Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 15(4): 253, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38594244

RESUMEN

Mitochondria are important for the activation of endothelial cells and the process of angiogenesis. NDUFS8 (NADH:ubiquinone oxidoreductase core subunit S8) is a protein that plays a critical role in the function of mitochondrial Complex I. We aimed to investigate the potential involvement of NDUFS8 in angiogenesis. In human umbilical vein endothelial cells (HUVECs) and other endothelial cell types, we employed viral shRNA to silence NDUFS8 or employed the CRISPR/Cas9 method to knockout (KO) it, resulting in impaired mitochondrial functions in the endothelial cells, causing reduction in mitochondrial oxygen consumption and Complex I activity, decreased ATP production, mitochondrial depolarization, increased oxidative stress and reactive oxygen species (ROS) production, and enhanced lipid oxidation. Significantly, NDUFS8 silencing or KO hindered cell proliferation, migration, and capillary tube formation in cultured endothelial cells. In addition, there was a moderate increase in apoptosis within NDUFS8-depleted endothelial cells. Conversely, ectopic overexpression of NDUFS8 demonstrated a pro-angiogenic impact, enhancing cell proliferation, migration, and capillary tube formation in HUVECs and other endothelial cells. NDUFS8 is pivotal for Akt-mTOR cascade activation in endothelial cells. Depleting NDUFS8 inhibited Akt-mTOR activation, reversible with exogenous ATP in HUVECs. Conversely, NDUFS8 overexpression boosted Akt-mTOR activation. Furthermore, the inhibitory effects of NDUFS8 knockdown on cell proliferation, migration, and capillary tube formation were rescued by Akt re-activation via a constitutively-active Akt1. In vivo experiments using an endothelial-specific NDUFS8 shRNA adeno-associated virus (AAV), administered via intravitreous injection, revealed that endothelial knockdown of NDUFS8 inhibited retinal angiogenesis. ATP reduction, oxidative stress, and enhanced lipid oxidation were detected in mouse retinal tissues with endothelial knockdown of NDUFS8. Lastly, we observed an increase in NDUFS8 expression in retinal proliferative membrane tissues obtained from human patients with proliferative diabetic retinopathy. Our findings underscore the essential role of the mitochondrial protein NDUFS8 in regulating endothelial cell activation and angiogenesis.


Asunto(s)
Angiogénesis , Proteínas Proto-Oncogénicas c-akt , Humanos , Ratones , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Movimiento Celular , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , ARN Interferente Pequeño/farmacología , Lípidos/farmacología , Adenosina Trifosfato/farmacología , Proliferación Celular/genética , NADH Deshidrogenasa/genética , NADH Deshidrogenasa/metabolismo
2.
Nat Commun ; 14(1): 8393, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110369

RESUMEN

Ferroptosis is an iron-dependent programmed cell death associated with severe kidney diseases, linked to decreased glutathione peroxidase 4 (GPX4). However, the spatial distribution of renal GPX4-mediated ferroptosis and the molecular events causing GPX4 reduction during ischemia-reperfusion (I/R) remain largely unknown. Using spatial transcriptomics, we identify that GPX4 is situated at the interface of the inner cortex and outer medulla, a hyperactive ferroptosis site post-I/R injury. We further discover OTU deubiquitinase 5 (OTUD5) as a GPX4-binding protein that confers ferroptosis resistance by stabilizing GPX4. During I/R, ferroptosis is induced by mTORC1-mediated autophagy, causing OTUD5 degradation and subsequent GPX4 decay. Functionally, OTUD5 deletion intensifies renal tubular cell ferroptosis and exacerbates acute kidney injury, while AAV-mediated OTUD5 delivery mitigates ferroptosis and promotes renal function recovery from I/R injury. Overall, this study highlights a new autophagy-dependent ferroptosis module: hypoxia/ischemia-induced OTUD5 autophagy triggers GPX4 degradation, offering a potential therapeutic avenue for I/R-related kidney diseases.


Asunto(s)
Lesión Renal Aguda , Ferroptosis , Daño por Reperfusión , Humanos , Riñón , Autofagia , Isquemia
3.
BMC Pediatr ; 23(1): 358, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37442965

RESUMEN

BACKGROUND: We aimed to analyze the characteristics of the body composition of children and adolescents aged 3-17 in Suzhou, China. METHODS: A cross-sectional study between January 2020 and June 2022 using bioelectrical impedance was conducted to determine the fat mass (FM), fat-free mass (FFM), skeletal muscle mass, and protein and mineral contents of 24,845 children aged 3-17 who attended the Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, China. Measurement data was presented in tables as mean ± SD, and groups were compared using the independent samples t-test. RESULTS: FM and fat-free mass increased with age in both boys and girls. The fat-free mass of girls aged 14-15 decreased after reaching a peak, and that of boys in the same age group was higher than that of the girls (p < 0.05). There were no significant differences in FM between boys and girls younger than 9- and 10-years old. The percentage body fat (PBF) and FM index of girls increased rapidly between 11 and 15 years of age (p < 0.05), and those of boys aged 11-14 were significantly lower (p < 0.05), suggesting that the increase in body mass index (BMI) was mainly contributed by muscle mass (MM) in boys. CONCLUSIONS: The body composition of children and adolescents varies according to their age and sex. A misdiagnosis of obesity made on the basis of BMI alone can be avoided if BMI is used in combination with FM index, percentage body fat, and other indexes.


Asunto(s)
Composición Corporal , Obesidad , Masculino , Femenino , Humanos , Niño , Adolescente , Estudios Transversales , Índice de Masa Corporal , China , Tejido Adiposo
4.
Int Immunopharmacol ; 122: 110617, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37478666

RESUMEN

This study aims to discern the possible molecular mechanism of the effect of ubiquitin-specific peptidase 18 (USP18) on the resistance to BRAF inhibitor vemurafenib in BRAF V600E mutant melanoma by regulating cyclic GMP-AMP synthase (cGAS). The cancer tissues of BRAF V600E mutant melanoma patients before and after vemurafenib treatment were collected, in which the protein expression of USP18 and cGAS was determined. A BRAF V600E mutant human melanoma cell line (A2058R) resistant to vemurafenib was constructed with its viability, apoptosis, and autophagy detected following overexpression and depletion assays of USP18 and cGAS. Xenografted tumors were transplanted into nude mice for in vivo validation. Bioinformatics analysis showed that the expression of cGAS was positively correlated with USP18 in melanoma, and USP18 was highly expressed in melanoma. The expression of cGAS and USP18 was up-regulated in cancer tissues of vemurafenib-resistant patients with BRAF V600E mutant melanoma. Knockdown of cGAS inhibited the resistance to vemurafenib in A2058R cells and the protective autophagy induced by vemurafenib in vitro. USP18 could deubiquitinate cGAS to promote its protein stability. In vivo experimentations confirmed that USP18 promoted vemurafenib-induced protective autophagy by stabilizing cGAS protein, which promoted resistance to vemurafenib in BRAF V600E mutant melanoma cells. Collectively, USP18 stabilizes cGAS protein expression through deubiquitination and induces autophagy of melanoma cells, thereby promoting the resistance to vemurafenib in BRAF V600E mutant melanoma.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Animales , Ratones , Humanos , Vemurafenib/farmacología , Vemurafenib/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Ratones Desnudos , Indoles/farmacología , Indoles/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Resistencia a Antineoplásicos/genética , Mutación , Línea Celular Tumoral , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Autofagia/genética , Nucleotidiltransferasas/genética , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/farmacología
5.
Redox Rep ; 28(1): 2152607, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36692085

RESUMEN

Renal proximal tubular cells are highly vulnerable to different types of assaults during filtration and reabsorption, leading to acute renal dysfunction and eventual chronic kidney diseases (CKD). The chemotherapeutic drug cisplatin elicits cytotoxicity causing renal tubular cell death, but its executing mechanisms of action are versatile and elusive. Here, we show that cisplatin induces renal tubular cell apoptosis and ferroptosis by disrupting glutathione (GSH) metabolism. Upon cisplatin treatment, GSH metabolism is impaired leading to GSH depletion as well as the execution of mitochondria-mediated apoptosis and lipid oxidation-related ferroptosis through activating IL6/JAK/STAT3 signaling. Inhibition of JAK/STAT3 signaling reversed cell apoptosis and ferroptosis in response to cisplatin induction. Using a cisplatin-induced acute kidney injury (CAKI) mouse model, we found that inhibition of JAK/STAT3 significantly mitigates cisplatin nephrotoxicity with a reduced level of serum BUN and creatinine as well as proximal tubular distortion. In addition, the GSH booster baicalein also reclaims cisplatin-induced renal tubular cell apoptosis and ferroptosis as well as the in vivo nephrotoxicity. In conclusion, cisplatin disrupts glutathione metabolism, leading to renal tubular cell apoptosis and ferroptosis. Rewiring glutathione metabolism represents a promising strategy for combating cisplatin nephrotoxicity.


Asunto(s)
Lesión Renal Aguda , Ferroptosis , Ratones , Animales , Cisplatino , Apoptosis , Riñón/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Glutatión/metabolismo
6.
Ann Clin Lab Sci ; 52(6): 956-966, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36564069

RESUMEN

OBJECTIVE: The long non-coding RNA (lncRNA) colorectal neoplasia differentially expressed (CRNDE) is considered a carcinogenic promoter in various human malignancies. However, the role and underlying mechanism of action of CRNDE during carcinogenesis in neuroblastoma remain unknown. METHODS: CRNDE transcript levels were detected in neuroblastoma tissues and adjacent normal tissues. The effects of CRNDE overexpression and knockdown on the viability of SH-SY5Y and SK-N-AS cells were determined using the Cell Counting Kit-8 (CCK-8) assay. Flow cytometry was performed to measure the role of CRNDE in apoptosis and the cell cycle in neuroblastoma cells. Moreover, the transwell assay was used to evaluate the role of CRNDE in the migration and invasion of tumor cells. The levels of ERK/MAPK pathway-related proteins were evaluated using western blotting. The in vivo role of CRNDE in tumor growth and apoptosis was evaluated in a xenograft mouse model of human neuroblastoma. RESULTS: The relative expression of CRNDE was significantly higher in neuroblastoma tissues than in the adjacent normal tissues. Moreover, knockdown of CRNDE inhibited tumor cell proliferation and induced apoptosis and cell cycle arrest, whereas elevation of CRNDE promoted cell growth and inhibited apoptosis in neuroblastoma cells. In addition, depletion of CRNDE suppressed migration and invasion, whereas overexpression of CRNDE enhanced the migratory and invasive potential of SH-SY5Y and SK-N-AS cells. At the mechanistic level, western blotting showed that CRNDE exerted its oncogenic role by affecting the ERK/MAPK signaling pathway. Furthermore, animal experiments confirmed that CRNDE promotes tumor growth and inhibits apoptosis in neuroblastoma in vivo. CONCLUSION: The present study revealed that CRNDE plays a critical role in the proliferation, apoptosis, migration, and invasion of neuroblastoma by altering the ERK/MAPK signaling pathway, representing a novel molecular target for the treatment of neuroblastoma.


Asunto(s)
MicroARNs , Neuroblastoma , ARN Largo no Codificante , Humanos , Animales , Ratones , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Carcinógenos , Línea Celular Tumoral , Neuroblastoma/genética , Transducción de Señal/genética , Carcinogénesis/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética
7.
Gene ; 741: 144537, 2020 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-32156528

RESUMEN

Breed improvement is an important genetic process affecting meat quality. Compared with Kazakh cattle, Xinjiang brown cattle have significantly improved carcass quality and meat quality. To elucidate the molecular mechanisms underlying the improvements in Xinjiang brown cattle and the differences in beef quality between the two breeds, we used RNA-Seq to study differentially expressed genes and miRNAs and regulatory pathways related to adipogenesis, myogenesis and fibrogenesis in the longissimus dorsi muscles of Xinjiang brown cattle and Kazakh cattle. The results showed that 1669 genes were differentially expressed in the longissimus dorsi muscle tissues of Xinjiang brown cattle and Kazakh cattle; 879 genes were upregulated and 790 genes were downregulated in Xinjiang brown cattle compared to Kazakh cattle. These genes were mainly involved in PPAR signaling, unsaturated fatty acid biosynthesis and vascular smooth muscle contraction. Additionally, 346 differentially expressed miRNAs were identified, of which 265 miRNAs were downregulated and 81 miRNAs were upregulated in Xinjiang brown cattle compared to Kazakh cattle. Association analysis of the differentially expressed genes and miRNAs revealed that 86 differentially expressed miRNAs related to adipogenesis were associated with 31 differentially expressed genes, 76 differentially expressed miRNAs associated with myogenesis were associated with 28 differentially expressed genes, and 54 differentially expressed miRNAs associated with fibrogenesis were associated with 19 differentially expressed genes. miRNA-target gene networks were also constructed. Finally, the expression levels of 19 genes and miRNAs were verified by qRT-PCR. Some differentially expressed genes, including FABP4, ACTA2 and ACTG2, were shown to play an important role in beef meat quality. This is the first study to perform transcriptomic analysis of muscle tissues from Xinjiang brown and Kazakh cattle.


Asunto(s)
Bovinos/genética , MicroARNs/genética , Músculo Esquelético/crecimiento & desarrollo , ARN Mensajero/genética , Animales , Cruzamiento , Bovinos/crecimiento & desarrollo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Carne , Desarrollo de Músculos/genética , Análisis de Secuencia de ARN , Transcriptoma/genética
8.
Oncol Lett ; 14(5): 5773-5778, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29113206

RESUMEN

The abnormal expression of nuclear paraspeckle assembly transcript 1 (NEAT1) may serve critical functions for the development and progression of various types of human tumor. However, the expression and biological function of NEAT1 in hepatoblastoma (HB) and the underlying mechanisms for the function of NEAT1 in HB remain largely uncharacterized. In the present study, the results of reverse transcription-quantitative polymerase chain reaction revealed that the expression of NEAT1 was significantly elevated in HB tissues. HB tissues with metastasis also exhibited significantly increased levels of NEAT1 compared with tissues without metastasis. The biological functions of NEAT1 were then assessed using gain-/loss-of-function studies. The results of in vitro assays revealed that inhibiting NEAT1 expression reduced the migration and invasion of HepG2 cells. By contrast, the induced expression of NEAT1 exhibited the opposite effect. The present study also demonstrated that the inhibition of NEAT1 expression prevented the epithelial-mesenchymal transition of HepG2 cells, whereas forced expression of NEAT1 exhibited the opposite effect. In addition, it was confirmed that NEAT1 could modulate the expression of microRNA (miR)-129-5p in HepG2 cells, and that NEAT1 may exert its effect on the metastatic behaviors and epithelial-mesenchymal transition of HepG2 cells by inhibiting miR-129-5p. In conclusion, the present study indicated that NEAT1 expression was aberrantly increased in HB and that it may promote the metastasis of HB cells by inhibiting miR-129-5p. Targeting NEAT1 may potentially be a novel therapeutic option for treating patients with HB.

9.
Cancer Biomark ; 20(4): 589-596, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-28800318

RESUMEN

Neuroblastoma is a malignancy [corrected] of childhood and accounts for 7-10% of childhood cancers, leading to approximately 15% of pediatric cancer deaths. MicroRNAs (miRNAs) are a family of short (about 18-25 nucleotides), noncoding and single stranded endogenous RNAs, which complementarily bind to the 3' untranslated regions of their target genes. Recently, glutamine metabolism has been recognized as an important nutrition source for tumor cells, and hence targeting glutamine metabolism could benefit to development of anti-cancer agents. In this study, we investigate the roles of miR-513c in human neuroblastoma. We report miR-513c is significantly downregulated in human neuroblastoma tissues compared with their adjacent normal tissues. Moreover, miR-513c is significantly downregulated in neuroblastoma cell lines compared with normal neuroblast cells. Overexpression of miR-513c suppresses neuroblastoma cells' migration, invasion, and proliferation. We demonstrate the glutaminase (GLS) is a direct target of miR-513c in human neuroblastoma cells. In addition, we found restoration of GLS expression recovered the neuroblastoma cells' migration, invasion, and proliferation. In summary, this study illustrates a miR-513c mediated neuroblastoma cells suppression, providing a new aspect on the miRNA-based therapeutic approach for the treatments of neuroblastoma.


Asunto(s)
Expresión Génica , Glutaminasa/genética , MicroARNs/genética , Neuroblastoma/genética , Interferencia de ARN , Regiones no Traducidas 3' , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos
10.
Biochem Biophys Res Commun ; 479(2): 159-165, 2016 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-27524236

RESUMEN

Prostate Cancer has become the second leading cause of male cancer-related incidence and mortality in United States. Hyperthermia (HT) is known to serve as a powerful tool in treatment of prostate cancer in clinical. The combination treatment with HT and cisplatin has a synergistic effect to inhibit prostate cancer progression and demonstrates better clinical effectiveness than HT or chemotherapy alone. But molecular mechanisms of this phenomenon have not been illuminated clearly. In this study, we used MTS assay to examine cell viabilities of PC-3, LNCaP, DU-145 and RM-1 cells after treated by HT and cisplatin. Then colony formation of PC-3 and DU-145 cells after treated with HT and cisplatin were photographed. To investigate whether the combination therapy would enhance apoptosis of PC-3 and DU-145 cells, we used Western blot analysis to detect expression level of proteins on apoptosis-regulated signaling pathway in PC-3 and DU-145 cells. Our results showed that the combination treatment decreased cell viabilities and colony formation of prostate cancer cells in a dose-dependent manner and this combination therapy enhanced apoptosis of PC-3 and DU-145 cells via activation of Caspase-3 and cleavage of PARP. We also found that the combination therapy could down-regulate the anti-apoptotic Bcl-2 and IAP family proteins. At last, the combination therapy activated AMPKα-JNK signaling pathway and inhibited Akt-mTOR-p70s6k signaling pathway to promote apoptosis of PC-3 and DU-145 cells. In conclusion, this study clearly elucidated how the combination therapy with HT and cisplatin promoted apoptosis of prostate cancer cells in synergy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Calor , Animales , Antineoplásicos/farmacología , Western Blotting , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
11.
Mol Med Rep ; 13(3): 2745-50, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26847592

RESUMEN

The histone deacetylase (HDAC) family is comprised of enzymes, which are involved in modulating the majority of critical cellular processes, including transcriptional regulation, apoptosis, proliferation and cell cycle progression. However, the biological function of HDAC5 in Wilms' tumor remains to be fully elucidated. The present study aimed to investigate the expression and function of HDAC5 in Wilm's tumor. It was demonstrated that the mRNA and protein levels of HDAC5 were upregulated in human Wilms' tumor tissues. Overexpression of HDAC5 in G401 cells was observed to significantly promote cellular proliferation, as demonstrated by the results of an MTT assay and bromodeoxyuridine incorporation assay. By contrast, HDAC5 knockdown using small interfering RNA suppressed the proliferation of the G401 cells. At the molecular level, the present study demonstrated that HDAC5 promoted the expression of c­Met, which has been previously identified as an oncogene. In addition, downregulation of c­Met inhibited the proliferative effects of HDAC5 in human Wilms' tumor cells. Taken together, these results suggested that HDAC5 promotes cellular proliferation through the upregulation of c­Met, and may provide a novel therapeutic target for the treatment of patients with Wilms' tumor.


Asunto(s)
Proliferación Celular , Histona Desacetilasas/metabolismo , Neoplasias Renales/enzimología , Proteínas Tirosina Quinasas Receptoras/genética , Tumor de Wilms/enzimología , Línea Celular Tumoral , Preescolar , Femenino , Expresión Génica , Histona Desacetilasas/genética , Humanos , Lactante , Neoplasias Renales/patología , Masculino , Proteínas Tirosina Quinasas Receptoras/metabolismo , Regulación hacia Arriba , Tumor de Wilms/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...