Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 300(7): 107447, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38844134

RESUMO

A high level of PD-L1 in cancer cells promotes tumor immune escape and inhibits tumor immunotherapy. Although PD-L1 gene expression is upregulated by multiple pathways, its gene transcriptional repression is still unclear. Here we found that loss of PPARα, one of the peroxisome-proliferator-activated receptors (PPARs) family members, promoted colorectal tumor immune escape. Mechanistically, PPARα directly bound to the PD-L1 promoter resulting in its gene transcriptional repression, which in turn increased T cell activity, and PPARα agonist enhanced this event. However, ERK induced PPARα-S12 phosphorylation leading to blockade of PPARα-mediated PD-L1 transcriptional repression, and the combination of ERK inhibitor with PPARα agonist significantly inhibited tumor immune escape. These findings suggest that the ERK-PPARα pathway inhibited PD-L1 gene transcriptional repression and promoted colorectal tumor immune escape.

2.
Mol Carcinog ; 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38751009

RESUMO

Programmed cell death 1 (PD-1), a key immune checkpoint receptor, has been extensively studied for its role in regulating immune responses in cancer. However, recent research has unveiled a complex and dual role for PD-1 in tumorigenesis. While PD-1 is traditionally associated with immune cells, this article explores its expression in various cancer cells and its impact on cancer progression. PD-1's functions extend beyond immune regulation, as it has been found to both promote and suppress tumor growth, depending on the cancer type. These findings have significant implications for the future of cancer treatment and our understanding of the immune response in the context of cancer. This article calls for further research into the multifaceted roles of PD-1 to optimize its therapeutic potential and improve patient outcomes in the fight against cancer.

3.
J Cell Biochem ; 124(8): 1145-1154, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37393598

RESUMO

As a master transcription factor, c-Myc plays an important role in promoting tumor immune escape. In addition, PPARγ (peroxisome proliferator-activated receptor γ) regulates cell metabolism, inflammation, and tumor progression, while the effect of PPARγ on c-Myc-mediated tumor immune escape is still unclear. Here we found that cells treated with PPARγ agonist pioglitazone (PIOG) reduced c-Myc protein expression in a PPARγ-dependent manner. qPCR analysis showed that PIOG had no significant effect on c-Myc gene levels. Further analysis showed that PIOG decreased c-Myc protein half-life. Moreover, PIOG increased the binding of c-Myc to PPARγ, and induced c-Myc ubiquitination and degradation. Importantly, c-Myc increased PD-L1 and CD47 immune checkpoint protein expression and promoted tumor immune escape, while PIOG inhibited this event. These findings suggest that PPARγ agonist inhibited c-Myc-mediated tumor immune escape by inducing its ubiquitination and degradation.


Assuntos
Neoplasias Colorretais , Pioglitazona , Tiazolidinedionas , Humanos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Regulação da Expressão Gênica , Pioglitazona/farmacologia , PPAR gama/agonistas , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Evasão Tumoral , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo
4.
Cancer Sci ; 114(7): 2871-2881, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37096255

RESUMO

Blockade of the programmed death 1 (PD-1)/ programmed death ligand 1 (PD-L1) immune checkpoint could increase antitumor immunotherapy for multiple types of cancer, but the response rate of patients is about 10%-40%. Peroxisome proliferator activated receptor γ (PPARγ) plays an important role in regulating cell metabolism, inflammation, immunity, and cancer progression, while the mechanism of PPARγ on cancer cell immune escape is still unclear. Here we found that PPARγ expression exhibits a positive correlation with activation of T cells in non-small-cell lung cancer (NSCLC) by clinical analysis. Deficiency of PPARγ promoted immune escape of NSCLC by inhibiting T-cell activity, which was associated with increased PD-L1 protein level. Further analysis showed that PPARγ reduced PD-L1 expression independent of its transcriptional activity. PPARγ contains the microtubule-associated protein 1A/1B-light chain 3 (LC3) interacting region motif, which acts as an autophagy receptor for PPARγ binding to LC3, leading to degradation of PD-L1 in lysosomes, which in turn suppresses NSCLC tumor growth by increasing T-cell activity. These findings suggest that PPARγ inhibits the tumor immune escape of NSCLC by inducing PD-L1 autophagic degradation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Antígeno B7-H1 , PPAR gama , Evasão Tumoral
5.
Cancer Sci ; 114(5): 1882-1897, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36445120

RESUMO

Naked cuticle homolog 1 (NKD1), which is expressed at low levels in many tumors, is considered an inhibitor of the Wnt/ß-catenin pathway, but it is highly expressed in colon cancer and can promote colon cancer cell proliferation. miRNAs are involved in the occurrence and progression of many tumors. However, miRNAs that can regulate NKD1 and the mechanisms by which NKD1 regulates tumor progression remain ambiguous. This research aims to reveal the potential regulatory network of NKD1 in colon cancer. miRNA data downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were analyzed by bioinformatics to screen for potential miRNAs targeting NKD1. Let-7b-5p was found to inhibit proliferation, migration, and invasion of colon cancer cells targeting NKD1. Further studies suggested that let-7b-5p can modulate Wnt signaling activity, and the nuclear accumulation of ß-catenin was significantly restrained by let-7b-5p through targeting NKD1. Moreover, NKD1 could prohibit the expression of the APC protein. Further studies manifested that NKD1 bound to APC and promoted the ubiquitination degradation of APC through restraining the expression of the deubiquitinating enzyme USP15 and blocking the combination between USP15 and APC. Functionally, NKD1 enhanced the proliferation and migration of colon cancer cells by inhibiting APC expression. This research revealed a novel mechanism by which the let-7b-5p-NKD1-APC-ß-catenin signaling pathway inhibited colon cancer cell progression.


Assuntos
Proteína da Polipose Adenomatosa do Colo , Proteínas de Ligação ao Cálcio , Neoplasias do Colo , MicroRNAs , Via de Sinalização Wnt , Humanos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , MicroRNAs/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Proteína da Polipose Adenomatosa do Colo/metabolismo
6.
J Biol Chem ; 297(3): 100954, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34270958

RESUMO

Peroxisome proliferator-activated receptor δ (PPARδ) is a nuclear receptor transcription factor that plays an important role in the regulation of metabolism, inflammation, and cancer. In addition, the nutrient-sensing kinase 5'AMP-activated protein kinase (AMPK) is a critical regulator of cellular energy in coordination with PPARδ. However, the molecular mechanism of the AMPK/PPARδ pathway on cancer progression is still unclear. Here, we found that activated AMPK induced PPARδ-S50 phosphorylation in cancer cells, whereas the PPARδ/S50A (nonphosphorylation mimic) mutant reversed this event. Further analysis showed that the PPARδ/S50E (phosphorylation mimic) but not the PPARδ/S50A mutant increased PPARδ protein stability, which led to reduced p62/SQSTM1-mediated degradation of misfolded PPARδ. Furthermore, PPARδ-S50 phosphorylation decreased PPARδ transcription activity and alleviated PPARδ-mediated uptake of glucose and glutamine in cancer cells. Soft agar and xenograft tumor model analysis showed that the PPARδ/S50E mutant but not the PPARδ/S50A mutant inhibited colon cancer cell proliferation and tumor growth, which was associated with inhibition of Glut1 and SLC1A5 transporter protein expression. These findings reveal a new mechanism of AMPK-induced PPARδ-S50 phosphorylation, accumulation of misfolded PPARδ protein, and inhibition of PPARδ transcription activity contributing to the suppression of colon tumor formation.


Assuntos
Adenilato Quinase/metabolismo , Proliferação de Células , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Glucose/metabolismo , Glutamina/metabolismo , PPAR gama/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Camundongos Nus , Fosforilação
7.
J Cell Biochem ; 122(3-4): 394-402, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33164261

RESUMO

Epidermal growth factor receptor (EGFR) induces peroxisome-proliferator-activated receptor-δ (PPARδ)-Y108 phosphorylation, while it is unclear the effect of phosphorylation of PPARδ on cancer cell metabolism. Here we found that EGF treatment increased its protein stability by inhibiting its lysosomal dependent degradation, which was reduced by gefitinib (EGFR inhibitor) treatment. PPARδ-Y108 phosphorylation in response to EGF recruited HSP90 (heat shock protein 90) to PPARδ resulting in increased PPARδ stability. In addition, PPARδ-Y108 phosphorylation promoted cancer cell metabolism, proliferation, and chemoresistance. Therefore, this study revealed a novel molecular mechanism of EGFR/HSP90/PPARδ pathway-mediated cancer cell metabolism, proliferation, and chemoresistance, which provides a strategy for cancer treatment.


Assuntos
Receptores ErbB/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , PPAR delta/metabolismo , Western Blotting , Proliferação de Células/genética , Proliferação de Células/fisiologia , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/genética , Proteínas de Choque Térmico HSP90/genética , Células HT29 , Células HeLa , Humanos , Imunoprecipitação , PPAR delta/genética , Fosforilação/genética , Fosforilação/fisiologia , Estabilidade Proteica
8.
Cell Commun Signal ; 19(1): 91, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493296

RESUMO

Autophagy is catabolic process by degradation of intracellular components in lysosome including proteins, lipids, and mitochondria in response to nutrient deficiency or stress such as hypoxia or chemotherapy. Increasing evidence suggests that autophagy could induce immune checkpoint proteins (PD-L1, MHC-I/II) degradation of cancer cells, which play an important role in regulating cancer cell immune escape. In addition to autophagic degradation of immune checkpoint proteins, autophagy induction in immune cells (macrophages, dendritic cells) manipulates antigen presentation and T cell activity. These reports suggest that autophagy could negatively or positively regulate cancer cell immune escape by immune checkpoint protein and antigens degradation, cytokines release, antigens generation. These controversial phenomenon of autophagy on cancer cell immune evasion may be derived from different experimental context or models. In addition, autophagy maybe exhibit a role in regulating host excessive immune response. So rational combination with autophagy could enhance the efficacy of cancer immunotherapy. In this review, the current progress of autophagy on cancer immune escape is discussed. Video Abstract.


Assuntos
Autofagia/genética , Imunoterapia , Neoplasias/imunologia , Evasão Tumoral/genética , Autofagia/imunologia , Antígeno B7-H1/genética , Humanos , Evasão da Resposta Imune/genética , Lisossomos/genética , Lisossomos/imunologia , Macrófagos/imunologia , Neoplasias/genética , Neoplasias/patologia , Linfócitos T/imunologia , Evasão Tumoral/imunologia
9.
Biochem Biophys Res Commun ; 532(4): 520-527, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32896379

RESUMO

DPEP1 is highly expressed in the colorectal carcinoma tissues and colon cancer cells. However, the function and underlying mechanism of DPEP1 in the colon cancer cells are still poorly understood. Here, we found that transcription factor MYC could occupy on the DPEP1 promoter and activate its activities, and DPEP1 was up-regulated by MYC proteins in mRNA and protein levels in a dose-dependent manner in colon cancer cells. The expression levels of DPEP1 were positively correlated with that of MYC in colorectal tumor tissues. Moreover, Laser confocal images and Co-immunoprecipitation (Co-IP) revealed that DPEP1 and MYC proteins could bind to each other in the colon cancer cells. In turn, DPEP1 could enhance the stability of MYC proteins by extending the half-life of MYC proteins in colon cancer cells. Thus, DPEP1 and MYC proteins might form a positive feedback loop to maintain their high expression levels in colon cancer cells. In function, the MTT, EdU, Clone Formation assays and xenograft tumors assays demonstrated that DPEP1 could boost the proliferation of colon cancer cells through the DPEP1/MYC positive feedback loop in vitro and in vivo. Theoretically, DPEP1 may serve as a colon cancer biomarker and a novel target of colorectal carcinogenesis therapy.


Assuntos
Neoplasias do Colo/metabolismo , Dipeptidases/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Dipeptidases/biossíntese , Dipeptidases/metabolismo , Retroalimentação Fisiológica , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Estabilidade Proteica , Ativação Transcricional
10.
BMC Cancer ; 20(1): 199, 2020 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-32164602

RESUMO

BACKGROUND: The fine-needle aspiration (FNA) biopsy was broadly applied to clinical diagnostics evaluation for thyroid carcinomas nodule, while companioning with higher uncertainty rate (15~30%) to identify malignancy for cytological indeterminate cases. It is requirement to discover novel molecular biomarkers to differentiate malignant thyroid nodule more precise. METHODS: We employed weighted gene co-expression network analysis (WGCNA) to discover genes significantly associated with malignant histopathology for cytological indeterminate nodules. In addition, identified significantly genes were validated through another independently investigations of thyroid carcinomas patient's samples via cBioportal and Geipa. The key function pathways of significant genes involving were blast through GenClip. RESULTS: Twenty-four signature genes were identified significantly related to thyroid nodules malignancy. Furthermore, five novel genes with missense mutation, FN1 (R534P), PROS1((K200I), (Q571K)), SCEL (T320S), SLC34A2(T688M) and TENM1 (S1131F), were highlighted as potential biomarkers to rule out nodules malignancy. It was identified that the key functional pathways involving in thyroid carcinomas. CONCLUSION: These results will be helpful to better understand the mechanism of thyroid nodules malignant transformation and characterize the potentially biomarkers for thyroid carcinomas early diagnostics.


Assuntos
Biomarcadores Tumorais/genética , Redes Reguladoras de Genes , Mutação de Sentido Incorreto , Neoplasias da Glândula Tireoide/diagnóstico , Nódulo da Glândula Tireoide/diagnóstico , Biópsia por Agulha Fina , Proteínas de Transporte/genética , Detecção Precoce de Câncer , Feminino , Fibronectinas/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteínas do Tecido Nervoso/genética , Proteína S/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Tenascina/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Nódulo da Glândula Tireoide/genética , Nódulo da Glândula Tireoide/patologia
11.
Carcinogenesis ; 38(7): 748-755, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28419191

RESUMO

Malignant cancer cell uncontrolled growth depends on the persistent nutrient availability such as glucose and amino acids, which is required for cancer cell energetic and biosynthetic pathways. As a nuclear hormone receptor, peroxisome-proliferator-activated receptor δ (PPARδ) plays a critical role in inflammation and cancer, however, it is still unclear the regulatory mechanism of PPARδ on cancer cell metabolism. Here, we found that PPARδ directly regulated neutral amino acid transporter SLC1-A5 (solute carrier family 1 member 5) and glucose transporter-1 (Glut1) gene transcription, leading to uptake of glucose and amino acid, activation of mTOR signaling, and tumor progression. In contrast, silence of PPARδ or its antagonist inhibited this event. More importantly, PPARδ promoted cancer cell metabolic reprogramming resulting in chemoresistance, which was alleviated by PPARδ antagonist. These findings revealed a novel mechanism of PPARδ-mediated tumor progression, which provided a potential strategy for cancer treatment.


Assuntos
Sistema ASC de Transporte de Aminoácidos/genética , Transportador de Glucose Tipo 1/genética , Antígenos de Histocompatibilidade Menor/genética , Neoplasias/metabolismo , PPAR gama/genética , Serina-Treonina Quinases TOR/genética , Sistema ASC de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Animais , Vias Biossintéticas , Metabolismo Energético/genética , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Células HCT116 , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Antígenos de Histocompatibilidade Menor/metabolismo , Neoplasias/genética , Neoplasias/patologia , PPAR gama/metabolismo , Transcrição Gênica
12.
J Cell Biochem ; 118(6): 1556-1562, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27918085

RESUMO

Abundant nutrient availability including glucose and amino acids plays an important role in maintaining cancer cell energetic and biosynthetic pathways. As a nuclear receptor, peroxisome-proliferator-activated receptor α (PPARα) regulates inflammation and cancer progression, however, it is still unclear the interaction of PPARα with the cancer cell glucose metabolism. Here we found that PPARα reduced Glut1 (Glucose transporter 1) protein and gene levels in HCT-116, SW480, HeLa, and MCF-7 cancer cell lines. In contrast, silenced PPARα reversed this event. Further analysis shows that PPARα directly targeted the consensus PPRE motif of Glut1 promoter region resulting in Glut1 transcription repression. PPARα-mediated Glut1 transcription repression led to decreased influx of glucose in cancer cells. These findings revealed a novel mechanism of PPARα-mediated cancer cell Glut1 transcription repression. J. Cell. Biochem. 118: 1556-1562, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Regulação para Baixo , Transportador de Glucose Tipo 1/genética , Neoplasias/metabolismo , PPAR alfa/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , Neoplasias/genética , Regiões Promotoras Genéticas
13.
Carcinogenesis ; 37(2): 215-222, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718225

RESUMO

Dysregulated expression of epidermal growth factor receptor (EGFR) has been implicated in many cancer events, while peroxisome proliferator-activated receptor γ (PPARγ) negatively regulates cancer progression. The molecular mechanism of EGFR interaction with PPARγ is still unclear. Here, we found that nuclear EGFR induced phosphorylation of PPARγ at Tyr-74 leading to PPARγ ubiquitination and degradation by mouse double minute 2 (MDM2) ubiquitin ligase. PPARγ degradation by EGFR/MDM2 signaling resulted in accumulation of nuclear factor-kappaB (NF-κB)/p65 protein levels and increasing NF-κB activation. In contrast, PPARγ-Y74A mutant reversed this event. Moreover, PPARγ-Y74A mutant suppressed cell proliferation and increased chemotherapeutic agent-induced cancer cell sensitivity. Importantly, the clinical findings show that the nuclear phosphorylation of PPARγ-Y74 and EGFR expression in colonic cancer tissues was higher than that in control normal tissues. Thus, our study revealed a novel molecular mechanism that nuclear EGFR/NF-κB signaling promoted cell proliferation by destructing PPARγ function, which provides a novel strategy for cancer treatment.


Assuntos
Neoplasias do Colo/patologia , Receptores ErbB/metabolismo , NF-kappa B/metabolismo , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Ativação Enzimática/fisiologia , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia
14.
Tumour Biol ; 37(4): 4275-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26803518

RESUMO

The inhibitor of growth-4 (ING-4) belongs to the inhibitor of growth (ING) family that is a type II tumor suppressor gene including five members (ING1-5). As a tumor suppressor, ING4 inhibits tumor growth, invasion, and metastasis by multiple signaling pathways. In addition to that, ING4 can facilitate cancer cell sensitivity to chemotherapy and radiotherapy. Although ING4 loss is observed for many types of cancers, increasing evidences show that ING4 can be used for gene therapy. In this review, the recent progress of ING4 regulating tumorigenesis is discussed.


Assuntos
Proteínas de Ciclo Celular/genética , Terapia Genética , Proteínas de Homeodomínio/genética , Neoplasias/genética , Proteínas Supressoras de Tumor/genética , Apoptose/genética , Carcinogênese/genética , Proteínas de Ciclo Celular/uso terapêutico , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/uso terapêutico , Humanos , Terapia de Alvo Molecular , Neoplasias/terapia , Proteínas Supressoras de Tumor/uso terapêutico
15.
Cell Physiol Biochem ; 37(3): 1123-33, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26402430

RESUMO

BACKGROUND: microRNAs (miRNAs) are small non-coding RNAs and have been shown to play a crucial role in the colorectal cancer (CRC) tumorigenesis and progression. The aim of this study was to investigate the clinical significance and prognostic value of miR-140-5p in CRC. The exact functions and the underlying molecular mechanisms of miR-140-5p in CRC was further determined. METHODS: miR-140-5p expression was detected in CRC samples, their adjacent nontumor tissues as well as CRC cell lines by RT-qPCR. Cell proliferation was detected using CCK-8, and cell invasion and migration were evaluated using Transwell assay. The direct regulation of VEGFA by miR-140-5p was identified using luciferase reporter assay. RESULTS: miR-140-5p was significantly dowregulated in CRC tissues and cell lines. Downregulation of miR-140-5p was significantly correlated with advanced CRC stage and poorer overall survival. Both gain-of-function and loss of function studies demonstrated that miR-140-5p acted as a tumor suppressor by inhibiting cell proliferation, migration and invasion. Integrated analysis identified VEGFA as a direct and functional target gene of miR-140-5p. Silencing VEGFA by small interfering RNA (siRNA) resembled the phenotype resulting from ectopic miR-140-5p expression, while overexpression of VEGFA attenuated the effect of miR-140-5p on CRC cells. CONCLUSIONS: Our results suggested a tumor suppressive role of miR-140-5p in CRC tumorigenesis and progression by targeting VEGFA.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Regiões 3' não Traduzidas , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Masculino , Prognóstico , Análise de Sobrevida
16.
Med Oncol ; 41(5): 114, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38619661

RESUMO

As one of the peroxisome-proliferator-activated receptors (PPARs) members, PPARγ is a ligand binding and activated nuclear hormone receptor, which is an important regulator in metabolism, proliferation, tumor progression, and immune response. Increased evidence suggests that activation of PPARγ in response to ligands inhibits multiple types of cancer proliferation, metastasis, and tumor growth and induces cell apoptosis including breast cancer, colon cancer, lung cancer, and bladder cancer. Conversely, some reports suggest that activation of PPARγ is associated with tumor growth. In addition to regulating tumor progression, PPARγ could promote or inhibit tumor immunotherapy by affecting macrophage differentiation or T cell activity. These controversial findings may be derived from cancer cell types, conditions, and ligands, since some ligands are independent of PPARγ activity. Therefore, this review discussed the dual role of PPARγ on tumor progression and immunotherapy.


Assuntos
Neoplasias da Mama , Neoplasias do Colo , Feminino , Humanos , Imunoterapia , Ligantes , PPAR gama
17.
Med Oncol ; 41(5): 124, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38652406

RESUMO

Ferroptosis is a form of intracellular iron-dependent cell death that differs from necrosis, autophagy and apoptosis. Intracellular iron mediates Fenton reaction resulting in lipid peroxidation production, which in turn promotes cell death. Although cancer cell exhibit's ability to escape ferroptosis by multiple pathways such as SLC7A11, GPX4, induction of ferroptosis could inhibit cancer cell proliferation, migration and invasion. In tumor microenvironment, ferroptosis could affect immune cell (T cells, macrophages etc.) activity, which in turn regulates tumor immune escape. In addition, ferroptosis in cancer cells could activate immune cell activity by antigen processing and presentation. Therefore, ferroptosis could be an effective strategy for cancer therapy such as chemotherapy, radiotherapy, and immunotherapy. In this paper, we reviewed the role of ferroptosis on tumor progression and therapy, which may provide a strategy for cancer treatment.


Assuntos
Ferroptose , Neoplasias , Microambiente Tumoral , Humanos , Ferroptose/efeitos dos fármacos , Neoplasias/terapia , Neoplasias/patologia , Neoplasias/metabolismo , Neoplasias/imunologia , Imunoterapia/métodos , Animais , Ferro/metabolismo
18.
Med Oncol ; 41(5): 94, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38526625

RESUMO

Casein kinase II (CK2) is an enzyme with pleiotropic kinase activity that catalyzes the phosphorylation of lots of substrates, including STAT3, p53, JAK2, PTEN, RELA, and AKT, leading to the regulation of diabetes, cardiovascular diseases, angiogenesis, and tumor progression. CK2 is observed to have high expression in multiple types of cancer, which is associated with poor prognosis. CK2 holds significant importance in the intricate network of pathways involved in promoting cell proliferation, invasion, migration, apoptosis, and tumor growth by multiple pathways such as JAK2/STAT3, PI3K/AKT, ATF4/p21, and HSP90/Cdc37. In addition to the regulation of cancer progression, increasing evidence suggests that CK2 could regulate tumor immune responses by affecting immune cell activity in the tumor microenvironment resulting in the promotion of tumor immune escape. Therefore, inhibition of CK2 is initially proposed as a pivotal candidate for cancer treatment. In this review, we discussed the role of CK2 in cancer progression and tumor therapy.


Assuntos
Caseína Quinase II , Neoplasias , Humanos , Caseína Quinase II/metabolismo , Transdução de Sinais/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias/tratamento farmacológico , Microambiente Tumoral
19.
Eur J Pharmacol ; 972: 176565, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38599309

RESUMO

Blockade of PD-1/PD-L1 immune checkpoint is wildly used for multiple types of cancer treatment, while the low response rate for patients is still completely unknown. As nuclear hormone receptor, PPARδ (peroxisome-proliferator-activated receptor) regulates cell proliferation, inflammation, and tumor progression, while the effect of PPARδ on tumor immune escape is still unclear. Here we found that PPARδ antagonist GSK0660 significantly reduced colon cancer cell PD-L1 protein and gene expression. Luciferase analysis showed that GSK0660 decreased PD-L1 gene transcription activity. Moreover, reduced PD-L1 expression in colon cancer cells led to increased T cell activity. Further analysis showed that GSK0660 decreased PD-L1 expression in a PPARδ dependent manner. Implanted tumor model analysis showed that GSK0660 inhibited tumor immune escape and the combined PD-1 antibody with GSK0660 effectively enhanced colorectal cancer immunotherapy. These findings suggest that GSK0660 treatment could be an effective strategy for cancer immunotherapy.


Assuntos
Antígeno B7-H1 , Imunoterapia , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inibidores , Humanos , Animais , Imunoterapia/métodos , Camundongos , Linhagem Celular Tumoral , PPAR delta/genética , PPAR delta/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias do Colo/imunologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Neoplasias do Colo/genética , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Evasão Tumoral/efeitos dos fármacos , Camundongos Endogâmicos BALB C
20.
Onkologie ; 36(10): 598-601, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24107916

RESUMO

The peroxisome proliferator-activated receptor-γ (PPARγ) belongs to the nuclear hormone receptor superfamily, and is expressed in adipose tissue, skeletal muscle, spleen, heart, liver, placenta, lung, and ovary. PPARγ is a critical regulator of inflammation, adipocyte differentiation, glucose homeostasis, and tumorigenesis. The mechanism of PPARγ on tumor suppression is still unclear, but plenty of evidence shows that PPARγ provides new therapeutic targets for cancer. Here we give a review of how PPARγ and its ligands regulate tumorigenesis by different pathways.


Assuntos
Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , PPAR gama/metabolismo , Transdução de Sinais , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA