Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Immunology ; 160(4): 345-356, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32311768

RESUMEN

Macrophages are particularly abundant and play an important role throughout the tumor progression process, namely, tumor-associated macrophages (TAM) in the tumor microenvironment. TAM can be polarized to disparate functional phenotypes, the M1 and M2 macrophages. M1-like type macrophages are defined as pro-inflammatory cells involved in killing cancer cells, while M2-like type cells can specially promote tumor growth and metastasis, tissue remodeling and immunosuppression. In this study, we first found that integrin ß3 was highly expressed on the surface of TAM, both in vivo and in vitro, that displayed the M2-like characteristics. Under intervention of CYC or triptolide, the integrin ß3 inhibitors, the M2 polarization of TAM could be inhibited. Moreover, in the cell model of M2 polarization, either blockade or knockout/knockdown of integrin ß3 could also suppress macrophage M2 polarization, which suggested that the M2 polarization was dependent on integrin ß3. Using knockdown of peroxisome proliferator-activated receptor-γ (PPARγ), an M2 regulator, we found that expression and activation of PPARγ participated in M2 polarization that was mediated by integrin ß3. Finally, to verify the activity of integrin ß3 inhibitors on TAM in vivo, 4T1 tumor-bearing mice were treated with CYC or triptolide; in response, the M1/M2 ratio of TAM was up-regulated, while the infiltration of total lymphocytes into tumor tissue was not altered. In general, our study found a connection between integrin ß3 and macrophage polarization, which provides a strategy for facilitating M2 to M1 repolarization and reconstructing the tumor immune microenvironment.


Asunto(s)
Neoplasias de la Mama/inmunología , Integrina beta3/metabolismo , Neoplasias Mamarias Animales/inmunología , PPAR gamma/metabolismo , Macrófagos Asociados a Tumores/inmunología , Animales , Diferenciación Celular , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Células Th2/inmunología , Microambiente Tumoral , Regulación hacia Arriba
2.
Exp Cell Res ; 322(1): 30-8, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24440556

RESUMEN

Post-translational regulation plays a critical role in the control of cell growth and proliferation. The phosphorylation of peroxisome proliferator-activated receptor γ (PPARγ) is the most important post-translational modification. The function of PPARγ phosphorylation has been studied extensively in the past. However, the relationship between phosphorylated PPARγ1 and tumors remains unclear. Here we investigated the role of PPARγ1 phosphorylation in human fibrosarcoma HT1080 cell line. Using the nonphosphorylation (Ser84 to alanine, S84A) and phosphorylation (Ser84 to aspartic acid, S84D) mutant of PPARγ1, the results suggested that phosphorylation attenuated PPARγ1 transcriptional activity. Meanwhile, we demonstrated that phosphorylated PPARγ1 promoted HT1080 cell proliferation and this effect was dependent on the regulation of cell cycle arrest. The mRNA levels of cyclin-dependent kinase inhibitor (CKI) p21(Waf1/Cip1) and p27(Kip1) descended in PPARγ1(S84D) stable HT1080 cell, whereas the expression of p18(INK4C) was not changed. Moreover, compared to the PPARγ1(S84A), PPARγ1(S84D) up-regulated the expression levels of cyclin D1 and cyclin A. Finally, PPARγ1 phosphorylation reduced sensitivity to agonist rosiglitazone and increased resistance to anticancer drug 5-fluorouracil (5-FU) in HT1080 cell. Our findings establish PPARγ1 phosphorylation as a critical event in human fibrosarcoma growth. These findings raise the possibility that chemical compounds that prevent the phosphorylation of PPARγ1 could act as anticancer drugs.


Asunto(s)
Proliferación Celular , Resistencia a Antineoplásicos , Fibrosarcoma/patología , PPAR gamma/metabolismo , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Línea Celular Tumoral , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes cdc , Células HEK293 , Humanos , PPAR gamma/genética , Fosforilación , Activación Transcripcional
3.
J Colloid Interface Sci ; 660: 203-214, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38244489

RESUMEN

Recently, wearable electronic products and gadgets have developed quickly with the aim of catching up to or perhaps surpassing the ability of human skin to perceive information from the external world, such as pressure and strain. In this study, by first treating the cellulosic fiber (modal textile) substrate with (3-aminopropyl) triethoxysilane (APTES) and then covering it with conductive nanocomposites, a bionic corpuscle layer is produced. The sandwich structure of tactile corpuscle-inspired bionic (TCB) piezoresistive sensors created with the layer-by-layer (LBL) technology consists of a pressure-sensitive module (a bionic corpuscle), interdigital electrodes (a bionic sensory nerve), and a PU membrane (a bionic epidermis). The synergistic mechanism of hydrogen bond and coupling agent helps to improve the adhesive properties of conductive materials, and thus improve the pressure sensitive properties. The TCB sensor possesses favorable sensitivity (1.0005 kPa-1), a wide linear sensing range (1700 kPa), and a rapid response time (40 ms). The sensor is expected to be applied in a wide range of possible applications including human movement tracking, wearable detection system, and textile electronics.


Asunto(s)
Nanocompuestos , Silanos , Dispositivos Electrónicos Vestibles , Humanos , Electrónica , Nanocompuestos/química , Textiles
4.
Cell Rep ; 43(2): 113779, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38358891

RESUMEN

R-loops are three-stranded structures that can pose threats to genome stability. RNase H1 precisely recognizes R-loops to drive their resolution within the genome, but the underlying mechanism is unclear. Here, we report that ARID1A recognizes R-loops with high affinity in an ATM-dependent manner. ARID1A recruits METTL3 and METTL14 to the R-loop, leading to the m6A methylation of R-loop RNA. This m6A modification facilitates the recruitment of RNase H1 to the R-loop, driving its resolution and promoting DNA end resection at DSBs, thereby ensuring genome stability. Depletion of ARID1A, METTL3, or METTL14 leads to R-loop accumulation and reduced cell survival upon exposure to cytotoxic agents. Therefore, ARID1A, METTL3, and METTL14 function in a coordinated, temporal order at DSB sites to recruit RNase H1 and to ensure efficient R-loop resolution. Given the association of high ARID1A levels with resistance to genotoxic therapies in patients, these findings open avenues for exploring potential therapeutic strategies for cancers with ARID1A abnormalities.


Asunto(s)
Adenina/análogos & derivados , Estructuras R-Loop , ARN , Ribonucleasa H , Humanos , Inestabilidad Genómica , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Metiltransferasas/genética
5.
iScience ; 27(6): 110014, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38947512

RESUMEN

The NAD+-dependent deacetylase SIRT7 is a pivotal regulator of DNA damage response (DDR) and a promising drug target for developing cancer therapeutics. However, limited progress has been made in SIRT7 modulator discovery. Here, we applied peptide-based deacetylase platforms for SIRT7 enzymatic evaluation and successfully identified a potent SIRT7 inhibitor YZL-51N. We initially isolated bioactive YZL-51N from cockroach (Periplaneta americana) extracts and then developed the de novo synthesis of this compound. Further investigation revealed that YZL-51N impaired SIRT7 enzymatic activities through occupation of the NAD+ binding pocket. YZL-51N attenuated DNA damage repair induced by ionizing radiation (IR) in colorectal cancer cells and exhibited a synergistic anticancer effect when used in combination with etoposide. Overall, our study not only identified YZL-51N as a selective SIRT7 inhibitor from insect resources, but also confirmed its potential use in combined chemo-radiotherapy by interfering in the DNA damage repair process.

6.
Biochem Pharmacol ; 183: 114354, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33279498

RESUMEN

Tumor-associated macrophages (TAMs) are the most widely infiltrating immune cells in the tumor microenvironment (TME). Clinically, the number of TAMs is closely correlated with poor outcomes in multiple cancers. The biological actions of TAMs are complex and diverse, including mediating angiogenesis, promoting tumor invasion and metastasis, and building an immunosuppressive microenvironment. Given these pivotal roles of TAMs in tumor development, TAM-based strategies are attractive and used in certain tumor therapies, including inhibition of angiogenic signalling, blockade of the immune checkpoint, and macrophage enhancement phagocytosis. Several attempts to develop TAM-targeted agents have been made to deplete TAMs or reprogram the behaviour of TAMs. Some have shown a favourable curative effect in monotherapy, combination with chemotherapy or immunotherapy in clinical trials. Additionally, a new macrophage-based cell therapeutic technology was recently developed by equipping macrophages with CAR molecules. It is expected to break through barriers to solid tumor treatment. Although TAM-related studies have yielded positive antitumor outcomes, further investigations are needed to better characterize TAMs, which will benefit further establishment of novel strategies for tumor therapy. Here, we concisely summarize the functions of TAMs in the TME and comprehensively introduce the latest TAM-based regimens in cancer treatment.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Inmunosupresores/administración & dosificación , Inmunoterapia/tendencias , Neoplasias/tratamiento farmacológico , Receptores Quiméricos de Antígenos/antagonistas & inhibidores , Receptores Quiméricos de Antígenos/inmunología , Microambiente Tumoral/efectos de los fármacos , Macrófagos Asociados a Tumores/efectos de los fármacos
7.
Biochem Pharmacol ; 183: 114298, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33153969

RESUMEN

B7-H3 is an immune checkpoint molecule from the B7 superfamily. It has been widely studied in tumor immune evasion in certain types of cancer. In our preliminary study, we found that B7-H3 is specifically enriched in tumor-associated macrophages (TAMs) in triple-negative breast cancer (TNBC) patients and strongly correlated with poor clinical prognosis. However, the role of B7-H3 in breast cancer remains elusive. Our current study aims to explore the potential of B7-H3 as a novel target in TNBC therapy. Here, we demonstrated that B7-H3 enriched on TAMs is tightly correlated with TNBC clinical progression. B7-H3high TAMs exhibit great pro-metastatic and immunosuppressive functions by intriguing extracellular matrix (ECM) reconstruction and tumor angiogenesis, therefore helping tumor cell dissemination and dampening T-cell infiltration in tumor microenvironment (TME). Importantly, targeting blockade of B7-H3 by anti-B7-H3 antibody improves the tumor vasculature disorder, thereby enhancing chemotherapy and PD-1 therapy efficacy. In conclusion, our study establishes the correlation between B7-H3high TAMs and TNBC progression for the first time. By exploring the possibility of targeting B7-H3 expressed in both tumor cells and TAMs, we suggest that B7-H3 could be a promising target in clinical TNBC treatment.


Asunto(s)
Antígenos B7/biosíntesis , Sistemas de Liberación de Medicamentos/métodos , Neovascularización Patológica/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antígenos B7/antagonistas & inhibidores , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/biosíntesis , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Paclitaxel/administración & dosificación , Células RAW 264.7 , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/patología
8.
Adv Sci (Weinh) ; 7(22): 2001417, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33240752

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, in which the higher frequency of cancer stem cells (CSCs) correlates with the poor clinical outcome. An aberrant activation of CDK5 is found to associate with TNBC progression closely. CDK5 mediates PPARγ phosphorylation at its Ser 273, which induces CD44 isoform switching from CD44s to CD44v, resulting in an increase of stemness of TNBC cells. Blocking CDK5/pho-PPARγ significantly reduces CD44v+ BCSCs population in tumor tissues, thus abrogating metastatic progression in TNBC mouse model. Strikingly, diminishing stemness transformation reverses immunosuppressive microenvironment and enhances anti-PD-1 therapeutic efficacy on TNBC. Mechanistically, CDK5 switches the E3 ubiquitin ligase activity of PPARγ and directly protects ESRP1 from a ubiquitin-dependent proteolysis. This finding firstly indicates that CDK5 blockade can be a potent strategy to diminish stemness transformation and increase the response to PD-1 blockade in TNBC therapy.

10.
Nat Commun ; 8(1): 766, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974683

RESUMEN

Tumor-associated macrophages are increasingly viewed as a target of great relevance in the tumor microenvironment, because of their important role in cancer progression and metastasis. However, the endogenous regulatory mechanisms underlying tumor-associated macrophage differentiation remain largely unknown. Here, we report that caspase-1 promotes tumor-associated macrophage differentiation by cleaving peroxisome proliferator-activated receptor gamma (PPARγ) at Asp64, thus generating a 41 kDa fragment. This truncated PPARγ translocates to mitochondria, where it directly interacts with medium-chain acyl-CoA dehydrogenase (MCAD). This binding event attenuates MCAD activity and inhibits fatty acid oxidation, thereby leading to the accumulation of lipid droplets and promoting tumor-associated macrophage differentiation. Furthermore, the administration of caspase-1 inhibitors or the infusion of bone marrow-derived macrophages genetically engineered to overexpress murine MCAD markedly suppresses tumor growth. Therefore, targeting the caspase-1/PPARγ/MCAD pathway might be a promising therapeutic approach to prevent tumor progression.Tumor associated macrophages (TAMs) promote cancer progression. Here, the author show that caspase-1 promotes TAMs differentiation by attenuating medium-chain acyl-CoA dehydrogenase activity and that inhibition of this axis results in suppression of tumour growth in a transgenic mouse model of breast cancer.


Asunto(s)
Caspasa 1/metabolismo , Diferenciación Celular/inmunología , Macrófagos/inmunología , Mitocondrias/metabolismo , Neoplasias/metabolismo , PPAR gamma/metabolismo , Microambiente Tumoral/inmunología , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa/metabolismo , Animales , Inhibidores de Caspasas/farmacología , Metabolismo Energético , Ácidos Grasos/metabolismo , Femenino , Humanos , Metabolismo de los Lípidos , Células MCF-7 , Macrófagos/efectos de los fármacos , Ratones , Ratones Desnudos , Neoplasias/inmunología , Células THP-1
11.
Oncotarget ; 7(47): 76984-76994, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27769068

RESUMEN

Peroxisome proliferator-activating receptor γ (PPARγ), a transcription factor, is involved in many important biological processes, including cell terminal differentiation, survival and apoptosis. However, the role of PPARγ, which regulates tumour promoter and oncogene expression, is not well understood in hepatocellular carcinoma (HCC). In the present study, based on evidence from clinical samples that phosphorylation of PPARγ at Ser84 is up-regulated in human liver tumours, we confirmed that phosphorylation of PPARγ was also significantly increased in an HCC mouse model and was increased by Mitogen-activated protein kinase (MEK)/ Extracellular-signal-regulated kinases (ERK) kinase. Next, we performed an RNA microarray analysis, and our data indicated that dephosphorylation of PPARγ at Ser84 affects the expression of glycolysis-related genes and pro-proliferation genes, which supposedly promote proliferation of HCC cells. Using a chromatin immunoprecipitation (ChIP) assay, we demonstrated that the observed PPARγ-mediated induction of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 (PFKFB4) expression was directly modulated by the transcriptional activity of its promoter. Furthermore, using knockdown of PFKFB4, we elucidated that the stimulation of PPARγ phosphorylation on glycolysis and proliferation in HCC is dependent on PFKFB4. Together, these findings extend our understanding of how liver tumour cells reprogram their glycolytic pathways by post-translational modification of specific transcription factors and lay a foundation for the screening of new targets for the treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Glucólisis , Neoplasias Hepáticas/patología , PPAR gamma/metabolismo , Fosfofructoquinasa-2/genética , Serina/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Trasplante de Neoplasias , Fosfofructoquinasa-2/metabolismo , Fosforilación , Regiones Promotoras Genéticas
12.
Neurosci Lett ; 584: 208-13, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25449866

RESUMEN

Many studies have shown that chronic stress can cause neuronal damage and depression, but this exact mechanism still remains unknown. Neurons are vulnerable to lipid peroxidation-induced damage because the major part of neuronal cell membrane is polyunsaturated fatty acids that are substrate for reactive oxygen species. Since endogenous antioxidant defense systems normally eliminate production of reactive oxygen species, deficient antioxidant defense can cause oxidative stress-induced damage. In the present study, to understand the role of endogenous antioxidant defense in chronic stress-induced neuronal damage, we analyzed lipid peroxidation, total antioxidant capacity, and activities of catalase and glutathione peroxidase in frontal cortex, hippocampus and striatum of rats exposed to chronic unpredictable stress. We found that chronic unpredictable stress for four weeks in rats induced depressive-like behaviors such as anhedonia, despair and decreased exploration. Malondialdehyde, a lipid peroxidation product, is increased, but total antioxidant capacity, glutathione peroxidase activity and catalase activity are decreased in brain of rats exposed to chronic unpredictable stress. Our findings suggest that down regulation of endogenous antioxidant defense induces lipid peroxidation contributing a role to chronic stress and depression.


Asunto(s)
Antioxidantes/metabolismo , Encéfalo/metabolismo , Estrés Psicológico/metabolismo , Anhedonia , Animales , Catalasa/metabolismo , Cuerpo Estriado/metabolismo , Conducta Exploratoria , Lóbulo Frontal/metabolismo , Glutatión Peroxidasa/metabolismo , Hipocampo/metabolismo , Peroxidación de Lípido , Masculino , Ratas Sprague-Dawley , Estrés Psicológico/psicología
13.
J Med Food ; 15(8): 713-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22784184

RESUMEN

High-fat diets (HFDs) have been found to influence central nervous system development and to cause cognitive impairments in human epidemiologic studies, as well as in animal investigations. These adverse effects on learning and memory induced by an HFD have been associated with an impaired hippocampus, including hippocampal oxidative damage. Previously, we had found that α-lipoic acid (α-LA) could ameliorate the oxidative stress in non-neural organs (liver, jejunum, and spleen) induced by a 10-week HFD (21.2% fat) food regimen in mice. In this study, we investigated whether a 10-week HFD (21.2% fat) induced oxidative stress in the hippocampus or impaired spatial learning in mice and whether LA ameliorated these effects. The HFD was found to induce oxidative stress (a decrease in catalase activity, glutathione peroxidase activity, and total antioxidative capacity and an increase in malondialdehyde levels) in the mouse hippocampus. In addition, we found that the HFD impaired spatial recognition memory of mice in the Y-maze paradigm. Furthermore, the hippocampal oxidative stress and impaired spatial recognition memory of the mice were reduced in HFD diets supplemented with 0.1% LA. These findings suggest that LA, as a strong antioxidant, may help prevent HFD-induced learning impairments by ameliorating associated oxidative stress in the hippocampus.


Asunto(s)
Antioxidantes/farmacología , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/efectos adversos , Aprendizaje por Laberinto/efectos de los fármacos , Ácido Tióctico/farmacología , Animales , Grasas de la Dieta/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Masculino , Memoria/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA