Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Exp Cell Res ; 419(1): 113279, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35810773

RESUMEN

Cholesterol homeostasis plays an important role in the maintenance of normal body functions. CYP27A1 is a key enzyme known to regulate cholesterol homeostasis, which catalyzes the conversion of cholesterol to 27-HC and has been implicated in the occurrence and metastasis of various cancer types. The present study aimed to explore the regulatory role of CYP27A1 in the development of clear cell renal cell carcinoma (ccRCC). In particular, the effect of CYP27A1 on the proliferation and migration of ccRCC cells was investigated. The construction of a stable 786-O cell line overexpressing CYP27A1/pLVX was mediated by lentiviral infection. The proliferative capacity was assessed using MTT and colony formation. Wound healing assay was used to measure cell migration. Production of intracellular cholesterol and 27-HC was detected by enzyme-linked immunosorbent assay. The LXRs/ABCA1 pathway of cholesterol metabolism regulation was studied by RT-qPCR and Western blotting analysis after cells were treated with stimulation agents of 27-HC or T0901317 and inhibition agents of siRNA or GSK2033. The results revealed that overexpression of CYP27A1 could increase the intracellular production of 27-HC and inhibit the proliferation and migration of 786-O cells. And the treatment of 786-O cells with 27-HC induced a similar effect. CYP27A1/27HC mediated activation of the liver X receptors (LXRs) could up-regulate the expression of ATP-binding cassette transporter A1 (ABCA1), further resulting in the reduction of intracellular cholesterol contents. All of these findings indicated a regulatory role of CYP27A1 in the proliferation and migration of ccRCC, via activating LXRs/ABCA1 to regulate cholesterol homeostasis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP , Carcinoma de Células Renales , Colestanotriol 26-Monooxigenasa , Neoplasias Renales , Receptores X del Hígado , Transportador 1 de Casete de Unión a ATP/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Proliferación Celular , Colestanotriol 26-Monooxigenasa/metabolismo , Colesterol , Humanos , Receptores X del Hígado/metabolismo
2.
Cell Commun Signal ; 20(1): 114, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35897036

RESUMEN

Metabolic reprogramming and immune escape play a major role in tumorigenesis. Increasing number of studies have shown that reprogramming of glutamine metabolism is a putative determinant of the anti-tumor immune response in the tumor microenvironment (TME). Usually, the predatory uptake of glutamine by tumor cells in the TME results in the limited utilization of glutamine by immune cells and affects the anti-tumor immune response. The cell-programmed glutamine partitioning also affects the anti-tumor immune response. However, the reprogramming of glutamine metabolism in tumors modulates immune escape by regulating tumor PD-L1 expression. Likewise, the reprogramming of glutamine metabolism in the immune cells also affects their immune function. Additionally, different types of glutamine metabolism inhibitors extensively regulate the immune cells in the TME while suppressing tumor cell proliferation. Herein, we discuss how metabolic reprogramming of tumor and immune cells regulates anti-tumor immune responses, as well as functional changes in different immune cells in the context of targeting tumor glutamine metabolism, which can better explain the potential of targeting glutamine metabolism in combination with immunotherapy for cancer. Video abstract.


Asunto(s)
Neoplasias , Microambiente Tumoral , Glutamina/metabolismo , Humanos , Inmunidad , Inmunoterapia/métodos , Neoplasias/metabolismo
3.
Acta Pharmacol Sin ; 43(11): 2841-2847, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35468993

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) is one of the pervasive side effects of chemotherapy, leading to poor quality of life in cancer patients. Discovery of powerful analgesics for CIPN is an urgent and substantial clinical need. Nerve growth factor (NGF), a classic neurotrophic factor, has been identified as a potential therapeutic target for pain. In this study, we generated a humanized NGF monoclonal antibody (DS002) that most effectively blocked the interaction between NGF and tropomyosin receptor kinase A (TrkA). We showed that DS002 blocked NGF binding to TrkA in a dose-dependent manner with an IC50 value of 6.6 nM; DS002 dose-dependently inhibited the proliferation of TF-1 cells by blocking the TrkA-mediated downstream signaling pathway. Furthermore, DS002 did not display noticeable species differences in its binding and blocking abilities. In three chemotherapy-induced rat models of CIPN, subcutaneous injection of DS002 produced a significant prophylactic effect against paclitaxel-, cisplatin- and vincristine-induced peripheral neuropathy. In conclusion, we demonstrate for the first time that an NGF inhibitor effectively alleviates pain in animal models of CIPN. DS002 has the potential to treat CIPN pain in the clinic.


Asunto(s)
Antineoplásicos , Enfermedades del Sistema Nervioso Periférico , Ratas , Animales , Factor de Crecimiento Nervioso , Anticuerpos Monoclonales/uso terapéutico , Calidad de Vida , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Dolor , Antineoplásicos/efectos adversos , Receptor trkA/metabolismo
4.
Cancer Cell Int ; 20(1): 538, 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33292266

RESUMEN

BACKGROUND: Bladder cancer (BC) is a commonly diagnosed malignant tumor in the urinary system, with a high morbidity and a high recurrence rate. Current studies indicated that metabolism-associated genes (MAGs) having critical roles in the etiology of BC. The present study aims to identify differentially expressed MAGs and construct a MAGs based prognostic risk signature for BC by using The Cancer Genome Atlas (TCGA) database and proteomics data. METHODS: RNA-sequence data from the TCGA database and proteomics data from our BC samples were used to identify differentially expressed MAGs and construct a MAGs based prognostic signature in BC. Subsequently, survival analysis and nomogram were used to evaluate the prognostic and predictive value of the MAGs based signature in BC. RNA isolation and reverse transcription­quantitative PCR (RT-qPCR) were further performed to investigate the expression levels of MAGs in BC cells and explore the relationship between MAGs and M2 tumor associated macrophages (TAMs) secreted transforming growth factor-ß1 (TGF-ß1) in BC cells. RESULTS: A total of 23 differentially expressed MAGs were identified and five MAGs were finally used to construct a MAGs based signature. Survival analysis revealed that the MAGs based signature was closely correlated with the survival outcomes of patients with BC. A nomogram with the MAGs based signature risk score and clinical features was also constructed to facilitate the individualized prediction of BC patients. RT-qPCR showed that five MAGs were significantly differentially expressed and the expression levels of three MAGs were positively correlated with M2 TAMs secreted TGF-ß1 in T24 cells. CONCLUSIONS: Our study identified novel prognostic MAGs and constructed a MAGs based signature, which can be used as an independent factor in evaluating the prognosis of patients with BC. Furthermore, M2 TAMs may promote the expression of MAGs via the TGF-ß1 signaling pathway in the microenvironment of BC. Further clinical trials and experimental explorations are needed to validate our observations in BC.

5.
Int J Cancer ; 145(5): 1280-1289, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31034097

RESUMEN

Penile squamous cell carcinoma (PSCC) is a malignancy that affects the skin and tissues of the penis, but the knowledge of pathogenesis and carcinogenesis is limited. Here, we characterize the PSCC genomic landscape using whole-exome sequencing. Of the 30 paired blood and tumor samples, we identified recurrent mutations in 11 genes; confirmed previous findings for FAT1 (4/30), HRAS (4/30), NOTCH1 (4/30), TP53 (3/30) and PIK3CA (3/30); and revealed novel candidate driver genes [CASP8 (4/30), SLITRK2 (3/30), FLG (3/30) and TRRAP (3/30)]. Our in vitro experiments suggested CASP8 was involved in mediating TRAIL-induced apoptosis of penile cancer cell lines. We also observed the frequently altered pathways for potential therapeutic implications: alterations in the Notch (30% of sample altered), RTK-RAS (26.7% altered) and Hippo (23.3% altered) pathways accounted for over 50% of tumors. The frequently altered genes (>10%) in these pathways were proved to be expressed in penile tumors by immunohistochemistry assay. These findings provide new insight into the mutational and pathway landscapes of PSCC and suggest potential novel therapeutic opportunities for this malignancy.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias del Pene/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/sangre , Carcinoma de Células Escamosas/epidemiología , Caspasa 8/biosíntesis , Caspasa 8/genética , China/epidemiología , Análisis Mutacional de ADN , Proteínas Filagrina , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Mutación , Neoplasias del Pene/sangre , Neoplasias del Pene/epidemiología , Transducción de Señal/genética , Secuenciación del Exoma
6.
Cell Physiol Biochem ; 45(5): 2054-2070, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29533936

RESUMEN

BACKGROUND/AIMS: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potential anti-cancer agent due to its selective toxicity. However, many human non-small cell lung cancer (NSCLC) cells are partially resistant to TRAIL, thereby limiting its clinical application. Therefore, there is a need for the development of novel adjuvant therapeutic agents to be used in combination with TRAIL. METHODS: In this study, the effect of N-acetyl-glucosamine (GlcNAc), a type of monosaccharide derived from chitosan, combined with TRAIL was evaluated in vitro and in vivo. Thirty NSCLC clinical samples were used to detect the expression of death receptor (DR) 4 and 5. After GlcNAc and TRAIL co-treatment, DR expression was determined by real-time PCR and western blotting. Cycloheximide was used to detect the protein half-life to further understand the correlation between GlcNAc and the metabolic rate of DR. Non-reducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis was used to detect receptor clustering, and the localization of DR was visualized by immunofluorescence under a confocal microscope. Furthermore, a co-immunoprecipitation assay was performed to analyze the formation of death-inducing signaling complex (DISC). O-linked glycan expression levels were evaluated following DR5 overexpression and RNA interference mediated knockdown. RESULTS: We found that the clinical samples expressed higher levels of DR5 than DR4, and GlcNAc co-treatment improved the effect of TRAIL-induced apoptosis by activating DR5 accumulation and clustering, which in turn recruited the apoptosis-initiating protease caspase-8 to form DISC, and initiated apoptosis. Furthermore, GlcNAc promoted DR5 clustering by improving its O-glycosylation. CONCLUSION: These results uncovered the molecular mechanism by which GlcNAc sensitizes cancer cells to TRAIL-induced apoptosis, thereby highlighting a novel effective agent for TRAIL-mediated NSCLC-targeted therapy.


Asunto(s)
Acetilglucosamina/farmacología , Apoptosis/efectos de los fármacos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/toxicidad , Células A549 , Acetilglucosamina/uso terapéutico , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Caspasa 8/metabolismo , Línea Celular Tumoral , Glicosilación/efectos de los fármacos , Humanos , Inmunoprecipitación , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Microscopía Confocal , Poli(ADP-Ribosa) Polimerasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Trasplante Heterólogo , Regulación hacia Arriba/efectos de los fármacos
7.
J Thromb Haemost ; 22(7): 1956-1972, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38554936

RESUMEN

BACKGROUND: Patients with cancer are at an increased risk of developing a hypercoagulative phenotype and venous thromboembolism. However, no clinical trial has yet confirmed that anticoagulant therapy improves cancer prognosis, and the mechanism underlying hypercoagulation in patients with bladder cancer is not well understood. OBJECTIVES: We hypothesized that the prognostic genes affect tumor progression via tumor-mediated coagulation. METHODS: We detected the most significant prognostic genes of bladder cancer with The Cancer Genome Atlas dataset and validated them in 2 Gene Expression Omnibus datasets and 1 ArrayExpress dataset. Immunohistochemical tests were performed on a cohort of 80 individuals to further examine the prognostic genes. For the most reliable prognostic gene, its influence on coagulation was evaluated with gene knockdown followed by next-generation sequencing and cellular and animal experiments. RESULTS: Depletion of microtubule interacting and trafficking domain containing 1 (MITD1), a major prognostic gene of bladder cancer, significantly increased the tissue factor (TF) expression. MITD1 deficiency led to cytokinesis arrest, which, in turn, promoted the TF expression via unfolded protein response and c-Jun. The knockdown of IRE1, an essential kinase of unfolded protein response or the inactivation of c-Jun using c-Jun N-terminal kinase inhibitors weakened MITD1 deficiency- or dithiothreitol-induced TF upregulation. Cells lacking MITD1 promoted coagulation and metastasis in the experimental metastasis assay. CONCLUSION: Our findings suggest the novel role of tumor prognostic genes upon the development of hypercoagulative phenotype and venous thromboembolism, thereby underlining the importance of anticoagulant therapy and shedding light on the therapeutic value of targeting MITD1 in bladder cancer.


Asunto(s)
Coagulación Sanguínea , Tromboplastina , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Humanos , Tromboplastina/metabolismo , Tromboplastina/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Pronóstico , Masculino , Bases de Datos Genéticas , Ratones Desnudos , Femenino , Transducción de Señal , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo
8.
Cancer Lett ; 593: 216964, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38762193

RESUMEN

Tumor-associated macrophages (TAMs) are important components of the tumor microenvironment (TME) and strongly associated with poor prognosis and drug resistance, including checkpoint blockade immunotherapy in solid tumor patients. However, the mechanism by which TAM affects immune metabolism reprogramming and immune checkpoint signalling pathway in the TME remains elusive. In this study we found that transforming growth factor-beta (TGF-ß) secreted by M2-TAMs increased the level of glycolysis in bladder cancer (BLCA) and played important role in PD-L1-mediated immune evasion through pyruvate kinase isoenzymes M2 (PKM2). Mechanistically, TGF-ß promoted high expression of PKM2 by promoting the nuclear translocation of PKM2 dimer in conjunction with phosphorylated signal transducer and activator of transcription (p-STAT3), which then exerted its kinase activity to promote PD-L1 expression in BLCA. Moreover, SB-431542 (TGF-ß blocker) and shikonin (PKM2 inhibitor) significantly reduced PD-L1 expression and inhibited BLCA growth and organoids by enhancing anti-tumor immune responses. In conclusion, M2-TAM-derived TGF-ß promotes PD-L1-mediated immune evasion in BLCA by increasing the PKM2 dimer-STAT3 complex nuclear translocation. Combined blockade of the TGF-ß receptor and inhibition of PKM2 effectively prevent BLCA progression and immunosuppression, providing a potential targeted therapeutic strategy for BLCA.


Asunto(s)
Antígeno B7-H1 , Proteínas de la Membrana , Escape del Tumor , Macrófagos Asociados a Tumores , Neoplasias de la Vejiga Urinaria , Animales , Humanos , Ratones , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Glucólisis , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Naftoquinonas , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Proteínas de Unión a Hormona Tiroide , Hormonas Tiroideas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/genética
9.
Biomed Mater ; 18(5)2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37399813

RESUMEN

Cancer severely threatens human health, which makes it particularly urgent to develop effective strategies for cancer diagnosis and therapy. Gene therapy and nucleic acid-based cancer diagnosis play important roles in cancer theranostic, but their applicability is challenged by the low cellular uptake and enzymatic degradation. In response, safe and efficient carrier metal-organic frameworks (MOFs) have been proposed. Zeolite imidazole frameworks (ZIFs), a promising MOF type, can easily encapsulate negatively charged nucleic acid while offering a high loading efficiency, adjustable structure, and conditional responsiveness (pH, adenosine triphosphate (ATP), or glutathione (GSH)). In this review, we studied recent articles on nucleic acid-loading ZIFs-based nanoplatforms in tumor theranostics on the Pubmed database, with a focus on the synthesis and applications in tumor diagnosis and treatment. The relevant favorable aspects, potential challenges, and future opportunities are also discussed in this review.


Asunto(s)
Estructuras Metalorgánicas , Neoplasias , Ácidos Nucleicos , Zeolitas , Humanos , Zeolitas/química , Neoplasias/diagnóstico , Neoplasias/terapia , Estructuras Metalorgánicas/química , Portadores de Fármacos/química
10.
J Exp Clin Cancer Res ; 42(1): 316, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38001512

RESUMEN

BACKGROUND: Cancer-associated fibroblasts (CAFs), the predominant stromal cell of tumor microenvironment (TME), play an important role in tumor progression and immunoregulation by remodeling extracellular matrix (ECM) and secreting cytokines. However, little is known about the details of the underlying mechanism in bladder cancer. METHODS: Bioinformatics analysis was performed to analyze the prognostic value of CAFs and CXCL12 using GEO, TCGA and SRA databases. The effects of CXCL12 on bladder cancer progression were investigated through in vitro and in vivo assays. The biological mechanism of the effect of CXCL12 on PDL1 were investigated using western blotting, immunoprecipitation, RT-PCR, immunofluorescence, mass spectrometry, protein stability, and flow cytometry. RESULTS: The results demonstrated that CAFs-derived CXCL12 promoted cancer cell migration and invasion and upregulated PDL1. Mechanistically, upon binding to its specific receptor, CXCL12 activated the downstream JAK2/STAT3 pathway and rapidly up-regulated the expression of deubiquitinase CYLD. CYLD deubiquitinated P62 causing P62 accumulation, which in turn inhibited the autophagic degradation of PDL1. In vivo experiments demonstrated that blocking CXCL12 inhibited tumor growth, reduced tumor PDL1 expression and increased immune cell infiltration. CONCLUSIONS: This study revealed a novel mechanism for the role of CXCL12 in P62-mediated PDL1 autophagic regulation. Combined application of CXCL12 receptor blocker and PD1/PDL1 blocker can more effectively inhibit PDL1 expression and enhance antitumor immune response. Targeting CAFs-derived CXCL12 may provide an effective strategy for immunotherapy in bladder cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias de la Vejiga Urinaria , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Citocinas/metabolismo , Pronóstico , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria/patología
11.
iScience ; 26(11): 108142, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37867947

RESUMEN

ALDH1A1 is one of the classical stem cell markers for bladder cancer. Lysine 2-hydroxyisobutyrylation (Khib) is a newfound modification to modulate the protein expression, and the underlying mechanisms of how ALDH1A1 was regulated by Khib modification in bladder cancer remains unknown. Here, ALDH1A1 showed a decreased K260hib modification, as identified by protein modification omics in bladder cancer. Decreasing ALDH1A1 expression significantly suppressed the proliferation, migration and invasion of bladder cancer cells. Moreover, K260hib modification is responsible for the activity of ALDH1A1 in bladder cancer, which is regulated by HDAC2/3. Higher K260hib modification on ALDH1A1 promotes protein degradation through chaperone-mediated autophagy (CMA), and ALDH1A1 K260hib could sensitize bladder cancer cells to chemotherapeutic drugs. Higher ALDH1A1 expression with a lower K260hib modification indicates a poor prognosis in patients with bladder cancer. Overall, we demonstrated that K260hib of ALDH1A1 can be used as a potential therapeutic target for bladder cancer treatment.

12.
Hum Cell ; 35(2): 649-664, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35044630

RESUMEN

As one of important components of tumor microenvironment, CAFs (cancer-associated fibroblasts) play a vital role in the development and metastasis of bladder cancer. The present study aimed to develop a CAFs-related gene signature to predict the prognosis of patients and the response to chemotherapy and immunotherapy based on research of multidatabase. Expression data and clinical information were obtained from TCGA and GEO databases. Different bioinformatic and statistical methods were combined to construct the robust CAFs-related gene signature for prognosis. The model was explored from four aspects: single-cell source, immune infiltration, correlation with cancer-related genes and pathways, and prediction of drug response. After screening, five genes (BNC2, LAMA2, MFAP5, NID1, and OLFML1) related to CAFs were used for constructing the signature to divide patients into high- and low-risk groups. Patients in low-risk group had better prognosis and multidatabase analysis confirmed the predictive value. The five genes were mainly expressed by fibroblasts and involved in regulation of pathways related with glycolysis, hypoxia, and epithelial-mesenchymal transition (EMT). BNC2, LAMA2, and NID1 were strongly associated with drug sensitivity. Moreover, the immunological status was different between high- and low-risk groups. High-risk patients had poor response to chemotherapy or immunotherapy. The CAFs-related gene signature might help to optimize risk stratification and provide a new insight in individual treatment for bladder cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer , Preparaciones Farmacéuticas , Neoplasias de la Vejiga Urinaria , Fibroblastos Asociados al Cáncer/patología , Transición Epitelial-Mesenquimal/genética , Humanos , Oncogenes , Preparaciones Farmacéuticas/metabolismo , Microambiente Tumoral/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética
13.
Cell Death Discov ; 7(1): 15, 2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33462221

RESUMEN

Programmed death receptor-ligand 1 (PD-L1) plays a crucial role in immune evasion by tumour cells. Most tumour cells exhibit energy dependency and acquire energy from glycolysis. However, the relationship between glucose metabolism and PD-L1 expression remains unclear. In this study, changes in PD-L1 expression in renal carcinoma cells were evaluated during glucose deficiency and recovery, and PD-L1 could inversely regulate glycolysis. In addition, the possible signalling pathways activated by a low level of glucose to regulate PD-L1 were tested experimentally. The results showed that glucose deficiency could upregulate PD-L1 expression in two renal cancer cell lines, 786-O and OS-RC-2. Although the native levels of PD-L1 differed in the two cell lines, the upregulated PD-L1 expression was repristinated after glucose recovery. Moreover, epidermal growth factor receptor (EGFR) expression was upregulated in both cell lines with glucose deficiency. The use of an EGFR inhibitor reversed the upregulation of PD-L1 expression induced by glucose deficiency and inhibited the phosphorylation of extracellular regulated protein kinases 1 and 2 (ERK1/2). EGFR activated by epidermal growth factor (EGF) induced PD-L1 expression and ERK1/2 phosphorylation. Furthermore, an ERK1/2 inhibitor inhibited the phosphorylation of c-Jun and decreased the elevated PD-L1 expression induced by glucose deficiency. In addition, this study also showed that 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFK-2/FBPase 3 or PFKFB3) mediated upregulation of the level of glycolysis to improve the adverse environment through PD-L1 induction. Therefore, glucose metabolism can regulate the expression of PD-L1 through the EGFR/ERK/c-Jun pathway in renal cancer, and elevated PD-L1 can also regulate glycolysis by improving the expression of PFKFB3. The findings of this study could provide a new multiple target treatment for renal cell carcinoma (RCC) therapy.

14.
Oncol Rep ; 45(1): 191-201, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33200223

RESUMEN

The prognosis­associated genes of urinary bladder cancer have been systematically investigated in the Pathology Atlas project based on The Cancer Genome Atlas data. However, the biological functions of most genes in bladder cancer remain unknown. The present study investigated the biological function of 12 of the most significant survival­associated genes (ABRACL, MITD1, ZNF524, EMP1, HSPB6, CXorf38, TRIM38, ZNF182, ZNF195, SPRN, PTPN6 and LIPT1) in urothelial cancer reported by the Pathology Atlas project, with respect to cell proliferation and migration. In vitro, proliferation and migration analyses of T24 cells were performed following the transfection of the 12 prognostic genes. The results were validated with a small interfering (si)RNA library. Immunohistochemistry (IHC) analysis of clinical samples was performed to determine the association between gene expression and tumor metastasis. Furthermore, RNA sequencing was used to investigate the downstream signals. Among the 12 prognostic genes, MIT­domain containing protein 1 (MITD1) transfection was demonstrated to inhibit T24 cell migration to a certain degree. Experiments performed with a 7­gene siRNA library demonstrated that MITD1 knockdown markedly upregulated cell migratory abilities. Mechanistically, the influence of MITD1 on cell signal transduction was assessed via RNA sequencing. Cell migration­associated genes, including KISS1, SPANXB1, SPINT1, PIWIL2, SNAI1, APLN and CTHRC1 were dysregulated. IHC analysis demonstrated that MITD1 protein expression was notably lower in metastatic lymph nodes compared with the primary tumors. Taken together, the results of the present study suggest that the prognostic gene, MITD1 may serve as a migration inhibitor, and be developed as a potential therapeutic target for improving the prognosis of bladder cancer.


Asunto(s)
Proteínas de la Membrana/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Neoplasias de la Vejiga Urinaria/genética , Anciano , Movimiento Celular , Supervivencia Celular , Femenino , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Persona de Mediana Edad , Pronóstico , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología
15.
Front Mol Biosci ; 8: 687305, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34805266

RESUMEN

Few studies have reported whether nutrients in the tumor microenvironment can regulate the expression of PD-L1. Since tumor cells are often situated in a low-glutamine environment, we investigated PD-L1 expression under glutamine deprivation in bladder cancer cells. PD-L1 expression and the activation of the EGFR/MEK/ERK/c-Jun signaling pathway under glutamine deprivation were investigated by qPCR, Western blot, and immunofluorescence analyses. C-Jun-mediated transcriptional regulation of the PD-L1 gene was assessed by ChIP. PD-L1 expression and activation of the EGFR/MEK/ERK/c-Jun signaling pathway were assessed in T24 cells, TCCSUP cells and BALB/c mice with or without glutamine supplementation. Additionally, the impact of PD-L1 expression under glutamine deprivation on the function of T cells was investigated by ELISA. The expression of PD-L1 and EGFR/MEK/ERK/c-Jun pathway activation were elevated by glutamine deprivation, and c-Jun was enriched in the enhancer region of PD-L1. The expression of PD-L1 was considerably impaired by inhibiting the EGFR/MEK/ERK/c-Jun pathway and was elevated by activating this signaling pathway. In addition, the elevated PD-L1 expression and MEK/ERK/c-Jun signaling pathway activation were reduced by glutamine supplementation in vitro and in vivo. PD-L1 upregulation by glutamine deprivation in bladder cancer cells could reduce IFN-γ production by T cells. The expression of PD-L1 was upregulated under glutamine deprivation through the EGFR/MEK/ERK/c-Jun pathway to impair T cell function.

16.
Front Oncol ; 11: 697894, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34327138

RESUMEN

Immunotherapy, especially PD-1/PD-L1 checkpoint blockade immunotherapy, has led tumor therapy into a new era. However, the vast majority of patients do not benefit from immunotherapy. One possible reason for this lack of response is that the association between tumors, immune cells and metabolic reprogramming in the tumor microenvironment affect tumor immune escape. Generally, the limited amount of metabolites in the tumor microenvironment leads to nutritional competition between tumors and immune cells. Metabolism regulates tumor cell expression of PD-L1, and the PD-1/PD-L1 immune checkpoint regulates the metabolism of tumor and T cells, which suggests that targeted tumor metabolism may have a synergistic therapeutic effect together with immunotherapy. However, the targeting of different metabolic pathways in different tumors may have different effects on tumor immune escape. Herein, we discuss the influence of glucose metabolism and glutamine metabolism on tumor immune escape and describe the theoretical basis for strategies targeting glucose or glutamine metabolism in combination with PD-1/PD-L1 checkpoint blockade immunotherapy.

17.
DNA Cell Biol ; 40(6): 798-810, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34030484

RESUMEN

Recent studies have revealed the significant role of TEA domain family member 4 (TEAD4) in the development and progression of cancer. However, the potential role of TEAD4 in the progression of bladder cancer (BC) remains to be explored. The aim of this study was to determine whether TEAD4 could serve as a pan-cancer predictor of the prognosis for BC. Based on data mined from public databases, expression levels and clinical value of TEAD4 were identified in BC and human pan-cancers. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis was performed to detect the TEAD4 expression levels in BC cell lines. Gene Set Enrichment Analysis (GSEA) was carried out for functional analysis in BC, and the relationship between infiltrating immune cells and TEAD4 expression was evaluated by the CIBERSORT algorithm in BC and pan-cancer data. TEAD4 was overexpressed and associated with poor prognosis in BC and several types of cancers. GSEA and CIBERSORT algorithm suggested that various pathways including immune-related pathways were enriched in TEAD4 high expression group and several immunocytes infiltrated were correlated with the expression of TEAD4. This study revealed TEAD4 is an immune regulating-related predictor of prognosis for BC and has generalization value in pan-cancer.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas Musculares/fisiología , Factores de Transcripción/fisiología , Neoplasias de la Vejiga Urinaria/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/fisiología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Factores de Transcripción de Dominio TEA
18.
Bioact Mater ; 6(2): 433-446, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32995671

RESUMEN

Bladder cancer is one of the concerning malignancies worldwide, which is lacking effective targeted therapy. Gene therapy is a potential approach for bladder cancer treatment. While, a safe and effective targeted gene delivery system is urgently needed for prompting the bladder cancer treatment in vivo. In this study, we confirmed that the bladder cancer had CD44 overexpression and small interfering RNAs (siRNA) with high interfere to Bcl2 oncogene were designed and screened. Then hyaluronic acid dialdehyde (HAD) was prepared in an ethanol-water mixture and covalently conjugated to the chitosan nanoparticles (CS-HAD NPs) to achieve CD44 targeted siRNA delivery. The in vitro and in vivo evaluations indicated that the siRNA-loaded CS-HAD NPs (siRNA@CS-HAD NPs) were approximately 100 nm in size, with improved stability, high siRNA encapsulation efficiency and low cytotoxicity. CS-HAD NPs could target to CD44 receptor and deliver the therapeutic siRNA into T24 bladder cancer cells through a ligand-receptor-mediated targeting mechanism and had a specific accumulation capacity in vivo to interfere the targeted oncogene Bcl2 in bladder cancer. Overall, a CD44 targeted gene delivery system based on natural macromolecules was developed for effective bladder cancer treatment, which could be more conducive to clinical application due to its simple preparation and high biological safety.

19.
Signal Transduct Target Ther ; 6(1): 315, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433803

RESUMEN

The evolution of coronaviruses, such as SARS-CoV-2, makes broad-spectrum coronavirus preventional or therapeutical strategies highly sought after. Here we report a human angiotensin-converting enzyme 2 (ACE2)-targeting monoclonal antibody, 3E8, blocked the S1-subunits and pseudo-typed virus constructs from multiple coronaviruses including SARS-CoV-2, SARS-CoV-2 mutant variants (SARS-CoV-2-D614G, B.1.1.7, B.1.351, B.1.617.1, and P.1), SARS-CoV and HCoV-NL63, without markedly affecting the physiological activities of ACE2 or causing severe toxicity in ACE2 "knock-in" mice. 3E8 also blocked live SARS-CoV-2 infection in vitro and in a prophylactic mouse model of COVID-19. Cryo-EM and "alanine walk" studies revealed the key binding residues on ACE2 interacting with the CDR3 domain of 3E8 heavy chain. Although full evaluation of safety in non-human primates is necessary before clinical development of 3E8, we provided a potentially potent and "broad-spectrum" management strategy against all coronaviruses that utilize ACE2 as entry receptors and disclosed an anti-coronavirus epitope on human ACE2.


Asunto(s)
Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Anticuerpos Monoclonales de Origen Murino/farmacología , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , SARS-CoV-2/inmunología , Enzima Convertidora de Angiotensina 2/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/inmunología , Antivirales/inmunología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Células Vero
20.
Dis Markers ; 2020: 8866730, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33456631

RESUMEN

BACKGROUND: In recent years, immune-associated genes (IAGs) have been documented as having critical roles in the occurrence and progression of muscle-invasive bladder cancer (MIBC). Novel immune-related biomarkers and a robust prognostic signature for MIBC patients are still limited. The study is aimed at developing an IAG-based signature to predict the prognosis of MIBC patients. METHODS: In the present study, we identified differentially expressed IAGs in MIBC by using transcriptomics data from The Cancer Genome Atlas (TCGA) database and proteomics data from our samples. We further constructed an IAG-based signature and evaluated its prognostic and predictive value by survival analysis and nomogram. Tumor Immune Estimation Resource (TIMER) was applied to explore the correlation between the IAG-based signature and immune cell infiltration in the microenvironment of MIBC. RESULTS: A total of 22 differentially expressed IAGs were identified, and 2 IAGs (NR2F6 and AHNAK) were used to establish a prognostic signature. Subsequently, survival analysis showed that high-risk scores were significantly correlated with poor overall survival (OS), progression-free survival (PFS), and disease-free survival (DFS) of MIBC patients. A prognostic nomogram was constructed by integrating clinical factors with the IAG-based signature risk score. In addition, the IAG-based signature risk score was positively associated with the infiltration of macrophages and dendritic cells in MIBC. CONCLUSIONS: We constructed and verified a novel IAG-based signature, which could predict the prognosis of MIBC and might reflect the status of the immune microenvironment of MIBC. Further studies in more independent clinical cohorts and further experimental exploration of the prognostic IAG-based signature are still needed.


Asunto(s)
Biomarcadores de Tumor/genética , Microambiente Tumoral/inmunología , Neoplasias de la Vejiga Urinaria/genética , Adulto , Anciano , Anciano de 80 o más Años , Anexina A6/genética , Anexina A6/metabolismo , Biomarcadores de Tumor/metabolismo , Femenino , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Músculos/patología , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Análisis de Supervivencia , Transcriptoma , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA