Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Injury ; 55(10): 111755, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39098255

RESUMEN

OBJECTIVE: The clinical effects of artificial dermis in treating skin and soft tissue defects accompanied by bone or tendon exposure were assessed. APPROACH: A retrospective analysis was conducted on the clinical data of 45 cases of skin and soft tissue defects accompanied by bone or tendon exposure admitted to the trauma surgery department of Fujian Provincial Hospital between February 2018 and August 2020. They were divided into the artificial dermis and control groups. The wound was assessed using the Vancouver Scar Scale (mVSS), and the postoperative visual analogue scale (VAS) scores were recorded at 3, 6, 9, and 12 months after surgery. At the 12-month after surgery, skin sensation recovery was evaluated using the criteria of the British Medical Research Council (BMRC). RESULTS: The cases included 26 males and 19 females, aged 50 to 85 years. All patients were followed up for an average of 13.8 months (range: 12-18 months). Compared with controls, the wound healing time of the observation group was longer (35.8 ± 10.6 vs. 28.5 ± 4.8, P = 0.007), without significant differences for the number of operations and length of hospitalization. The mVSS scores were not different between groups (Pgroup = 0.294), but the scores decreased with time (Ptime < 0.001), and the group×time interaction was significant (Pinteraction < 0.001). Similarly, the VAS scores were not different between groups (Pgroup = 0.667), but the scores decreased with time (Ptime < 0.001); the group×time interaction was not significant (Pinteraction = 0.274). At the 12-month mark following the operation, in the artificial dermis group, the MCRR score was S3+ in 23 patients, while it ranged from S0 to S3 in two patients; in the control group, S3+ was observed in 17 patients, and S0-S3 in three (P = 0.815). CONCLUSION: Artificial dermis treatment is considered a safe and effective alternative therapy for patients with skin and soft tissue defects accompanied by bone or tendon exposure who cannot tolerate or are unwilling to undergo autologous skin flap transplantation. It offers the advantages of minimal donor site trauma, simplicity in operation, and favorable postoperative functional recovery.

2.
J Orthop Surg (Hong Kong) ; 32(2): 10225536241267725, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39058795

RESUMEN

PURPOSE: The purpose of the present work was to assess the specific effects and underlying mechanisms of Daprodustat (GSK1278863) on skeletal muscle injury induced by ischemia reperfusion (I/R). METHODS: C57BL/6 mice were randomized into the skeletal muscle I/R injury (I/R), Daprodustat (GSK1278863) pretreatment and I/R (I/R + GSK) and sham operation (Sham) groups. The skeletal muscle I/R injury model was established by placing an orthodontic rubber band at the left hip joint for 3 h and releasing it for 3 h. H&E staining, wet weight/dry weight ratio assessment, TUNEL assay, ELISA, qRT-PCR and immunoblot were utilized to assess the effects of Daprodustat. RESULTS: Daprodustat pretreatment significantly ameliorated apoptosis in skeletal muscle cells, reduced oxidative damage and suppressed inflammatory cytokines. Mechanistically, Daprodustat positively affected NF-κB signaling activation. CONCLUSION: These data demonstrated that Daprodustat may provide a potential clinical approach for preventing or treating skeletal muscle injury induced by I/R.


Asunto(s)
Ratones Endogámicos C57BL , Músculo Esquelético , Daño por Reperfusión , Animales , Daño por Reperfusión/prevención & control , Daño por Reperfusión/etiología , Ratones , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Apoptosis/efectos de los fármacos , Masculino , Modelos Animales de Enfermedad , FN-kappa B/metabolismo , Distribución Aleatoria
3.
Arterioscler Thromb Vasc Biol ; 43(12): 2348-2368, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37881938

RESUMEN

BACKGROUND: Hemangioblasts are mesoderm-derived multipotent stem cells for differentiation of all hematopoietic and endothelial cells in the circulation system. However, the underlying molecular mechanism is poorly understood. METHODS: CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (type II CRISPR RNA-guided endonuclease) editing was used to develop aggf1-/- and emp2-/- knockout zebra fish. Whole-mount in situ hybridization and transgenic Tg(gata1-EGFP [enhanced green fluorescent protein]), Tg(mpx-EGFP), Tg(rag2-DsRed [discosoma sp. red fluorescent protein]), Tg(cd41-EGFP), Tg(kdrl-EGFP), and Tg(aggf1-/-;kdrl-EGFP) zebra fish were used to examine specification of hemangioblasts and hematopoietic stem and progenitor cells (HSPCs), hematopoiesis, and vascular development. Quantitative real-time polymerase chain reaction and Western blot analyses were used for expression analysis of genes and proteins. RESULTS: Knockout of aggf1 impaired specification of hemangioblasts and HSPCs, hematopoiesis, and vascular development in zebra fish. Expression of npas4l/cloche-the presumed earliest marker for hemangioblast specification-was significantly reduced in aggf1-/- embryos and increased by overexpression of aggf1 in embryos. Overexpression of npas4l rescued the impaired specification of hemangioblasts and HSPCs and development of hematopoiesis and intersegmental vessels in aggf1-/- embryos, placing aggf1 upstream of npas4l in hemangioblast specification. To identify the underlying molecular mechanism, we identified emp2 as a key aggf1 downstream gene. Similar to aggf1, emp2 knockout impaired the specification of hemangioblasts and HSPCs, hematopoiesis, and angiogenesis by increasing the phosphorylation of ERK1/2 (extracellular signal-regulated protein kinase 1/2). Mechanistic studies showed that aggf1 knockdown and knockout significantly decreased the phosphorylated levels of mTOR (mammalian target of rapamycin) and p70 S6K (ribosomal protein S6 kinase), resulting in reduced protein synthesis of Emp2 (epithelial membrane protein 2), whereas mTOR activator MHY1485 (4,6-dimorpholino-N-(4-nitrophenyl)-1,3,5-triazin-2-amine) rescued the impaired specification of hemangioblasts and HSPCs and development of hematopoiesis and intersegmental vessels and reduced Emp2 expression induced by aggf1 knockdown. CONCLUSIONS: These results indicate that aggf1 acts at the top of npas4l and becomes the earliest marker during specification of hemangioblasts. Our data identify a novel signaling axis of Aggf1 (angiogenic factor with G-patch and FHA domain 1)-mTOR-S6K-ERK1/2 for specification of hemangioblasts and HSPCs, primitive and definitive hematopoiesis, and vascular development. Our findings provide important insights into specification of hemangioblasts and HSPCs essential for the development of the circulation system.


Asunto(s)
Hemangioblastos , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Hemangioblastos/metabolismo , Hematopoyesis/genética , Mamíferos , Serina-Treonina Quinasas TOR/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
5.
Obesity (Silver Spring) ; 31(1): 123-138, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36504350

RESUMEN

OBJECTIVE: Genetic variants in ninjurin-2 (NINJ2; nerve injury-induced protein 2) confer risk of ischemic strokes and coronary artery disease as well as endothelial activation and inflammation. However, little is known about NINJ2's in vivo functions and underlying mechanisms. METHODS: The phenotypes of NINJ2 knockout mice were analyzed, and mechanisms of NINJ2 that regulate body weight, insulin resistance, and glucose homeostasis and lipogenesis were investigated in vivo and in vitro. RESULTS: This study found that mice lacking NINJ2 showed impaired adipogenesis, increased insulin resistance, and abnormal glucose homeostasis, all of which are risk factors for strokes and coronary artery disease. Mechanistically, NINJ2 directly interacts with insulin receptor/insulin-like growth factor 1 receptor (INSR/IGF1R), and NINJ2 knockdown can block insulin-induced mitotic clonal expansion during preadipocyte differentiation by inhibiting protein kinase B/extracellular signal-regulated kinase (AKT/ERK) signaling and by decreasing the expression of key adipocyte transcriptional regulators CCAAT/enhancer-binding protein ß (C/EBP-ß), C/EBP-α, and peroxisome proliferator-activated receptor γ (PPAR-γ). Furthermore, the interaction between NINJ2 and INSR/IGF1R is needed for maintaining insulin sensitivity in adipocytes and muscle via AKT and glucose transporter type 4. Notably, adenovirus-mediated NINJ2 overexpression can ameliorate diet-induced insulin resistance in mice. CONCLUSIONS: In conclusion, these findings reveal NINJ2 as an important new facilitator of insulin receptors, and the authors propose a unique regulatory mechanism between insulin signaling, adipogenesis, and insulin resistance.


Asunto(s)
Moléculas de Adhesión Celular Neuronal , Resistencia a la Insulina , Animales , Ratones , Células 3T3-L1 , Adipogénesis/genética , Diferenciación Celular/genética , Enfermedad de la Arteria Coronaria , Glucosa/metabolismo , Insulina , Resistencia a la Insulina/genética , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Moléculas de Adhesión Celular Neuronal/genética
6.
Circ Genom Precis Med ; 15(5): e003603, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35895078

RESUMEN

BACKGROUND: Idiopathic ventricular tachycardia (VT) occurs in structurally normal hearts and accounts for a significant number of all types of VT. The genome-wide association study is the most effective strategy for identifying novel genetic variants for common diseases. However, no genome-wide association study has been reported for idiopathic VT. METHODS: We conducted the first genome-wide association study for idiopathic VT in the Chinese Han population using a discovery population with 246 cases and 648 controls and a replication population with 222 cases and >4072 controls. Candidate VT genes were functionally characterized in zebrafish. Real-time RT-PCR analysis was used to determine the effects of candidate genes on expression of ion channels and regulators. Patch-clamping was used to record L-type calcium current from neonatal rat cardiomyocytes with overexpression of candidate genes. RESULTS: We identified 4 significant loci represented by rs78960694 (minor allele frequency [MAF]=5.02% in cases and 1.84% in controls; P=4.30×10-12, odds ratio [OR]=3.91) and rs2229095 (MAF=3.25% in cases and 1.63% in controls; P=1.02×10-7, OR=3.44) near and in CCR7, respectively, rs68126098 in NELL1 (MAF=40.98% in cases and 32.07% in controls; P=2.40×10-8, OR=1.53), rs2390325 between PKN2 and LMO4 (MAF=21.19% in cases and 15.12% in controls; P=1.92×10-7, OR=1.62), and rs270065 in CSMD1 (MAF=33.63% in cases and 40.25% in controls; P=9.51×10-7, OR=0.69). Note that the associations of idiopathic VT for CCR7 variant rs78960694 and NELL1 variant rs68126098 reach genome-wide significance (P<5.00×10-8). Overexpression of either PKN2 or CCR7 increased the heart rate in zebrafish, and enhanced expression of CACNA1C, RYR2, or NOS1AP in zebrafish embryos, HEK293, and AC16 cardiomyocytes. Overexpression of either PKN2 or CCR7 significantly increased L-type Ca2+ current density. CONCLUSIONS: The first genome-wide association study identifies 4 novel loci and 2 risk genes (PKN2 and CCR7) for idiopathic VT. These findings identify new molecular determinants for cardiac calcium homeostasis and rhythm maintenance and provide novel targets for diagnosis and treatment for idiopathic VT.


Asunto(s)
Calcio , Proteína Quinasa C , Taquicardia Ventricular , Animales , Humanos , Ratas , Proteínas Adaptadoras Transductoras de Señales/genética , Calcio/metabolismo , Canales de Calcio Tipo L , Células HEK293 , Homeostasis , Proteínas con Dominio LIM/metabolismo , Receptores CCR7/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Taquicardia Ventricular/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteína Quinasa C/genética
7.
FASEB J ; 36(6): e22366, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35608889

RESUMEN

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.


Asunto(s)
Paraspeckles , ARN Largo no Codificante , Núcleo Celular/metabolismo , Dominios Proteicos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
8.
Biochim Biophys Acta Mol Basis Dis ; 1868(9): 166429, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35533905

RESUMEN

Our earlier studies identified MOG1 as a Nav1.5-binding protein that promotes Nav1.5 intracellular trafficking to plasma membranes. Genetic studies have identified MOG1 variants responsible for cardiac arrhythmias. However, the physiological functions of MOG1 in vivo remain incompletely characterized. In this study, we generated Mog1 knockout (Mog1-/-) mice. Mog1-/- mice did not develop spontaneous arrhythmias at the baseline, but exhibited a prolongation of QRS duration. Mog1-/- mice treated with isoproterenol (ISO), but not with flecainide, exhibited an increased risk of arrhythmias and even sudden death. Mog1-/- mice had normal cardiac morphology, however, LV systolic dysfunction was identified and associated with an increase in ventricular fibrosis. Whole-cell patch-clamping and Western blotting analysis clearly demonstrated the normal cardiac expression and function of Nav1.5 in Mog1-/- mice. Further RNA-seq and iTRAQ analysis identified critical pathways and genes, including extracellular matrix (Mmp2), gap junction (Gja1), and mitochondrial components that were dysregulated in Mog1-/- mice. RT-qPCR, Western blotting, and immunofluorescence assays revealed reduced cardiac expression of Gja1 in Mog1-/- mice. Dye transfer assays confirmed impairment of gap-junction function; Cx43 gap-junction enhancer ZP123 decreased arrhythmia inducibility in ISO-treated Mog1-/- mice. Transmission electron microscopy analysis revealed abnormal sarcomere ultrastructure and altered mitochondrial morphology in Mog1-/- mice. Mitochondrial dynamics was found to be disturbed, and associated with a trend toward increased mitochondrial fusion in Mog1-/- mice. Meanwhile, the level of ATP supply was increased in the hearts of Mog1-/- mice. These results indicate that MOG1 plays an important role in cardiac electrophysiology and cardiac contractile function.


Asunto(s)
Conexina 43 , Canal de Sodio Activado por Voltaje NAV1.5 , Proteína de Unión al GTP ran , Animales , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/genética , Conexina 43/genética , Conexina 43/metabolismo , Fibrosis , Isoproterenol/efectos adversos , Ratones , Ratones Noqueados , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Proteína de Unión al GTP ran/genética
9.
Acta Physiol (Oxf) ; 235(1): e13800, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35156297

RESUMEN

AIM: Loss-of-function KCNMA1 variants cause Liang-Wang syndrome (MIM #618729), a newly identified multiple malformation syndrome with a broad spectrum of developmental and neurological phenotypes. However, the full spectrum of clinical features and underlying pathogenic mechanisms need full elucidation. METHODS: Exome sequencing was used to identify pathogenic variants. Patch-clamp recordings were performed to access the effects of KCNMA1 variants on BK channels. Total and membrane protein expression levels of BK channels were characterized using Western blotting. RESULTS: We report identification and functional characterization of two new de novo loss-of-function KCNMA1 variants p.(A172T) and p.(A314T) with characteristics of Liang-Wang syndrome. Variant p.(A172T) is associated with developmental delay, cognitive impairment and ataxia. Mechanistically, p.(A172T) abolishes BK potassium current, inhibits Mg2+ -dependent gating, but shifts conductance-voltage (G-V) curves to more positive potentials when complexed with WT channels. Variant p.(A314T) is associated with developmental delay, intellectual disability, cognitive impairment, mild ataxia and generalized epilepsy; suppresses BK current amplitude; and shifts G-V curves to more positive potentials when expressed with WT channels. In addition, two new patients with previously reported gain-of-function variants p.(N536H) and p.(N995S) are found to show epilepsy and paroxysmal dyskinesia as reported previously, but also exhibit additional symptoms of cognitive impairment and dysmorphic features. Furthermore, variants p.(A314T) and p.(N536H) reduced total and membrane levels of BK proteins. CONCLUSION: Our findings identified two new loss-of-function mutations of KCNMA1 associated with Liang-Wang syndrome, expanded the spectrum of clinical features associated with gain-of-function KCNMA1 variants and emphasized the overlapping features shared by gain-of-function and loss-of-function mutations.


Asunto(s)
Epilepsia , Discapacidad Intelectual , Ataxia/genética , Epilepsia/genética , Epilepsia/patología , Humanos , Discapacidad Intelectual/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Fenotipo
10.
Heart Rhythm ; 19(3): 478-489, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34843967

RESUMEN

BACKGROUND: Mutations in cardiac sodium channel Nav1.5 cause Brugada syndrome (BrS). MOG1 is a chaperone that binds to Nav1.5, facilitates Nav1.5 trafficking to the cell surface, and enhances the amplitude of sodium current INa. OBJECTIVE: The purpose of this study was to identify structural elements involved in MOG1-Nav1.5 interaction and their relevance to the pathogenesis of BrS. METHODS: Systematic analyses of large deletions, microdeletions, and point mutations, and glutathione S-transferases pull-down, co-immunoprecipitation, cell surface protein quantification, and patch-clamping of INa were performed. RESULTS: Large deletion analysis defined the MOG1-Nav1.5 interaction domain to amino acids S476-H585 of Nav1.5 Loop I connecting transmembrane domains I and II. Microdeletion and point mutation analyses further defined the domain to F530T531F532R533R534R535. Mutations F530A, F532A, R533A, and R534A, but not T531A and R535A, significantly reduced MOG1-Nav1.5 interaction and eliminated MOG1-enhanced INa. Mutagenesis analysis identified D24, E36, D44, E53, and E101A of MOG1 as critical residues for interaction with Nav1.5 Loop I. We then characterized 3 mutations at the MOG1-Nav1.5 interaction domain: p.F530V, p.F532C, and p.R535Q reported from patients with long QT syndrome and BrS. We found that p.F532C reduced MOG1-Nav1.5 interaction and eliminated MOG1 function on INa; p.R535Q is also a loss-of-function mutation that reduces INa amplitude in a MOG1-independent manner, whereas p.F530V is benign as it does not have an apparent effect on MOG1 and INa. CONCLUSION: Our findings define the MOG1-Nav1.5 interaction domain to a 5-amino-acid motif of F530T531F532R533R534 in Loop I. Mutation p.F532C associated with BrS abolishes Nav1.5 interaction with MOG1 and reduces MOG1-enhanced INa density, thereby uncovering a novel molecular mechanism for the pathogenesis of BrS.


Asunto(s)
Síndrome de Brugada , Síndrome de QT Prolongado , Corazón , Humanos , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo
11.
Mol Med ; 27(1): 80, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34284715

RESUMEN

PURPOSE: Osteoarthritis (OA) is the most common inflammatory disease associated with pain and cartilage destruction. Interleukin (IL)-1ß is widely used to induce inflammatory response in OA models. This study aimed to explore the role of Danshensu (DSS) in IL-1ß-induced inflammatory responses in OA. METHODS: IL-1ß was used to induce chondrocyte inflammation. Cell viability was evaluated by Cell Counting Kit-8 (CCK-8) assay. IL-6, COX-2, TNF-α, and iNOS mRNA levels were detected by qRT-PCR. MMP3, MMP13, ADAMTS4, ADAMTS5, Aggrecan, Collagen, p-IκBα, and p-p65 protein levels were detected by Western blot. An OA mouse model was established by surgical destabilization of the medial meniscus (DMM), and the Osteoarthritis Research Society International (OARSI) score was evaluated by H&E staining. RESULTS: DSS did not affect the levels of inflammatory indicators including IL-6, COX-2, TNF-α, iNOS, PEG2, and NO but suppressed COX-2 and iNOS protein expression in IL-1ß treated chondrocytes. In addition, DSS downregulated IL-1ß-enhanced expression of MMP3, MMP13, ADAMTS4, and ADAMTS5 and upregulated aggrecan and collagen expression. Moreover, DSS significantly inhibited IL-1ß-induced phosphorylation of p-IκBα and p-p65 in a dose-dependent manner in chondrocytes, suggesting it plays a role in the NF-κB signaling pathway. Furthermore, DSS significantly reduced DMM-induced cartilage OARSI score in mice, further demonstrating its protective role in OA progression in vivo. CONCLUSIONS: Our study revealed the protective role of DSS in OA, suggesting that DSS might act as a potential treatment for OA.


Asunto(s)
Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Interleucina-1beta/metabolismo , Lactatos/farmacología , FN-kappa B/metabolismo , Osteoartritis/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Biomarcadores , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Matriz Extracelular/metabolismo , Mediadores de Inflamación/metabolismo , Lactatos/administración & dosificación , Lactatos/química , Ratones , Osteoartritis/tratamiento farmacológico , Osteoartritis/etiología , Osteoartritis/patología
13.
Acta Physiol (Oxf) ; 231(3): e13567, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33032360

RESUMEN

AIMS: MOG1 is a small protein that can bind to small GTPase RAN and regulate transport of RNA and proteins between the cytoplasm and nucleus. However, the in vivo physiological role of mog1 in the heart needs to be fully defined. METHODS: Mog1 knockout zebrafish was generated by TALEN. Echocardiography, histological analysis, and electrocardiograms were used to examine cardiac structure and function. RNA sequencing and real-time RT-PCR were used to elucidate the molecular mechanism and to analyse the gene expression. Isoproterenol was used to induce cardiac hypertrophy. Whole-mount in situ hybridization was used to observe cardiac morphogenesis. RESULTS: Mog1 knockout zebrafish developed cardiac hypertrophy and heart failure (enlarged pericardium, increased nppa and nppb expression and ventricular wall thickness, and reduced ejection fraction), which was aggravated by isoproterenol. RNAseq and KEGG pathway analyses revealed the effect of mog1 knockout on the pathways of cardiac hypertrophy, dilatation and contraction. Mechanistic studies revealed that mog1 knockout decreased expression of tbx5, which reduced expression of cryab and hspb2, resulting in cardiac hypertrophy and heart failure. Overexpression of cryab, hspb2 and tbx5 rescued the cardiac oedema phenotype of mog1 KO zebrafish. Telemetry electrocardiogram monitoring showed QRS and QTc prolongation and a reduced heart rate in mog1 knockout zebrafish, which was associated with reduced scn1b expression. Moreover, mog1 knockout resulted in abnormal cardiac looping during embryogenesis because of the reduced expression of nkx2.5, gata4 and hand2. CONCLUSION: Our data identified an important molecular determinant for cardiac hypertrophy and heart failure, and rhythm maintenance of the heart.


Asunto(s)
Insuficiencia Cardíaca , Pez Cebra , Animales , Cardiomegalia/genética , Corazón , Insuficiencia Cardíaca/genética , Transducción de Señal
14.
Cancer Lett ; 497: 28-40, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33069768

RESUMEN

Angiogenesis factors are widely known to promote tumor growth by increasing tumor angiogenesis in the tumor microenvironment, however, little is known whether their intracellular function is involved in tumorigenesis. Here we show that AGGF1 acts as a tumor suppressor by regulating p53 when acting inside tumor cells. AGGF1 antagonizes MDM2 function to inhibit p53 ubiquitination, increases the acetylation, phosphorylation, stability and expression levels of p53, activates transcription of p53 target genes, and regulates cell proliferation, cell cycle, and apoptosis. AGGF1 also interacts with p53 through the FHA domain. Somatic AGGF1 variants in the FHA domain in human tumors, including p.Q467H, p.Y469 N, and p.N483T, inhibit AGGF1 activity on tumor suppression. These results identify a key role for AGGF1 in an AGGF1-MDM2-p53 signaling axis with important functions in tumor suppression, and uncover a novel trans-tumor-suppression mechanism dependent on p53. This study has potential implications in diagnosis and therapies of cancer.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Procesamiento Postranscripcional del ARN , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Angiogénicas/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Pronóstico , Proteínas Proto-Oncogénicas c-mdm2/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Aging (Albany NY) ; 12(12): 12074-12085, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32580167

RESUMEN

Osteoarthritis (OA) is a whole-joint disease with extremely high prevalence. In all treatment approaches of OA, blocking the degradation of the cartilage extracellular matrix is an important treatment. In OA, overexpression of derivative enzymes leads to excessive catabolism and reduced synthesis of cartilage including type II collagen and aggrecan, which results in irreversible destruction of the joint. SOX9 is a transcription factor that regulates the synthesis of type II collagen and aggrecan and is significantly downregulated in OA. GPR120 has been reported to affect the pathophysiology of OA. In this study, we used the GPR120 agonist GW9508 and TUG891 in ATDC5 chondrocytes exposed to interleukin (IL)-1ß to investigate the involvement of GPR120 in SOX9-mediated expression of type II collagen and aggrecan. Our findings show that agonism of GPR120 can reduce inflammation by inhibiting the expression of IL-6 and IL-8 induced by IL-1ß. We also show that GW9508 and TUG891 rescue the expression of type II collagen and aggrecan by preventing the reduction of SOX9 expression. Additionally, we demonstrate that the effects of GW9508 on SOX9 expression are mediated through CREB and that GPR120 is indeed required for this effect. Thus, agonism of GPR120 by GW9508 might be a potential therapeutic strategy to halt or prevent cartilage degradation.


Asunto(s)
Interleucina-1beta/inmunología , Metilaminas/uso terapéutico , Osteoartritis/prevención & control , Propionatos/uso terapéutico , Receptores Acoplados a Proteínas G/agonistas , Factor de Transcripción SOX9/metabolismo , Agrecanos/metabolismo , Animales , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/uso terapéutico , Cartílago Articular/citología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/inmunología , Cartílago Articular/patología , Línea Celular , Condrocitos/efectos de los fármacos , Condrocitos/patología , Colágeno Tipo II/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Matriz Extracelular/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/metabolismo , Metilaminas/farmacología , Ratones , Osteoartritis/inmunología , Osteoartritis/patología , Fenilpropionatos/farmacología , Fenilpropionatos/uso terapéutico , Propionatos/farmacología , Proteolisis/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
16.
Gene ; 753: 144805, 2020 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-32445923

RESUMEN

Genomic variants in both ADTRP and TFPI genes are associated with risk of coronary artery disease (CAD). ADTRP regulates TFPI expression and endothelial cell functions involved in the initiation of atherosclerotic CAD. ADTRP also specifies primitive myelopoiesis and definitive hematopoiesis by upregulating TFPI expression. However, the underlying molecular mechanism is unknown. Here we show that transcription factor POU1F1 is the key by which ADTRP regulates TFPI expression. Luciferase reporter assays, chromatin-immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA) in combination with analysis of large and small deletions of the TFPI promoter/regulatory region were used to identify the molecular mechanism by which ADTRP regulates TFPI expression. Genetic association was assessed using case-control association analysis and phenome-wide association analysis (PhenGWA). ADTRP regulates TFPI expression at the transcription level in a dose-dependent manner. The ADTRP-response element was localized to a 50 bp region between -806 bp and -756 bp upstream of TFPI transcription start site, which contains a binding site for POU1F1. Deletion of POU1F1-binding site or knockdown of POU1F1 expression abolished ADTRP-mediated transcription of TFPI. ChIP and EMSA demonstrated that POU1F1 binds to the ADTRP response element. Genetic analysis identified significant association between POU1F1 variants and risk of CAD. PhenGWA identified other phenotypic traits associated with the ADTRP-POU1F1-TFPI axis such as lymphocyte count (ADTRP), waist circumference (TFPI), and standing height (POU1F1). These data identify POU1F1 as a transcription factor that regulates TFPI transcription in response to ADTRP, and link POU1F1 variants to risk of CAD for the first time.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Lipoproteínas/biosíntesis , Proteínas de la Membrana/metabolismo , Factor de Transcripción Pit-1/metabolismo , Aterosclerosis/genética , Estudios de Casos y Controles , Línea Celular , Inmunoprecipitación de Cromatina/métodos , Enfermedad de la Arteria Coronaria/genética , Bases de Datos Genéticas , Células Endoteliales/metabolismo , Genes Homeobox , Células HeLa , Humanos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Regiones Promotoras Genéticas , Elementos de Respuesta , Sitio de Iniciación de la Transcripción , Transcripción Genética
17.
Future Oncol ; 16(11): 665-674, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32250163

RESUMEN

Aim: This experimental design was based on DHRS12 to explore its biological effects on osteosarcoma (OS). Materials & methods: The expression level of endogenous DHRS12 was analyzed by immunohistochemical analysis. DHRS12 was overexpressed in MG-63 and HOS cells by plasmid transfection. Cell proliferation, invasion, migration, apoptosis and western blot were used in the experiment. Results: The expression of DHRS12 was significantly reduced in OS. Overexpression of DHRS12 inhibited the proliferation, migration and invasion of MG-63 and HOS cells and induced apoptosis of OS cells. Overexpression of DHRS12 upregulated Bax, Caspase 9 and Caspase 3. Overexpression of DHRS12 resulted in inactivation of the Wnt3a/ß-catenin signaling pathway. Conclusion: Overexpression of DHRS12 inhibited the progression of OS via the Wnt3a/ß-catenin pathway.


Asunto(s)
Osteosarcoma/patología , Deshidrogenasas-Reductasas de Cadena Corta/metabolismo , Vía de Señalización Wnt , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/mortalidad , Deshidrogenasas-Reductasas de Cadena Corta/genética , Tasa de Supervivencia , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
18.
J Shoulder Elbow Surg ; 28(7): 1308-1315, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31230782

RESUMEN

BACKGROUND: Iatrogenic supraclavicular nerve injury is frequent during surgical repair of clavicle fractures through a transverse incision. The use of an oblique incision may be a potential approach to avoiding this complication. This study compared the clinical effectiveness of oblique and transverse incisions in the treatment of fractures in the middle and outer thirds of the clavicle. METHODS: This prospective observational study included patients with fracture of the mid-to-outer third of the clavicle between August 2011 and August 2016. We allocated the patients into 2 groups based on their choice of treatment: oblique incision (n = 62) and transverse incision (n = 64). We compared the following parameters between the 2 groups: operative time, intraoperative blood loss, postoperative fracture healing time, incision size, clinical complications, postoperative subjective satisfaction, and shoulder function. RESULTS: Operative time, postoperative fracture healing time, postoperative shoulder function (Constant-Murley and disabilities of the arm, shoulder and hand [DASH] scores), and clinical complications did not differ significantly between groups (all P > .05). The oblique incision group had less intraoperative blood loss (41.4 ± 16.4 vs. 65.3 ± 10.4 mL, P < .001) and smaller surgical incisions (3.6 ± 1.6 vs. 10.3 ± 2.6 cm, P < .001). The oblique incision group showed better outcomes for postoperative satisfaction (85.5% vs. 64.1%, P = .015), absence of shoulder numbness at the last follow-up (89.3% vs. 70.3%, P = .010), and satisfaction with the scar (90.3% vs. 3.1%, P < .001). CONCLUSION: Oblique incisions have several advantages over transverse incisions: less bleeding, smaller incisions, less iatrogenic injury to supraclavicular nerves, and higher patient satisfaction. These 2 approaches have equivalent effects on recovery of shoulder joint function.


Asunto(s)
Clavícula/cirugía , Fijación Interna de Fracturas/efectos adversos , Fijación Interna de Fracturas/métodos , Fracturas Óseas/cirugía , Traumatismos de los Nervios Periféricos/prevención & control , Adulto , Pérdida de Sangre Quirúrgica , Clavícula/lesiones , Femenino , Curación de Fractura , Humanos , Masculino , Persona de Mediana Edad , Tempo Operativo , Satisfacción del Paciente , Traumatismos de los Nervios Periféricos/etiología , Complicaciones Posoperatorias/etiología , Periodo Posoperatorio , Estudios Prospectivos , Hombro/fisiopatología , Factores de Tiempo , Resultado del Tratamiento
19.
Ann Hum Genet ; 83(4): 239-248, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30821358

RESUMEN

Atrial fibrillation (AF) affects 33.5 million individuals worldwide. It accounts for 15% of strokes and increases risk of heart failure and sudden death. The voltage-gated cardiac sodium channel complex is responsible for the generation and conduction of the cardiac action potential, and composed of the main pore-forming α-subunit Nav 1.5 (encoded by the SCN5A gene) and one or more auxiliary ß-subunits, including Nav ß1 to Nav ß4 encoded by SCN1B to SCN4B, respectively. We and others identified loss-of-function mutations in SCN1B and SCN2B and dominant-negative mutations in SCN3B in patients with AF. Three missense variants in SCN4B were identified in sporadic AF patients and small nuclear families; however, the association between SCN4B variants and AF remains to be further defined. In this study, we performed mutational analysis in SCN4B using a panel of 477 AF patients, and identified one nonsynonymous genomic variant p.Gly8Ser in four patients. To assess the association between the p.Gly8Ser variant and AF, we carried out case-control association studies with two independent populations (944 AF patients vs. 9,81 non-AF controls in the first discovery population and 732 cases and 1,291 controls in the second replication population). Significant association was identified in the two independent populations and in the combined population (p = 4.16 × 10-4 , odds ratio [OR] = 3.14) between p.Gly8Ser and common AF as well as lone AF (p = 0.018, OR = 2.85). These data suggest that rare variant p.Gly8Ser of SCN4B confers a significant risk of AF, and SCN4B is a candidate susceptibility gene for AF.


Asunto(s)
Alelos , Sustitución de Aminoácidos , Fibrilación Atrial/genética , Variación Genética , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/genética , Anciano , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Estudios de Casos y Controles , Biología Computacional/métodos , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mutación , Polimorfismo de Nucleótido Simple , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/metabolismo
20.
Hum Mutat ; 40(3): 310-325, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30488537

RESUMEN

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.


Asunto(s)
Fibrilación Atrial/genética , Lamina Tipo A/genética , Mutación/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Humanos , Activación del Canal Iónico , Carioferinas/metabolismo , Lamina Tipo A/química , Ratones , Membrana Nuclear/metabolismo , Unión Proteica , Transporte de Proteínas , ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales de Sodio/metabolismo , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA