Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Endocrinol ; 262(1)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38692289

RESUMEN

CD44, a cell surface adhesion receptor and stem cell biomarker, is recently implicated in chronic metabolic diseases. Ablation of CD44 ameliorates adipose tissue inflammation and insulin resistance in obesity. Here, we investigated cell type-specific CD44 expression in human and mouse adipose tissue and further studied how CD44 in preadipocytes regulates adipocyte function. Using Crispr Cas9-mdediated gene deletion and lentivirus-mediated gene re-expression, we discovered that deletion of CD44 promotes adipocyte differentiation and adipogenesis, whereas re-expression of CD44 abolishes this effect and decreases insulin responsiveness and adiponectin secretion in 3T3-L1 cells. Mechanistically, CD44 does so via suppressing Pparg expression. Using quantitative proteomics analysis, we further discovered that cell cycle-regulated pathways were mostly decreased by deletion of CD44. Indeed, re-expression of CD44 moderately restored expression of proteins involved in all phases of the cell cycle. These data were further supported by increased preadipocyte proliferation rates in CD44-deficient cells and re-expression of CD44 diminished this effect. Our data suggest that CD44 plays a crucial role in regulating adipogenesis and adipocyte function possibly through regulating PPARγ and cell cycle-related pathways. This study provides evidence for the first time that CD44 expressed in preadipocytes plays key roles in regulating adipocyte function outside immune cells where CD44 is primarily expressed. Therefore, targeting CD44 in (pre)adipocytes may provide therapeutic potential to treat obesity-associated metabolic complications.


Asunto(s)
Células 3T3-L1 , Adipocitos , Adipogénesis , Ciclo Celular , Receptores de Hialuranos , PPAR gamma , Adipogénesis/genética , Adipogénesis/fisiología , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/genética , Animales , PPAR gamma/metabolismo , PPAR gamma/genética , Ratones , Ciclo Celular/genética , Ciclo Celular/fisiología , Humanos , Adipocitos/metabolismo , Eliminación de Gen , Diferenciación Celular/genética , Masculino , Tejido Adiposo/metabolismo , Tejido Adiposo/citología , Transducción de Señal/fisiología
2.
Dis Model Mech ; 17(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38756069

RESUMEN

Alström syndrome (AS), a multisystem disorder caused by biallelic ALMS1 mutations, features major early morbidity and mortality due to cardiac complications. The latter are biphasic, including infantile dilated cardiomyopathy and distinct adult-onset cardiomyopathy, and poorly understood. We assessed cardiac function of Alms1 knockout (KO) mice by echocardiography. Cardiac function was unaltered in Alms1 global KO mice of both sexes at postnatal day 15 (P15) and 8 weeks. At 23 weeks, female - but not male - KO mice showed increased left atrial area and decreased isovolumic relaxation time, consistent with early restrictive cardiomyopathy, as well as reduced ejection fraction. No histological or transcriptional changes were seen in myocardium of 23-week-old female Alms1 global KO mice. Female mice with Pdgfra-Cre-driven Alms1 deletion in cardiac fibroblasts and in a small proportion of cardiomyocytes did not recapitulate the phenotype of global KO at 23 weeks. In conclusion, only female Alms1-deficient adult mice show echocardiographic evidence of cardiac dysfunction, consistent with the cardiomyopathy of AS. The explanation for sexual dimorphism remains unclear but might involve metabolic or endocrine differences between sexes.


Asunto(s)
Síndrome de Alstrom , Cardiomiopatías , Ecocardiografía , Ratones Noqueados , Animales , Femenino , Masculino , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/patología , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Síndrome de Alstrom/complicaciones , Síndrome de Alstrom/genética , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Ratones , Miocardio/patología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fenotipo , Caracteres Sexuales
3.
Mol Metab ; 84: 101933, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38583571

RESUMEN

OBJECTIVE: Alström Syndrome (AS), caused by biallelic ALMS1 mutations, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and fatty liver. Prior studies suggest that hyperphagia is accounted for by loss of ALMS1 function in hypothalamic neurones, whereas disproportionate metabolic complications may be due to impaired adipose tissue expandability. We tested this by comparing the metabolic effects of global and mesenchymal stem cell (MSC)-specific Alms1 knockout. METHODS: Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα-Cre driver was used to abrogate Alms1 function selectively in MSCs and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα+ Alms1-KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. RESULTS: Assessed on 45% fat diet to promote adipose expansion, global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα-cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfrα expression. CONCLUSIONS: Mesenchymal deletion of Alms1 recapitulates metabolic features of AS, including fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. Hyperphagia in females may depend on Alms1 deficiency in oligodendrocyte precursor cells rather than neurones. AS should be regarded as a forme fruste of lipodystrophy.


Asunto(s)
Síndrome de Alstrom , Células Madre Mesenquimatosas , Ratones Noqueados , Animales , Ratones , Masculino , Femenino , Células Madre Mesenquimatosas/metabolismo , Síndrome de Alstrom/metabolismo , Síndrome de Alstrom/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Resistencia a la Insulina , Hígado Graso/metabolismo , Hígado Graso/genética , Obesidad/metabolismo , Obesidad/genética , Hiperfagia/metabolismo , Hiperfagia/genética , Tejido Adiposo/metabolismo , Ratones Endogámicos C57BL , Composición Corporal
4.
Front Mol Biosci ; 11: 1395607, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38545415
6.
bioRxiv ; 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37873427

RESUMEN

Background: Alström Syndrome (AS), a multi-system disease caused by mutations in the ALMS1 gene, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and hepatosteatosis. How loss of ALMS1 causes this phenotype is poorly understood, but prior studies have circumstancially implicated impaired adipose tissue expandability. We set out to test this by comparing the metabolic effects of selective Alms1 knockout in mesenchymal cells including preadipocytes to those of global Alms1 knockout. Methods: Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα -Cre driver was used to abrogate Alms1 function selectively in mesenchymal stem cells (MSCs) and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα + Alms1 -KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. Results: Global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα - cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfr α expression. Conclusions: Mesenchymal deletion of Alms1 recapitulates the metabolic features of AS, including severe fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. AS should be regarded as a forme fruste of lipodystrophy. Therapies should prioritise targeting positive energy balance.

7.
Commun Med (Lond) ; 3(1): 29, 2023 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-36806755

RESUMEN

BACKGROUND: Considerable variability exists in response to metformin with few effective biomarkers to guide the treatment. Here we evaluated whether whole blood derived mitochondrial DNA copy number (mtDNA-CN) is a biomarker of metformin response as measured by glucose reduction or weight loss. METHODS: Using data from the trial of Metformin (n = 304) and AcaRbose (n = 300) in Chinese as the initial Hypoglycaemic treatment (MARCH), we examined the association between mtDNA-CN and two metformin response outcomes of HbA1c reduction and weight loss. The acarbose arm was used as a comparator group. Whole blood mtDNA-CN was estimated by deep whole genome sequencing with adjustments for confounders. Multiple linear regression and repeated measurement analyses were used to evaluate the association between mtDNA-CN and drug response outcomes. RESULTS: Here we show that glucose reduction is not significantly associated with mtDNA-CN and in either treatment arm. In the metformin arm, each increase of 1 SD in mtDNA-CN is significantly (P = 0.006) associated with a 0.43 kg more weight loss. Repeated measurement analysis shows that after 16 weeks of metformin monotherapy, patients in the top tertile of mtDNA-CN consistently lost 1.21 kg more weight than those in the bottom tertile (P < 0.001). In comparison, mtDNA-CN is not significantly associated with acarbose-induced weight loss. CONCLUSIONS: Patients with higher mtDNA-CN are likely to lose more weight upon metformin treatment, suggesting mtDNA-CN as a potential novel biomarker for more effective weight management in type 2 diabetes.


Treatment of diabetes with the drug metformin can lead to beneficial weight loss. However, there is considerable variability in how patients respond to metformin and few markers or tests are available to guide prescribing. Here, we look at data from patients who took part in a trial comparing metformin with another diabetes drug and determine whether a particular marker­mitochondrial DNA copy number (mtDNA-CN)­is associated with weight loss with these treatments. mtDNA-CN is a proxy for the function of the mitochondria, an important organelle for the generation of metabolic energy in eukaryotic cells. Our results show that patients with diabetes with a higher mtDNA-CN lost more weight upon metformin treatment. This marker could potentially be used to guide treatment with metformin. Our findings warrant further exploration of mtDNA-CN as a marker of response to other drugs.

8.
Trends Endocrinol Metab ; 33(5): 318-332, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35249813

RESUMEN

CD44, a cell-surface glycoprotein, has long been studied as a cancer molecule due to its essential role in physiological activities in normal cells and pathological activities in cancer cells, such as cell proliferation, adhesion, and migration; angiogenesis; inflammation; and cytoskeleton rearrangement. Yet, recent evidence suggests a role of CD44 in metabolism, especially insulin resistance in obesity and diabetes. In line with the current concept of fibroinflammation in obesity and insulin resistance, CD44 as the main receptor of the extracellular matrix component, hyaluronan (HA), has been shown to regulate diet-induced insulin resistance in muscle and other insulin-sensitive tissues. In this review, we integrate current evidence for a role of CD44 in regulating glucose and lipid homeostasis and speculate about its involvement in the pathogenesis of chronic metabolic diseases, including obesity and diabetes. We summarize the current development of CD44-targeted therapies and discuss its potential for the use in treating metabolic diseases.


Asunto(s)
Resistencia a la Insulina , Glucosa , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Insulina , Obesidad
9.
Mol Metab ; 49: 101197, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33647469

RESUMEN

OBJECTIVE: Increased deposition of the extracellular matrix (ECM) in adipose tissue (AT) during obesity contributes to insulin resistance. The integrin receptors transmit changes in the extracellular environment causing corresponding intracellular adaptations. Integrin-linked kinase (ILK), an adaptor protein, is a central hub for intracellular signaling of integrins. This study determined the role of ILK in adipose function and insulin resistance. METHODS: The pathogenic role of ILK in obesity and insulin resistance was studied in human adipose tissue and adipocyte-specific ILK-deficient mice (ILKlox/loxAdCre). ILKlox/loxAdCre mice together with wild-type littermates (ILKlox/lox) were fed a chow diet or 60% high-fat (HF) diet for 16 weeks. In vivo insulin sensitivity was determined by hyperinsulinemic-euglycemic clamps. RESULTS: AT ILK expression was increased by HF diet feeding in mice and increased in visceral fat of morbidly obese humans. The HF-fed ILKlox/loxAdCre mice displayed reduced fat mass and improved glucose tolerance relative to the HF-fed ILKlox/lox mice. During a hyperinsulinemic-euglycemic clamp, the HF-fed ILKlox/loxAdCre mice exhibited partially improved insulin resistance in AT. Lipolysis was suppressed to a greater extent by insulin and glucose uptake in brown AT (BAT) increased. Increased inhibition of lipolysis may have been attributed to increased vascularization in white AT, while increased glucose uptake in BAT was associated with increased Akt phosphorylation and P38/JNK dephosphorylation. Notably, AT insulin sensitivity in lean mice was not affected by ILK deletion. Moreover, reduced fat mass in the HF-fed ILKlox/loxAdCre mice may have been attributed to decreased free fatty acid uptake into adipocytes via the downregulation of CD36 gene expression. Consistent with the results in the mice, knockdown and knockout of ILK in 3T3-L1 cells decreased lipid accumulation and CD36 gene expression during adipogenesis. CONCLUSIONS: These data show that adipocyte ILK is important for regulating HF diet-mediated insulin resistance in AT in a manner consistent with AT function.


Asunto(s)
Adipocitos/metabolismo , Resistencia a la Insulina/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Células 3T3-L1 , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa , Matriz Extracelular/metabolismo , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Insulina/metabolismo , Grasa Intraabdominal/metabolismo , Lipólisis , Masculino , Ratones , Obesidad Mórbida/metabolismo , Transducción de Señal
10.
Medicine (Baltimore) ; 99(37): e22170, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32925784

RESUMEN

BACKGROUND: Evidence suggests that metastasis is chiefly responsible for the poor prognosis of colon adenocarcinoma (COAD). The tumor microenvironment plays a vital role in regulating this biological process. However, the mechanisms involved remain unclear. The aim of this study was to identify crucial metastasis-related biomarkers in the tumor microenvironment and investigate its association with tumor-infiltrating immune cells. METHODS: We obtained gene expression profiles and clinical information from The Cancer Genome Atlas database. According to the "Estimation of STromal and Immune cells in MAlignant Tumor tissue using Expression data" algorithm, each sample generated the immune and stromal scores. Following correlation analysis, the metastasis-related gene was identified in The Cancer Genome Atlas database and validated in the GSE40967 dataset from Gene Expression Omnibus. The correlation between metastasis-related gene and infiltrating immune cells was assessed using the Tumor IMmune Estimation Resource database. RESULTS: The analysis included 332 patients; the metastatic COAD samples showed a low immune score. Correlation analysis results showed that interferon regulatory factor 1 (IRF1) was associated with tumor stage, lymph node metastasis, and distant metastasis. Furthermore, significant associations between IRF1 and CD8+ T cells, T cell (general), dendritic cells, T-helper 1 cells, and T cell exhaustion were demonstrated by Spearmans correlation coefficients and P values. CONCLUSIONS: The present findings suggest that IRF1 is associated with metastasis and the degree of immune infiltration of CD8+ T cells (general), dendritic cells, T-helper 1 cells, and T cell exhaustion in COAD. These results may provide information for immunotherapy in colon cancer.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/patología , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Factor 1 Regulador del Interferón/inmunología , Linfocitos T/inmunología , Biomarcadores de Tumor , Linfocitos T CD8-positivos/metabolismo , Bases de Datos Genéticas , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática/inmunología , Estadificación de Neoplasias , Pronóstico , Microambiente Tumoral
11.
Int J Mol Sci ; 21(16)2020 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-32785142

RESUMEN

Aberrant extracellular matrix (ECM) remodelling in muscle, liver and adipose tissue is a key characteristic of obesity and insulin resistance. Despite its emerging importance, the effective ECM targets remain largely undefined due to limitations of current approaches. Here, we developed a novel ECM-specific mass spectrometry-based proteomics technique to characterise the global view of the ECM changes in the skeletal muscle and liver of mice after high fat (HF) diet feeding. We identified distinct signatures of HF-induced protein changes between skeletal muscle and liver where the ECM remodelling was more prominent in the muscle than liver. In particular, most muscle collagen isoforms were increased by HF diet feeding whereas the liver collagens were differentially but moderately affected highlighting a different role of the ECM remodelling in different tissues of obesity. Moreover, we identified a novel association between collagen 24α1 and insulin resistance in the skeletal muscle. Using quantitative gene expression analysis, we extended this association to the white adipose tissue. Importantly, collagen 24α1 mRNA was increased in the visceral adipose tissue, but not the subcutaneous adipose tissue of obese diabetic subjects compared to lean controls, implying a potential pathogenic role of collagen 24α1 in obesity and type 2 diabetes.


Asunto(s)
Colágeno/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina , Grasa Intraabdominal/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , Animales , Colágeno/genética , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Expresión Génica , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , ARN Mensajero/genética
12.
Front Neurol ; 11: 591, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655488

RESUMEN

Posterior reversible encephalopathy syndrome (PRES) is a reversible neuroradiological syndrome characterized by reversible vasogenic edema. The pathophysiological mechanism is still unclear, but PRES may be triggered by various etiologies. To date, only a few PRES cases linked to cerebrospinal fluid (CSF) hypovolemia were reported. The association between PRES and CSF hypovolemia needs to be explored. We presented a case of PRES with CSF hypovolemia as a result of an inadvertent dural puncture and reviewed the literature to identify the clinical characterization and pathophysiological mechanism of PRES following CSF hypovolemia. A total of 31 cases of PRES-CSF hypovolemia was included for analysis. The median age was 33 years, with a notable female predominance (87.1%). Fifteen patients (48.4%) didn't have either a history of hypertension nor an episode of hypertension. The most common cause of CSF hypovolemia was epidural or lumbar puncture (n = 21), followed by CSF shunt (n = 6). The median interval between the procedure leading to CSF hypovolemia and PRES was 4 days. Seizure, altered mental state, and headache were the most frequent presenting symptom. The parietooccipital pattern was most frequent (71.0%). Conservative management remains the mainstay of treatment with excellent outcomes. Three patients had a second episode of PRES. CSF hypovolemia is a plausible cause of PRES via a unique pathophysiologic mechanism including arterial hyperperfusion and venous dysfunction. Patients with CSF hypovolemia is more susceptible to PRES, which is potentially life-threatening. Given that CSF hypovolemia is a common complication of anesthetic, neurological, and neurosurgical procedures, PRES should be early considered for prompt diagnosis and appropriate management.

13.
Cancer Cell Int ; 19: 282, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31728130

RESUMEN

BACKGROUND: Angiogenesis plays critical roles in the progression and metastasis of malignant tumors. Gastric neuroendocrine carcinoma is an uncommon stomach cancer that is rich in blood vessels and exhibits highly malignant biological behavior with a poor prognosis. The role of CDK5RAP3 in GNEC has not been reported to date. METHODS: Immunohistochemistry was used to assess the expression of CDK5RAP3 in GNEC tissues and adjacent non-tumor tissues. Cell lines with stable overexpression or knockdown of CDK5RAP3 were constructed using lentiviral transfection. Wound-healing assays, invasion and metastasis assays, tube formation assays, and tumor xenograft transplantation assays were performed to evaluate the effect of CDK5RAP3 on GNEC angiogenesis in vitro and in vivo. Real-time PCR, ELISA, western blot analysis, and confocal-immunofluorescence staining were used to explore the molecular mechanism of CDK5RAP3's effect on angiogenesis. RESULTS: Compared with their respective adjacent non-tumor tissues, protein levels of CDK5RAP3 were significantly decreased in GNEC tissues. Furthermore, low expression of CDK5RAP3 was correlated with more advanced TNM stage, increased tumor microvessel density, and poor prognosis. Functionally, we found that GNEC cells with CDK5RAP3 knockdown promoted human umbilical vein endothelial cells migration and tube formation via activation of AKT/HIF-1α/VEGFA signaling, resulting in increased levels of VEGFA in GNEC cell supernatant. In addition, CDK5RAP3 overexpression in GNEC cells caused the opposing effect. Consistent with these results, nude mouse tumorigenicity assays showed that CDK5RAP3 expression downregulated angiogenesis in vivo. Lastly, patients with low CDK5RAP3 expression and high VEGFA expression exhibited the worst prognosis. CONCLUSIONS: This study demonstrated that CDK5RAP3 inhibits angiogenesis by downregulating AKT/HIF-1α/VEGFA signaling in GNEC and improves patient prognosis, suggesting that CDK5RAP3 could be a potential therapeutic target for GNEC.

14.
J Exp Clin Cancer Res ; 38(1): 410, 2019 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-31533855

RESUMEN

BACKGROUND: UFM1 has been found to be involved in the regulation of tumor development. This study aims to clarify the role and potential molecular mechanisms of UFM1 in the invasion and metastasis of gastric cancer. METHODS: Expression of UFM1 in gastric tumor and paired adjacent noncancerous tissues from 437 patients was analyzed by Western blotting, immunohistochemistry, and realtime PCR. Its correlation with the clinicopathological characteristics and prognosis of gastric cancer patients was analyzed. The effects of UFM1 on the invasion and migration of gastric cancer cells were determined by the wound and trans-well assays, and the effect of UFM1 on subcutaneous tumor formation was verified in nude mice. The potential downstream targets of UFM1 and related molecular mechanisms were clarified by the human protein kinase assay and co-immunoprecipitation technique. RESULTS: Compared with the corresponding adjacent tissues, the transcription level and protein expression level of UFM1 in gastric cancer tissues were significantly downregulated (P < 0.05). The 5-year survival rate of gastric cancer patients with low UFM1 expression was significantly lower than the patients with high UFM1 expression (42.1% vs 63.0%, P < 0.05). The invasion and migration abilities of gastric cancer cells with stable UFM1 overexpression were significantly decreased, and the gastric cancer cells with UFM1 stable knockdown showed the opposite results; similar results were also obtained in the nude mouse model. Further studies have revealed that UFM1 could increase the ubiquitination level of PDK1 and decrease the expression of PDK1 at protein level, thereby inhibiting the phosphorylation level of AKT at Ser473. Additionally, the effect of UFM1 on gastric cancer cell function is dependent on the expression of PDK1. The expression level of UFM1 can improve the poor prognosis of PDK1 in patients with gastric cancer. CONCLUSION: UFM1 suppresses the invasion and metastasis of gastric cancer by increasing the ubiquitination of PDK1 through negatively regulating PI3K/AKT signaling.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas/metabolismo , Neoplasias Gástricas/patología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Oncol Rep ; 41(5): 2739-2752, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30864700

RESUMEN

Cyclin­dependent kinase 5 regulatory subunit­â€‹associated protein 3 (CDK5RAP3 or C53) is involved in the development of various types of tumor, and alternative splicing of C53 results in numerous transcription variants that encode different isoforms. The present study aimed to clone human C53 isoform d (IC53d) and explore its role in the proliferation of gastric cancer cells. Reverse transcription­quantitative polymerase chain reaction was used to detect the expression levels of IC53d in 80 primary gastric adenocarcinoma tissues and adjacent normal tissues. In addition, the association between IC53d and clinicopathological parameters was determined. Gastric cancer cell lines stably overexpressing IC53d were established to observe its effects on cell proliferation, invasion and migration, and on in vivo tumorigenicity, and the mechanism of action was explored. The results of the presen study demonstrated that IC53d was upregulated in gastric cancer tissues and was associated with tumor T­stage. Furthermore, overexpression of IC53d promoted the proliferation, colony formation and G1/S phase transition of gastric cancer cells, leading to enhancement of tumorigenesis in vitro and in vivo. Overexpression of IC53d also promoted phosphorylation of protein kinase B (AKT) and glycogen synthase kinase 3ß (GSK3ß), which increased the expression of cyclin D1. In addition, high cyclin D1 expression was associated with a significantly worse prognosis for patients compared with in patients with low cyclin D1 expression. These results indicated that IC53d may promote the phosphorylation of AKT and GSK3ß, which in turn may increase cyclin D1 expression, enhancing G1/S phase transition, accelerating cell cycle progression, promoting the proliferation of gastric cancer cells, and inducing a poor prognosis in patients with gastric cancer.


Asunto(s)
Adenocarcinoma/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Gástricas/patología , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/metabolismo , Femenino , Estudios de Seguimiento , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Estadificación de Neoplasias , Fosforilación , Pronóstico , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño , Transducción de Señal , Estómago/patología , Neoplasias Gástricas/mortalidad , Análisis de Supervivencia , Proteínas Supresoras de Tumor , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Cancer ; 9(23): 4404-4412, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30519346

RESUMEN

Purpose To discuss the relationship between the clinicopathological data, long-term survival of gastric cancer patients and different expression levels of Cyclin-Dependent Kinase 5 (CDK5) and Protein Phosphatase 2A (PP2A). Method The expression levels of CDK5 and PP2A were detected by immunohistochemistry in specimens from 124 patients with primary gastric cancer. The correlation among the expression of CDK5 and PP2A, clinicopathological factors and prognosis was investigated. Result The expression level of CDK5 was correlated with the TNM stage (p=0.030) and N stage (p=0.001), while the expression level of PP2A was correlated with the TNM stage and N stage (p=0.001 and p=0.004) as well as the degree of differentiation (p=0.046). The expression of CDK5 was positively correlated with the expression of PP2A in gastric cancer. Co-expression of CDK5 and PP2A is an independent prognostic factor that affected overall survival, and provided more accurate prognostic value for the overall survival of gastric cancer patients. Conclusion The expression of CDK5 and PP2A is positively correlated in gastric cancer. Co-expression of CDK5 and PP2A was an independent prognostic factor in patients with gastric cancer.

17.
World J Gastroenterol ; 24(34): 3898-3907, 2018 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-30228783

RESUMEN

AIM: To investigate the effects of different levels of expression of CDK5RAP3 and DDRGK1 on long-term survival of patients undergoing radical gastrectomy. METHODS: The expression of CDK5RAP3 and DDRGK1 was detected by immunohistochemistry in 135 patients who received standard gastrectomy were enrolled in the study. Western Blot was used to detect the expression of CDK5RAP3 and DDRGK1 in gastric cancer and its adjacent tissues and cell lines. The correlations between the expression of CDK5RAP3 and DDRGK1 and clinicopathological factors were analyzed, and the value of each parameter to the prognosis of the patients was compared. Receiver operating characteristic analysis was used to compare the accuracy of the prediction of clinical outcome by the parameters. RESULTS: CDK5RAP3 and DDRGK1 expression was down-regulated in the gastric cancer compared to its respective adjacent non-tumor tissues. The expression of CDK5RAP3 was closely related to the age of the patients (P = 0.035) and the T stage of the tumor (P = 0.017). The expression of DDRGK1 was correlated with the sex of the patients (P = 0.080), the degree of tumor differentiation (P = 0.036), the histological type (P = 0.036) and the N stage of the tumor (P = 0.014). Low expression CDK5RAP3 or DDRGK1 is a poor prognostic factor for gastric cancer patients. Prognostic analysis showed that the co-expression of CDK5RAP3 and DDRGK1 was an independent prognostic factor correlating with the overall survival of gastric cancer patients. Combined expression analysis of CDK5RAP3 and DDRGK1 may provide a more accurate prognostic value for overall survival. CONCLUSION: The co-expression of CDK5RAP3 and DDRGK1 is an independent prognostic factor for gastric cancer, which can provide a more accurate model for the long-term prognosis.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/metabolismo , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Gástricas/patología , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma/mortalidad , Adenocarcinoma/cirugía , Proteínas de Ciclo Celular , Femenino , Estudios de Seguimiento , Gastrectomía , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Estudios Retrospectivos , Estómago/patología , Estómago/cirugía , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/cirugía , Proteínas Supresoras de Tumor
18.
Zhongguo Zhong Yao Za Zhi ; 43(4): 772-778, 2018 Feb.
Artículo en Chino | MEDLINE | ID: mdl-29600654

RESUMEN

Glioblastoma is a common brain tumor and the overall survival rate of the patients is very low, so it is an effective way to develop the potential chemotherapy or adjuvant chemotherapy drugs in glioblastoma treatment. As a well-known antimalarial drug, artesunate(ARTs) has clear side effects, and recently it has been reported to have antitumor effects, but rarely reported in glioblastoma. Different concentrations of ARTs were used to treat the glioblastoma cells, and then the inhibitory effect of ARTs on glioblastoma proliferation was detected by MTT assay; Ki67 immunofluorescence assay was used to detect the proliferation of cells; Soft agar experiment was used to explain the clonal formation abilities in vitro; Flow Cytometry was used to detect the cell cycle; and Western blot assay was used to determine the expression of key cell cycle protein. MTT assay results indicated that ARTs-treated glioblastoma cell A172, U251, U87 were significantly inhibited in a time-and-dose dependent manner as compared to the control group(DMSO treatment group). Soft agar experiment showed that ARTs could significantly reduce the clonal formation ability of glioblastoma. Furthermore, Flow cytometry analysis showed that ARTs could obviously increase the cell proportion in G0/G1 phase and reduce the cell proportion in S phase. Western blot results showed that the expressions of cell cycle-related proteins CDK2, CDK4, cyclin D1 and cyclin B1 were all obviously down-regulated. Above all, ARTs may inhibit the proliferation of glioblastoma cells by arresting cell cycle in G0/G1 phase through down-regulating the expression of CDK2, CDK4, cyclin D1, cyclin B1. These results may not only provide a novel method for rediscovering and reusing ARTs but also provide a new potential drug for treating glioblastoma.


Asunto(s)
Antineoplásicos/farmacología , Artesunato/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Glioblastoma/patología , Apoptosis , Línea Celular Tumoral , Ciclina B1/metabolismo , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Glioblastoma/tratamiento farmacológico , Humanos
19.
Cell Signal ; 36: 222-229, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28506929

RESUMEN

Gastric cancer is difficult to cure due to its clinical heterogeneity and the complexity of its molecular mechanisms. KDM2B, a member of the JHDM family, functions as a histone lysine demethylase. However, the role and mechanisms of KDM2B in gastric cancer have not been elucidated. Here, we showed that KDM2B is commonly expressed in gastric cancer cells. The downregulation of KDM2B immediately induces autophagy, followed by the inhibition of proliferation. The compound 3-methyladenine (3-MA), an inhibitor of autophagy, largely rescues autophagy and the inhibition of cell proliferation induced by KDM2B knockdown. In this process, we observed a downregulation of the phosphorylation of Akt and its downstream effectors mTOR and p70S6K and an upregulation of Erk phosphorylation after KDM2B knockdown. In a xenograft model, the downregulation of KDM2B can inhibit tumour growth. The conversion of LC3-I to LC3-II also decreased concomitantly in vivo, which is a hallmark of autophagy. Taken together, our study was the first to demonstrate a novel regulatory role of KDM2B in autophagy and cell growth in gastric cancer cells. Our findings suggest that KDM2B may serve as a novel therapeutic target for gastric cancer therapy.


Asunto(s)
Autofagia , Proteínas F-Box/metabolismo , Técnicas de Silenciamiento del Gen , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones SCID , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...