in the Genome Aggregation Database and Exome Aggregation Consortium database at an averaged frequency of 0.026%. An estimated 1.15 million Asian people carry the variant and might be at risk of AF. The KI rats may provide an inflammation-based, genetic animal model for AF valuable for testing anti-inflammation or other therapies for AF.-Cheng, C., Liu, H., Tan, C., Tong, D., Zhao, Y., Liu, X., Si, W., Wang, L., Liang, L., Li, J., Wang, C., Chen, Q., Du, Y., Wang, Q. K., Ren, X. Mutation in NPPA causes atrial fibrillation by activating inflammation and cardiac fibrosis in a knock-in rat model.


Assuntos
Fibrilação Atrial/genética , Fator Natriurético Atrial/genética , Interleucina-1beta/metabolismo , Mutação de Sentido Incorreto , Fator de Necrose Tumoral alfa/metabolismo , Animais , Fibrilação Atrial/patologia , Células Cultivadas , GMP Cíclico/metabolismo , Feminino , Fibrose , Células HEK293 , Humanos , Imunidade Inata , Masculino , Miofibroblastos/metabolismo , Miofibroblastos/patologia , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
16.
Anticancer Drugs ; 31(10): 1046-1056, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32649369

RESUMO

Novel manganese complex, PdpaMn ([(Pdpa)MnCl2]), was developed to induce apoptosis in breast cancer cells. The impact of phosphoinositide-(3)-kinase pathway onto fatty acid synthase (FASN) has an effect on cellular metabolism in breast cancer. However, reverse actions from FASN towards PI3K/Akt are still indefinable. Perhaps, loss of FASN could regulate glycolysis. Previously we established that PdpaMn inhibits FASN and involve in mitochondrial function. This study investigated the activity of PdpaMn on glycolysis and its mechanism. PdpaMn was used to suppress FASN expression in tumor. Expression of ATP and lactic acid level was measured to investigate the glycolysis variance in cells and animals. MCF-7 and 4T1 cells were treated with G28UCM, an inhibitor of FASN and PdpaMn, western blotting to detect PI3K/Akt signaling pathway. The capacity of proliferation was investigated by western blotting and immunohistochemistry. PdpaMn selectively inhibits cancer cells and tumor growth but also block FASN expression and suppresses the content of free fatty acid. Lactate dehydrogenase (LDHA) protein level was down-regulated as G28UCM and PdpaMn inhibited FASN, glucose transporter (Glut1), and pyruvate kinase (PKM2) proteins level were not affected. PI3K, p-Akt in the experimental group evidently declined compared to the control group. Proliferation was suppressed in FASN-arbitrated glycolysis. Our study supports the hypothesis that loss of FASN by PdpaMn suppressed glycolysis via down-regulating PI3K/Akt signaling pathway revealing the direct link between FASN and glycolysis. The results have paved the way to unravel the mechanisms of FASN and mitochondrial will be useful for designing novel co-targeting strategies for breast cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Ácido Graxo Sintase Tipo I/metabolismo , Glicólise/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , Animais , Antineoplásicos/química , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Ácido Graxo Sintase Tipo I/antagonistas & inibidores , Feminino , Humanos , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Camundongos Endogâmicos BALB C , Compostos Organometálicos/química , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/química , Pirróis/química , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
RNA Biol ; 17(10): 1391-1401, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32602777

RESUMO

Each gene typically has multiple alternatively spliced transcripts. Different transcripts are assumed to play a similar biological role; however, some transcripts may simply lose their function due to loss of important functional domains. Here, we show that two different transcripts of lncRNA gene ANRIL associated with coronary artery disease (CAD) play antagonizing roles against each other. We previously reported that DQ485454, the short transcript, is downregulated in coronary arteries from CAD patients, and reduces monocyte adhesion to endothelial cells (ECs) and transendothelial monocyte migration (TEM). Interestingly, the longest transcript NR_003529 is significantly upregulated in coronary arteries from CAD patients. Overexpression of ANRIL transcript NR_003529 increases monocyte adhesion to ECs and TEM, whereas knockdown of NR_003529 expression reduces monocyte adhesion to ECs and TEM. Much more dramatic effects were observed for the combination of overexpression of NR_003529 and knockdown of DQ485454 or the combination of knockdown of NR_003529 and overexpression of DQ485454. The antagonizing effects of ANRIL transcripts NR_003529 and DQ485454 were associated with their opposite effects on expression of downstream target genes EZR, CXCL11 or TMEM106B. Our results demonstrate that different transcripts of lncRNA can exert antagonizing effects on biological functions, thereby providing important insights into the biology of lncRNA. The data further support the hypothesis that ANRIL is the causative gene at the 9p21 CAD susceptibility locus.


Assuntos
Processamento Alternativo , Doença da Artéria Coronariana/genética , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , Biomarcadores , Adesão Celular/genética , Doença da Artéria Coronariana/diagnóstico , Doença da Artéria Coronariana/metabolismo , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Suscetibilidade a Doenças , Técnicas de Silenciamento de Genes , Humanos , Monócitos/metabolismo , Monócitos/patologia , Isoformas de RNA , Migração Transendotelial e Transepitelial/genética
18.
J Mol Cell Cardiol ; 129: 79-91, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30772377

RESUMO

Voltage-gated sodium channel Nav1.5 is critical for generation and conduction of cardiac action potentials. Mutations and expression level changes of Nav1.5 are associated with cardiac arrhythmias and sudden death. The ubiquitin (Ub) conjugation machinery utilizes three enzyme activities, E1, E2, and E3, to regulate protein degradation. Previous studies from us and others showed that Nedd4-2 acts as an E3 ubiquitin-protein ligase involved in ubiquitination and degradation of Nav1.5, however, more key regulators remain to be identified. In this study, we show that UBC9, a SUMO-conjugating enzyme, regulates ubiquitination and degradation of Nav1.5. Overexpression of UBC9 significantly decreased Nav1.5 expression and reduced sodium current densities, whereas knockdown of UBC9 expression significantly enhanced Nav1.5 expression and increased sodium current densities, in both HEK293 cells and primary neonatal cardiomyocytes. Overexpression of UBC9 increased ubiquitination of Nav1.5, and proteasome inhibitor MG132 blocked the effect of UBC9 overexpression on Nav1.5 degradation. Co-immunoprecipitation showed that UBC9 interacts with Nedd4-2. UBC9 with mutation C93S, which suppresses SUMO-conjugating activity of UBC9, was as active as wild type UBC9 in regulating Nav1.5 levels, suggesting that UBC9 regulates Nav1.5 expression levels in a SUMOylation-independent manner. Our findings thus identify a key structural element of the ubiquitin-conjugation machinery for Nav1.5 and provide important insights into the regulatory mechanism for ubiquitination and turnover of Nav1.5.


Assuntos
Ativação do Canal Iônico , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Proteólise , Sódio/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação , Animais , Animais Recém-Nascidos , Regulação para Baixo/genética , Células HEK293 , Células HeLa , Humanos , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Ratos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Enzimas de Conjugação de Ubiquitina/genética , Regulação para Cima/genética
19.
Hum Mutat ; 40(7): 879-885, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30920082

RESUMO

X-linked hypophosphatemia (XLH) is the most common hereditary rickets, caused by mutations in PHEX encoding the phosphate regulating endopeptidase homolog X-linked. Here, we report a nonsense variant in exon 11 of PHEX (c.1209G>A p.Trp403*) cosegregating with XLH in a Chinese family with a LOD score of 2.70. Real-time reverse transcription polymerase chain reaction analysis demonstrated that p.Trp403* variant did not cause nonsense-mediated mRNA decay (NMD), but significantly increased the expression level of FGF23 mRNA in the patients. Interestingly, p.Trp403* significantly reduced phosphorylation of p38 mitogen-activated protein kinase (MAPK) but not ERK1/2. Moreover, overexpression of FGF23 significantly decreased phosphorylation of p38 MAPK, whereas knockdown of FGF23 by siRNA significantly increased phosphorylation of p38 MAPK. These data suggest that p.Trp403* may not function via an NMD mechanism, and instead causes XLH via a novel signaling mechanism involving PHEX, FGF23, and p38 MAPK. This finding provides important insights into genetic and molecular mechanisms for the pathogenesis of XLH.


Assuntos
Raquitismo Hipofosfatêmico Familiar/genética , Fatores de Crescimento de Fibroblastos/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Mutação Puntual , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Códon sem Sentido , Raquitismo Hipofosfatêmico Familiar/metabolismo , Feminino , Fator de Crescimento de Fibroblastos 23 , Predisposição Genética para Doença , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Linhagem , Fosforilação , Regulação para Cima
20.
Hum Mutat ; 40(3): 310-325, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30488537

RESUMO

Atrial fibrillation (AF) is the most common cardiac arrhythmia. Here, we show the identification and functional characterization of one AF-associated mutation p.Arg399Cys in lamin A/C. Co-immunoprecipitation and GST pull-down assays demonstrate that lamin A/C interacts with NUP155, which is a nucleoporin and causes AF when mutated. Lamin A/C mutation p.Arg399Cys impairs the interaction between lamin A/C and NUP155, and increases extractability of NUP155 from the nuclear envelope (NE). Mutation p.Arg399Cys leads to aggregation of lamin A/C in the nucleus, although it does not impair the integrity of NE upon cellular stress. Mutation p.Arg399Cys inhibits the export of HSP70 mRNA and the nuclear import of HSP70 protein. Electrophysiological studies show that mutation p.Arg399Cys decreases the peak cardiac sodium current by decreasing the cell surface expression level of cardiac sodium channel Nav 1.5, but does not affect IKr potassium current. In conclusion, our results indicate that lamin A/C mutation p.Arg399Cys weakens the interaction between nuclear lamina (lamin A/C) and the nuclear pore complex (NUP155), leading to the development of AF. The findings provide a novel molecular mechanism for the pathogenesis of AF.


Assuntos
Fibrilação Atrial/genética , Lamina Tipo A/genética , Mutação/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Sequência de Bases , Células HEK293 , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Humanos , Ativação do Canal Iônico , Carioferinas/metabolismo , Lamina Tipo A/química , Camundongos , Membrana Nuclear/metabolismo , Ligação Proteica , Transporte Proteico , RNA/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Canais de Sódio/metabolismo , Estresse Fisiológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA