Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Gene Ther ; 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39033218

RESUMEN

Nuclear factor kappa-B (NF-κB) is a nuclear transcription factor that plays a key factor in promoting inflammation, which can lead to the development of cancer in a long-lasting inflammatory environment. The activation of NF-κB is essential in the initial phases of tumor development and progression, occurring in both pre-malignant cells and cells in the microenvironment such as phagocytes, T cells, and B cells. In addition to stimulating angiogenesis, inhibiting apoptosis, and promoting the growth of tumor cells, NF-κB activation also causes the epithelial-mesenchymal transition, and tumor immune evasion. Therapeutic strategies that focus on immune checkpoint molecules have revolutionized cancer treatment by enabling the immune system to activate immunological responses against tumor cells. This review focused on understanding the NF-κB signaling pathway in the context of cancer.

2.
Aquat Toxicol ; 273: 107021, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38996480

RESUMEN

Permethrin (Per) is a widely used and frequently detected pyrethroid pesticide in agricultural products and the environment. It may pose potential toxicity to non-target organisms. Per has been reported to affect lipid homeostasis, although the mechanism is undefined. This study aims to explore the characteristic transcriptomic profiles and clarify the underlying signaling pathways of Per-induced lipid metabolism disorder in zebrafish liver. The results showed that environmental exposure to Per caused changes in the liver index, histopathology, and oxidative stress in zebrafish. Moreover, transcriptome results showed that Per heavily altered the pathways involved in metabolism, the immune system, and the endocrine system. We conducted a more in-depth analysis of the genes associated with lipid metabolism. Our findings revealed that exposure to Per led to a disruption in lipid metabolism by activating the KRAS-PPAR-GLUT signaling pathways through oxidative stress. The disruption of lipid homeostasis caused by exposure to Per may also contribute to obesity, hepatitis, and other diseases. The results may provide new insights for the risk of Permethrin to aquatic organisms and new horizons for the pathogenesis of hepatotoxicity.

3.
J Biol Chem ; 300(7): 107447, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38844134

RESUMEN

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.


Asunto(s)
Antígeno B7-H1 , Neoplasias Colorrectales , PPAR alfa , Escape del Tumor , PPAR alfa/metabolismo , PPAR alfa/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Humanos , Fosforilación , Animales , Ratones , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Sistema de Señalización de MAP Quinasas
4.
Mol Carcinog ; 63(8): 1436-1448, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38751009

RESUMEN

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.


Asunto(s)
Carcinogénesis , Neoplasias , Receptor de Muerte Celular Programada 1 , Humanos , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Carcinogénesis/inmunología , Carcinogénesis/genética , Animales , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología
5.
Eur J Pharmacol ; 972: 176565, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38599309

RESUMEN

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.


Asunto(s)
Antígeno B7-H1 , Inmunoterapia , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Humanos , Animales , Inmunoterapia/métodos , Ratones , Línea Celular Tumoral , PPAR delta/genética , PPAR delta/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias del Colon/inmunología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Neoplasias del Colon/genética , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Escape del Tumor/efectos de los fármacos , Ratones Endogámicos BALB C
6.
Med Oncol ; 41(5): 114, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38619661

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Neoplasias del Colon , Femenino , Humanos , Inmunoterapia , Ligandos , PPAR gamma
7.
Med Oncol ; 41(5): 124, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38652406

RESUMEN

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.


Asunto(s)
Ferroptosis , Neoplasias , Microambiente Tumoral , Humanos , Ferroptosis/efectos de los fármacos , Neoplasias/terapia , Neoplasias/patología , Neoplasias/metabolismo , Neoplasias/inmunología , Inmunoterapia/métodos , Animales , Hierro/metabolismo
8.
Med Oncol ; 41(5): 94, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38526625

RESUMEN

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.


Asunto(s)
Quinasa de la Caseína II , Neoplasias , Humanos , Quinasa de la Caseína II/metabolismo , Transducción de Señal/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
9.
Adv Sci (Weinh) ; 10(34): e2304068, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37870169

RESUMEN

Immune cells can protect against tumor progression by killing cancer cells, while aberrant expression of the immune checkpoint protein PD-L1 (programmed death ligand 1) in cancer cells facilitates tumor immune escape and inhibits anti-tumor immunotherapy. As a serine/threonine kinase, CK2 (casein kinase 2) regulates tumor progression by multiple pathways, while it is still unclear the effect of CK2 on tumor immune escape. Here it is found that ING4 induced PD-L1 autophagic degradation and inhibites non-small cell lung cancer (NSCLC) immune escape by increasing T cell activity. However, clinical analysis suggests that high expression of CK2 correlates with low ING4 protein level in NSCLC. Further analysis shows that CK2 induce ING4-S150 phosphorylation leading to ING4 ubiquitination and degradation by JFK ubiquitin ligase. In contrast, CK2 gene knockout increases ING4 protein stability and T cell activity, subsequently, inhibites NSCLC immune escape. Furthermore, the combined CK2 inhibitor with PD-1 antibody effectively enhances antitumor immunotherapy. These findings provide a novel strategy for cancer immunotherapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Antígeno B7-H1/metabolismo , Quinasa de la Caseína II/uso terapéutico , Inmunoterapia , Proteínas de Homeodominio , Proteínas de Ciclo Celular , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/uso terapéutico
10.
J Cell Biochem ; 124(8): 1145-1154, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37393598

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales , Pioglitazona , Tiazolidinedionas , Humanos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Regulación de la Expresión Génica , Pioglitazona/farmacología , PPAR gamma/agonistas , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Escape del Tumor , Proteínas Proto-Oncogénicas c-myc/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo
11.
Cancer Sci ; 114(7): 2871-2881, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37096255

RESUMEN

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.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Antígeno B7-H1 , PPAR gamma , Escape del Tumor
12.
Eur J Pharmacol ; 950: 175749, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37105516

RESUMEN

Blockade of PD-1/PD-L1 immune checkpoint could be an effective antitumor strategy for multiple types of cancer, but it is low response rate for colorectal cancer patients with unclear mechanism. Here we found that PPARγ agonist pioglitazone could reduce PD-L1 protein levels without effect on its gene expression. Further analysis showed that pioglitazone induced PD-L1 autophagic degradation in a PPARγ-dependent manner. Pioglitazone promoted PD-L1 translocation to lysosome by immunofluorescence analysis, which was associated with the increased binding of PPARγ to PD-L1. Moreover the combined pioglitazone with PD-1 antibody enhanced colorectal tumor immunotherapy, which was involved in reduced PD-L1 levels and increased CD8+ T cells. These findings suggest that PPARγ agonist could induce PD-L1 autophagic degradation resulting in increased colorectal tumor immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias Colorrectales , Humanos , Antígeno B7-H1/metabolismo , Pioglitazona/farmacología , PPAR gamma , Receptor de Muerte Celular Programada 1/metabolismo , Inmunoterapia/métodos , Neoplasias Colorrectales/tratamiento farmacológico
13.
Cancer Sci ; 114(5): 1882-1897, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36445120

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon , Proteínas de Unión al Calcio , Neoplasias del Colon , MicroARNs , Vía de Señalización Wnt , Humanos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/metabolismo
14.
Cell Death Discov ; 8(1): 427, 2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36289191

RESUMEN

Ferroptosis is triggered by intracellular iron leading to accumulation of lipid peroxidation consequent promotion of cell death. Cancer cell exhibits ability to evade ferroptosis by activation of antioxidant signaling pathways such as SLC7A11/GPX4 axis. In addition to transcriptional regulation on ferroptosis by NRF2, SREBP1, YAP, and p53, ferroptosis is modulated by ubiquitination or autophagic degradation. Moreover, zinc or Ca2+ could modulate ferroptosis by inducing lipid peroxidation and ferroptosis. Induction of ferroptosis enhances immune cell activity such as T cells or macrophages, which is associated with the release of DAMPs (damage-associated molecular patterns) and IFNγ. Therefore, combined immune checkpoint inhibitors with ferroptosis inducers effectively enhance antitumor immunotherapy, whereas induction of ferroptosis could impair T cell activity or survival, suggesting that rational combined therapy for cancer is essential. In this review, we discussed the regulatory role of ferroptosis on tumor progression and immunotherapy.

15.
Cell Commun Signal ; 19(1): 91, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34493296

RESUMEN

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.


Asunto(s)
Autofagia/genética , Inmunoterapia , Neoplasias/inmunología , Escape del Tumor/genética , Autofagia/inmunología , Antígeno B7-H1/genética , Humanos , Evasión Inmune/genética , Lisosomas/genética , Lisosomas/inmunología , Macrófagos/inmunología , Neoplasias/genética , Neoplasias/patología , Linfocitos T/inmunología , Escape del Tumor/inmunología
16.
Int J Biol Sci ; 17(13): 3281-3287, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512146

RESUMEN

The adaptive immune checkpoints such as PD-1(programmed death-1)/PD-L1 (programmed death-ligand 1) play an important role in cancer immunotherapy, whereas increasing evidence suggests that cancer cell evades immune surveillance by innate immune checkpoints such as SIRPα (signal-regulatory protein α)/CD47 (cluster of differentiation 47). In multiple types of cancer cells and solid tumor tissues, highly expressed CD47 protein level has been observed, which is triggered by some transcription factors including NFκB, Myc, and HIF. As a transmembrane protein, the binding of CD47 to SIRPα ligand on phagocytes results in phagocytosis resistance and cancer cell immune escape. In contrast, CD47-SIRPα interaction blockade enhances cancer cell clearance by phagocytes such as macrophages and dendritic cells (DCs) to activate an innate immune response, whereas this process could promote antigen cross-presentation by antigen present cells (APCs) leading to T cell priming, consequently, activates an adaptive antitumor immune response. In this review, we discussed the current SIRPα-CD47 axis-mediated cancer cell immune escape and immunotherapy, which could provide an effective antitumor strategy by the innate and adaptive immune response.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Inmunoterapia/métodos , Neoplasias/inmunología , Receptores Inmunológicos/metabolismo , Escape del Tumor , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/metabolismo , Neoplasias/terapia , Fagocitosis
17.
J Biol Chem ; 297(3): 100954, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34270958

RESUMEN

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.


Asunto(s)
Adenilato Quinasa/metabolismo , Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Glucosa/metabolismo , Glutamina/metabolismo , PPAR gamma/metabolismo , Animales , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Ratones Desnudos , Fosforilación
18.
J Cell Biochem ; 122(3-4): 394-402, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33164261

RESUMEN

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.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , PPAR delta/metabolismo , Western Blotting , Proliferación Celular/genética , Proliferación Celular/fisiología , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Proteínas HSP90 de Choque Térmico/genética , Células HT29 , Células HeLa , Humanos , Inmunoprecipitación , PPAR delta/genética , Fosforilación/genética , Fosforilación/fisiología , Estabilidad Proteica
19.
Cell Death Dis ; 11(11): 955, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33159034

RESUMEN

Programmed death ligand 1 (PD-L1, CD274) is an essential immune checkpoint protein that binds to programmed death 1 (PD-1) on T-lymphocytes. T cell plays a critical role in killing cancer cells while the cancer cell exhibits immune escape by the expression of PD-L1. The binding of PD-L1 to PD-1 inhibits T cell proliferation and activity, leading to tumor immunosuppression. Increasing evidence shows that PD-L1 protein undergoes degradation in proteasomes or lysosomes by multiple pathways, leading to enhanced immunotherapy for cancer. Although some specific drugs induce PD-L1 degradation and increase antitumor activity, the combination of these drugs with PD-L1/PD-1 blockade significantly enhances cancer immunotherapy. In this review, we have discussed the interaction of PD-L1 degradation with cancer immunotherapy.


Asunto(s)
Antígeno B7-H1/metabolismo , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Proteolisis , Animales , Humanos , Neoplasias/metabolismo , Neoplasias/patología
20.
Biochem Biophys Res Commun ; 532(4): 520-527, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32896379

RESUMEN

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.


Asunto(s)
Neoplasias del Colon/metabolismo , Dipeptidasas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Dipeptidasas/biosíntesis , Dipeptidasas/metabolismo , Retroalimentación Fisiológica , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Estabilidad Proteica , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...