Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Exp Med ; 221(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38695876

RESUMEN

Platinum-based chemotherapy drugs can lead to the development of anorexia, a detrimental effect on the overall health of cancer patients. However, managing chemotherapy-induced anorexia and subsequent weight loss remains challenging due to limited effective therapeutic strategies. Growth differentiation factor 15 (GDF15) has recently gained significant attention in the context of chemotherapy-induced anorexia. Here, we report that hepatic GDF15 plays a crucial role in regulating body weight in response to chemo drugs cisplatin and doxorubicin. Cisplatin and doxorubicin treatments induce hepatic Gdf15 expression and elevate circulating GDF15 levels, leading to hunger suppression and subsequent weight loss. Mechanistically, selective activation by chemotherapy of hepatic IRE1α-XBP1 pathway of the unfolded protein response (UPR) upregulates Gdf15 expression. Genetic and pharmacological inactivation of IRE1α is sufficient to ameliorate chemotherapy-induced anorexia and body weight loss. These results identify hepatic IRE1α as a molecular driver of GDF15-mediated anorexia and suggest that blocking IRE1α RNase activity offers a therapeutic strategy to alleviate the adverse anorexia effects in chemotherapy.


Asunto(s)
Anorexia , Doxorrubicina , Endorribonucleasas , Factor 15 de Diferenciación de Crecimiento , Hígado , Proteínas Serina-Treonina Quinasas , Pérdida de Peso , Proteína 1 de Unión a la X-Box , Animales , Humanos , Ratones , Anorexia/inducido químicamente , Anorexia/metabolismo , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Doxorrubicina/efectos adversos , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Factor 15 de Diferenciación de Crecimiento/efectos adversos , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos , Proteína 1 de Unión a la X-Box/metabolismo , Proteína 1 de Unión a la X-Box/genética
2.
Lipids Health Dis ; 23(1): 81, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509584

RESUMEN

BACKGROUND: Obesity is associated with extensive white adipose tissue (WAT) expansion and remodeling. Healthy WAT expansion contributes to the maintenance of energy balance in the liver, thereby ameliorating obesity-related hepatic steatosis. Tissue-resident mesenchymal stromal cell populations, including PDGFRß + perivascular cells, are increasingly recognized pivotal as determinants of the manner in which WAT expands. However, the full array of regulatory factors controlling WAT stromal cell functions remains to be fully elucidated. Hypoxia-inducible factors (HIFs) are critical regulators in WAT stromal cell populations such as adipocyte precursor cells (APCs). It is revealed that HIF1α activation within PDGFRß + stromal cells results in the suppression of de novo adipogenesis and the promotion of a pro-fibrogenic cellular program in obese animals. However, the role of HIF2α in PDGFRß + cells remains undetermined in vivo. METHODS: New genetic models were employed in which HIF1α (encoded by the Hif1a gene) and HIF2α (encoded by the Epas1 gene) are selectively inactivated in PDGFRß + cells in an inducible manner using tamoxifen (TAM). With these models, both in vitro and in vivo functional analysis of PDGFRß + cells lacking HIF proteins were performed. Additionally, comprehensive metabolic phenotyping in diet-induced mouse models were performed to investigate the roles of PDGFRß + cell HIF proteins in WAT remodeling, liver energy balance and systemic metabolism. RESULTS: Unlike HIF1α inactivation, the new findings in this study suggest that inducible ablation of HIF2α in PDGFRß + cells does not cause apparent effects on WAT expansion induced by obesogenic diet. The adipogenic ability of PDGFRß + APCs is not significantly altered by genetic HIF2α ablation. Moreover, no difference of key parameters associated with healthy WAT remodeling such as improvements of WAT insulin sensitivity, reduction in metabolic inflammation, as well as changes in liver fat accumulation or systemic glucose metabolism, is detected in PDGFRß + cell Epas1-deficient mice. CONCLUSION: The new findings in this study support that, in contrast to HIF1α, PDGFRß + cell HIF2α appears dispensable for WAT metabolic remodeling and the resulting effects on liver metabolic homeostasis in diet-induced obesity, underscoring the isoform-specific roles of HIFα proteins in the regulation of adipose tissue biology.


Asunto(s)
Tejido Adiposo Blanco , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Obesidad , Animales , Ratones , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Lípidos , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
3.
FASEB J ; 37(9): e23109, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37527216

RESUMEN

LAMA5 (laminin α5) is a member of the laminin family. Despite the recent research implicating LAMA5 in cancer, the function of LAMA5 has remained uncertain in the progression of ovarian cancer (OC). Here, we investigated the functional influences of LAMA5 knockdown on OC in vitro and in vivo. In this study, we used immunohistochemistry (IHC) analysis to detect the relative expression of LAMA5 in OC and non-cancer tissues, and we analyzed its connection with the overall survival (OS) of OC patients. To prove the role of LAMA5 in cell proliferation, migration, and invasion, LAMA5 expression in OC cell lines was inhibited by lentivirus. Compared with normal fallopian tube tissue, epithelial ovarian cancer (EOC) tissue showed critically higher LAMA5 expression levels; additionally, high LAMA5 levels were a poor predictor of OS. We found that cell progression was restrained in LAMA5-knockdown OC cell lines in vivo and in vitro. Finally, LAMA5 might be a commanding inducer of the expression of epithelial-mesenchymal transition (EMT) and Notch signaling pathway-related markers. Together, our research indicates that LAMA5 is highly connected to OC progression as it may play a role in the EMT process through the Notch signaling pathway.


Asunto(s)
Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Transducción de Señal , Proliferación Celular/genética , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica
4.
Pathol Oncol Res ; 28: 1610258, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250390

RESUMEN

[This corrects the article DOI: 10.3389/pore.2021.1609855.].

5.
Cancer Commun (Lond) ; 41(12): 1314-1330, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34699691

RESUMEN

Over the past few years, immune checkpoint inhibitors (ICIs) have greatly improved the survival for patients with non-small cell lung cancer (NSCLC) without driver mutations. Compared with wild-type tumors, tumors with epidermal growth factor receptor (EGFR) mutations show more heterogeneity in the expression level of programmed cell death-ligand 1 (PD-L1), tumor mutational burden (TMB), and other immune microenvironment characteristics. Whether ICIs are suitable for NSCLC patients with EGFR mutations is still worth exploring. In previous studies, no significantly improved benefits were observed with immunotherapy monotherapy in NSCLC patients with EGFR mutation. Here, we summarized and analyzed data from the clinical trials of ICIs or combined therapy in NSCLC patients with EGFR mutations. We also focused on the mechanisms affecting the efficacy of ICIs in NSCLC patients with EGFR mutations, the characteristics of potential responders, and provided insights into areas worth further investigations in future studies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Antígeno B7-H1/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Humanos , Inhibidores de Puntos de Control Inmunológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutación , Microambiente Tumoral
6.
Pathol Oncol Res ; 27: 1609855, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512203

RESUMEN

Survival is low in ovarian cancer (OC). Most OC patients demonstrate advanced metastases, and recurrence is common. Dysregulation of laminin interactions is associated with cancer development. However, it is unknown whether laminin subunits can be considered as biomarkers for OC diagnosis, prognosis, and treatment. We used cBioPortal, GEO, ONCOMINE, GEPIA, Human Protein Atlas, Kaplan-Meier Plotter, TIMER, and Metascape to determine the associations among laminin expression, prognosis, and immune cell infiltration in OC. LAMA5, LAMB3, and LAMC2 mRNAs and LAMA3, LAMB1/B2/B3, and LAMC1/C2 proteins were overexpressed in OC tissues compared with normal ovaries. LAMA4, LAMB1, and LAMC1 mRNA upregulation was positively correlated with worse overall survival (OS) and progression-free survival (PFS) in OC. Elevated LAMA2 and LAMC2 mRNA expression levels were related to better PFS or OS, respectively. The results speculated that LAMA5 could potentially be a good prognostic factor in OC. Its expression proves valuable for predicting OS in patients diagnosed with stage Ⅳ and grade 3 OC and PFS in patients diagnosed with all OC stages or grades. LAMB3 and LAMC2 expression was correlated with platinum resistance development. ROC analysis of laminins in OC sets revealed that LAMA2/A4/A5, LAMB1/B2/B3, and LAMC2 could be used to differentiate between malignant tumors and non-neoplastic tissues. LAMA1/A5 and LAMC1 were significantly and negatively correlated with various tumor immune infiltrates (TILs), especially with dendritic cells, CD8+ T cells or neutrophil. LAMA4 and LAMB1 might be associated with tumor purity in OC. Overall, LAMA5 and LAMC1 could help predict OC survival and diagnosis and might be deemed important OC oncogenes.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Laminina/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Biomarcadores de Tumor/genética , Linfocitos T CD8-positivos/patología , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/patología , Femenino , Humanos , Estimación de Kaplan-Meier , Oncogenes/genética , Pronóstico , Regulación hacia Arriba/genética
7.
Pathol Oncol Res ; 27: 1610006, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975347

RESUMEN

Objective: To investigate whether serum Tie-1 (sTie-1) is a valuable marker for predicting progression and prognosis of cervical cancer. Methods: Enzyme-linked immunosorbent assay (ELISA) was used to detect serum sTie-1 concentrations in 75 cervical cancer patients, 40 cervical intraepithelial neoplasia (CIN) patients, and 55 healthy controls without cervical lesions, and sTie-1 levels were compared between the groups. Receiver operating characteristic curves was used to evaluate the diagnostic value of sTie-1. The relationship between sTie-1 concentrations in patients with cervical cancer and clinicopathological features and prognosis were analyzed, and the risk factors for postoperative recurrence were determined using univariate and multivariable Cox proportional hazards regression. Results: We found that sTie-1 concentrations gradually increased according to lesion severity (i.e., cancer vs. CIN; p < 0.05) and were significantly elevated in adenocarcinoma compared with healthy controls. sTie-1 levels strongly distinguished between cervical cancer patients and the healthy controls (area under the curve = 0.846; cut-off value = 1,882.64 pg/ml; sensitivity = 74.6%; specificity = 96.4%). Moreover, sTie-1 levels in cervical cancer patients were significantly associated with tumor size, advanced tumor stage, lymph node metastasis, and reduced 4-years progression-free survival. Cervical cancer patients with high sTie-1 concentrations had a 3.123-fold [95% confidence interval (CI): 1.087-8.971, p = 0.034] higher risk for tumor recurrence. Conclusions: Elevated sTie-1 levels in patients with cervical carcinoma were associated with tumor progression and poor prognosis, indicating that sTie-1 may be a valuable marker for predicting progression and prognosis of cervical cancer.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/sangre , Carcinoma de Células Escamosas/patología , Receptor TIE-1/sangre , Neoplasias del Cuello Uterino/patología , Adulto , Anciano , Progresión de la Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Pronóstico , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA