Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 81(23): 5963-5976, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34645611

RESUMEN

Colorectal adenocarcinoma is a leading cause of death worldwide, and immune infiltration in colorectal tumors has been recognized recently as an important pathophysiologic event. In this context, tumor-associated macrophages (TAM) have been related to chemoresistance to 5-fluorouracil (5-FU), the first-line chemotherapeutic agent used in treating colorectal cancers. Nevertheless, the details of this chemoresistance mechanism are still poorly elucidated. In the current study, we report that macrophages specifically overexpress dihydropyrimidine dehydrogenase (DPD) in hypoxia, leading to macrophage-induced chemoresistance to 5-FU via inactivation of the drug. Hypoxia-induced macrophage DPD expression was controlled by HIF2α. TAMs constituted the main contributors to DPD activity in human colorectal primary or secondary tumors, while cancer cells did not express significant levels of DPD. In addition, contrary to humans, macrophages in mice do not express DPD. Together, these findings shed light on the role of TAMs in promoting chemoresistance in colorectal cancers and identify potential new therapeutic targets. SIGNIFICANCE: Hypoxia induces HIF2α-mediated overexpression of dihydropyrimidine dehydrogenase in TAMs, leading to chemoresistance to 5-FU in colon cancers.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Resistencia a Antineoplásicos , Fluorouracilo/farmacología , Regulación Enzimológica de la Expresión Génica , Hipoxia/fisiopatología , Macrófagos Asociados a Tumores/enzimología , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Dihidrouracilo Deshidrogenasa (NADP)/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Tumorales Cultivadas , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Gastroenterology ; 161(6): 1813-1829, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34606846

RESUMEN

Chronic inflammation is a known risk factor for gastrointestinal cancer. The evidence that nonsteroidal anti-inflammatory drugs suppress the incidence, growth, and metastasis of gastrointestinal cancer supports the concept that a nonsteroidal anti-inflammatory drug target, cyclooxygenase, and its downstream bioactive lipid products may provide one of the links between inflammation and cancer. Preclinical studies have demonstrated that the cyclooxygenase-2-prostaglandin E2 pathway can promote gastrointestinal cancer development. Although the role of this pathway in cancer has been investigated extensively for 2 decades, only recent studies have described its effects on host defenses against transformed epithelial cells. Overcoming tumor-immune evasion remains one of the major challenges in cancer immunotherapy. This review summarizes the impacts of the cyclooxygenase-2-prostaglandin E2 pathway on gastrointestinal cancer development. Our focus was to highlight recent advances in our understanding of how this pathway induces tumor immune evasion.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Neoplasias Gastrointestinales/enzimología , Mediadores de Inflamación/metabolismo , Escape del Tumor , Microambiente Tumoral/inmunología , Animales , Antineoplásicos/uso terapéutico , Fibroblastos Asociados al Cáncer/enzimología , Fibroblastos Asociados al Cáncer/inmunología , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/inmunología , Neoplasias Gastrointestinales/patología , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/enzimología , Linfocitos Infiltrantes de Tumor/inmunología , Transducción de Señal , Escape del Tumor/efectos de los fármacos , Macrófagos Asociados a Tumores/enzimología , Macrófagos Asociados a Tumores/inmunología
3.
Front Immunol ; 12: 667850, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995401

RESUMEN

Proprotein convertases (PC) are a family of 9 serine proteases involved in the processing of cellular pro-proteins. They trigger the activation, inactivation or functional changes of many hormones, neuropeptides, growth factors and receptors. Therefore, these enzymes are essential for cellular homeostasis in health and disease. Nine PC subtilisin/kexin genes (PCSK1 to PCSK9) encoding for PC1/3, PC2, furin, PC4, PC5/6, PACE4, PC7, SKI-1/S1P and PCSK9 are known. The expression of PC1/3, PC2, PC5/6, Furin and PC7 in lymphoid organs such as lymph nodes, thymus and spleen has suggested a role for these enzymes in immunity. In fact, knock-out of Furin in T cells was associated with high secretion of pro-inflammatory cytokines and autoantibody production in mice. This suggested a key role for this enzyme in immune tolerance. Moreover, Furin through its proteolytic activity, regulates the suppressive functions of Treg and thus prevents chronic inflammation and autoimmune diseases. In macrophages, Furin is also involved in the regulation of their inflammatory phenotype. Similarly, PC1/3 inhibition combined with TLR4 stimulation triggers the activation of the NF-κB signaling pathway with an increased secretion of pro-inflammatory cytokines. Factors secreted by PC1/3 KD macrophages stimulated with LPS exert a chemoattractive effect on naive auxiliary T lymphocytes (Th0) and anti-tumoral activities. The link between TLR and PCs is thus very important in inflammatory response regulation. Furin regulates TL7 and TLR8 processing and trafficking whereas PC1/3 controls TLR4 and TLR9 trafficking. Since PC1/3 and Furin are key regulators of both the innate and adaptive immune responses their inhibition may play a major role in oncoimmune therapy. The role of PCs in the oncoimmune response and therapeutic strategies based on PCs inhibition are proposed in the present review.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Linfocitos Infiltrantes de Tumor/enzimología , Neoplasias/enzimología , Proproteína Convertasas/metabolismo , Microambiente Tumoral , Macrófagos Asociados a Tumores/enzimología , Animales , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Transducción de Señal , Receptores Toll-Like/metabolismo , Macrófagos Asociados a Tumores/inmunología
4.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33753566

RESUMEN

BACKGROUND: Regorafenib and other multikinase inhibitors may enhance antitumor efficacy of anti-program cell death-1 (anti-PD1) therapy in hepatocellular carcinoma (HCC). Its immunomodulatory effects, besides anti-angiogenesis, were not clearly defined. METHODS: In vivo antitumor efficacy was tested in multiple syngeneic liver cancer models. Murine bone marrow-derived macrophages (BMDMs) were tested in vitro for modulation of polarization by regorafenib and activation of cocultured T cells. Markers of M1/M2 polarization were measured by quantitative reverse transcription PCR (RT-PCR), arginase activity, flow cytometry, and ELISA. Knockdown of p38 kinase and downstream Creb1/Klf4 signaling on macrophage polarization were confirmed by using knockdown of the upstream MAPK14 kinase, chemical p38 kinase inhibitor, and chromatin immunoprecipitation. RESULTS: Regorafenib (5 mg/kg/day, corresponding to about half of human clinical dosage) inhibited tumor growth and angiogenesis in vivo similarly to DC-101 (anti-VEGFR2 antibody) but produced higher T cell activation and M1 macrophage polarization, increased the ratio of M1/M2 polarized BMDMs and proliferation/activation of cocultured T cells in vitro, indicating angiogenesis-independent immunomodulatory effects. Suppression of p38 kinase phosphorylation and downstream Creb1/Klf4 activity in BMDMs by regorafenib reversed M2 polarization. Regorafenib enhanced antitumor efficacy of adoptively transferred antigen-specific T cells. Synergistic antitumor efficacy between regorafenib and anti-PD1 was associated with multiple immune-related pathways in the tumor microenvironment. CONCLUSION: Regorafenib may enhance antitumor immunity through modulation of macrophage polarization, independent of its anti-angiogenic effects. Optimization of regorafenib dosage for rational design of combination therapy regimen may improve the therapeutic index in the clinic.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Macrófagos Asociados a Tumores/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Factor 4 Similar a Kruppel/metabolismo , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/inmunología , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/enzimología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Transducción de Señal , Microambiente Tumoral , Macrófagos Asociados a Tumores/enzimología , Macrófagos Asociados a Tumores/inmunología
5.
Cell Death Dis ; 12(2): 162, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33558466

RESUMEN

Hepatocellular carcinoma (HCC) is a common high-mortality cancer, mainly due to diagnostic difficulties during its early clinical stages. In this study, we aimed to identify genes that are important for HCC diagnosis and treatment, and we investigated the underlying mechanism of prognostic differences. Differentially expressed genes (DEGs) were identified by using the limma package, and receiver operating characteristic curve analysis was performed to identify diagnostic markers for HCC. Bioinformatics and clinical specimens were used to assess epithelial cell transforming 2 (ECT2) in terms of expression, prognostic value, pathways, and immune correlations. In vitro experiments were used to investigate the underlying mechanism and function of ECT2, and the results were confirmed through in vivo experiments. The integrated analysis revealed 53 upregulated DEGs, and one candidate biomarker for diagnosis (ECT2) was detected. High expression of ECT2 was found to be an independent prognostic risk factor for HCC. ECT2 expression showed a strong correlation with tumor-associated macrophages. We found that ECT2 overexpression increased the migration and proliferation of HCC cells. It also promoted the expression of PLK1, which subsequently interacted with PTEN and interfered with its nuclear translocation, ultimately enhancing aerobic glycolysis and promoting M2 macrophage polarization. M2 macrophages suppress the functions of NK cells and T cells, and this was confirmed in the in vivo experiments. Overall, ECT2 may promote the polarization of M2 macrophages by enhancing aerobic glycolysis and suppressing the functions of immune cells. ECT2 could serve as a candidate diagnostic and prognostic biomarker for HCC.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Proteínas de Ciclo Celular/metabolismo , Plasticidad de la Célula , Neoplasias Hepáticas/enzimología , Fosfohidrolasa PTEN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Macrófagos Asociados a Tumores/enzimología , Apoptosis , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/genética , Movimiento Celular , Proliferación Celular , Bases de Datos Genéticas , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Glucólisis , Células Hep G2 , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Fosfohidrolasa PTEN/genética , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Microambiente Tumoral , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/patología , Regulación hacia Arriba , Quinasa Tipo Polo 1
6.
Aging (Albany NY) ; 12(23): 24394-24423, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33260160

RESUMEN

FBXW7 functions as an E3 ubiquitin ligase to mediate oncoprotein degradation via the ubiquitin-proteasome system in cancer cells, effectively inhibiting the growth and survival of tumor cells. However, little is known about the functions of FBXW7 in macrophages and the tumor immune microenvironment. In this study, we find that FBXW7 suppresses M2-like tumor-associated macrophage (TAM) polarization to limit tumor progression. We identified a significant increase in the proportion of M2-like TAMs and aggravated tumor growth in mice with myeloid FBXW7 deficiency by subcutaneous inoculation with Lewis lung carcinoma cells (LLCs). When stimulated with LLCs supernatant in vitro, FBXW7-knockout macrophages displayed increased M2 macrophage polarization and enhanced ability of supporting cancer cells growth. In mechanism, we confirmed that FBXW7 inhibited M2-like TAM polarization by mediating c-Myc degradation via the ubiquitin-proteasome system. These findings highlight the role of FBXW7 in M2-like TAM polarization and provide new insights into the potential targets for cancer immunotherapies.


Asunto(s)
Carcinoma Pulmonar de Lewis/enzimología , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Macrófagos Asociados a Tumores/enzimología , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Proliferación Celular , Células Cultivadas , Proteína 7 que Contiene Repeticiones F-Box-WD/deficiencia , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Regulación Neoplásica de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal , Carga Tumoral , Microambiente Tumoral , Macrófagos Asociados a Tumores/patología , Ubiquitinación
7.
Front Immunol ; 11: 576310, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133093

RESUMEN

Shp1, encoded by the gene Ptpn6, is a protein tyrosine phosphatase that transduces inhibitory signals downstream of immunoreceptors in many immune cell types. Blocking Shp1 activity represents an exciting potential immunotherapeutic strategy for the treatment of cancer, as Shp1 inhibition would be predicted to unleash both innate and adaptive immunity against tumor cells. Antibodies blocking the interaction between CD47 on tumor cells and SIRPα on macrophages enhance macrophage phagocytosis, show efficacy in preclinical tumor models, and are being evaluated in the clinic. Here we found that Shp1 bound to phosphorylated peptide sequences derived from SIRPα and transduced the anti-phagocytic signal, as Shp1 loss in mouse bone marrow-derived macrophages increased phagocytosis of tumor cells in vitro. We also generated a novel mouse model to evaluate the impact of global, inducible Ptpn6 deletion on anti-tumor immunity. We found that inducible Shp1 loss drove an inflammatory disease in mice that was phenotypically similar to that seen when Ptpn6 is knocked out from birth. This indicates that acute perturbation of Shp1 in vivo could drive hyperactivation of immune cells, which could be therapeutically beneficial, though at the risk of potential toxicity. In this model, we found that Shp1 loss led to robust anti-tumor immunity against two immune-rich syngeneic tumor models that are moderately inflamed though not responsive to checkpoint inhibitors, MC38 and E0771. Shp1 loss did not promote anti-tumor activity in the non-inflamed B16F10 model. The observed activity in MC38 and E0771 tumors was likely due to effects of both innate and adaptive immune cells. Following Shp1 deletion, we observed increases in intratumoral myeloid cells in both models, which was more striking in E0771 tumors. E0771 tumors also contained an increased ratio of effector to regulatory T cells following Shp1 loss. This was not observed for MC38 tumors, though we did find increased levels of IFNγ, a cytokine produced by effector T cells, in these tumors. Overall, our preclinical data suggested that targeting Shp1 may be an attractive therapeutic strategy for boosting the immune response to cancer via a mechanism involving both innate and adaptive leukocytes.


Asunto(s)
Adenocarcinoma/enzimología , Neoplasias de la Mama/enzimología , Neoplasias del Colon/enzimología , Melanoma Experimental/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Neoplasias Cutáneas/enzimología , Macrófagos Asociados a Tumores/enzimología , Inmunidad Adaptativa , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma/terapia , Animales , Antígenos de Diferenciación/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Femenino , Humanos , Inmunidad Innata , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Receptores Inmunológicos/metabolismo , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Células THP-1 , Carga Tumoral , Microambiente Tumoral , Macrófagos Asociados a Tumores/inmunología
8.
Front Immunol ; 11: 574990, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33123153

RESUMEN

Sphingosine kinase 1 (SPHK1) is a crucial molecule that catalyzes sphingosine to synthesize sphingosine-1-phosphate (S1P), facilitating cell survival signaling. Pyroptosis is a perplexing inflammatory mode of cell death primarily triggered by caspase-1, evoked by the NLRP3 inflammasome. Sphingosine is identified as a danger-associated molecular pattern (DAMP), which activates the NLRP3 inflammasome assembly and induces the pyroptosis. It has been demonstrated that macrophages play a pro-tumorigenic role and are closely associated with tumor progression. Attenuation of SPHK1 activity contributes significantly to macrophage pyroptosis and tumor inhibition. Calcium and integrin-binding protein 1 (CIB1) plays an important role in the translocation of SPHK1 from the cytoplasm to the plasma membrane, whereas CIB2 blocks the subcellular trafficking of SPHK1. Therefore, knockout of CIB1 or over-expression of CIB2 will result in sphingosine accumulation and contribute significantly to cancer treatment by several approaches. First, it directly provokes cancer cell apoptosis or triggers robust anti-tumor immunity by pyroptosis-induced inflammation. Second, it could restrain SPHK1 translocation from the cytoplasm to the plasma membrane and further pyroptosis, which not only drive M2 macrophages death but also facilitate tumor microenvironment inflammation as well as the further release of sphingosine from damaged macrophages. The perspective might provide novel insight into the association between SPHK1 and pyroptosis and suggest the potential target for cancer therapy.


Asunto(s)
Alarminas/metabolismo , Lisofosfolípidos/metabolismo , Neoplasias/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Piroptosis , Esfingosina/análogos & derivados , Animales , Antineoplásicos/uso terapéutico , Proteínas de Unión al Calcio/metabolismo , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Piroptosis/efectos de los fármacos , Transducción de Señal , Esfingosina/metabolismo , Microambiente Tumoral , Macrófagos Asociados a Tumores/enzimología , Macrófagos Asociados a Tumores/inmunología
9.
Front Immunol ; 11: 1759, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973744

RESUMEN

The histological architecture of certain aggressive B-cell lymphomas (prototypically Burkitt's lymphoma, BL) is characterized by a "starry-sky" (SS) appearance. This is caused by tumor-associated macrophages (TAMs), which appear in standard histological preparations as "stars" in a darkly stained "sky" of lymphoma cells. SS-TAMs accumulate in response to constitutive apoptosis in these tumors and are activated by the apoptotic tumor cells to a pro-oncogenic phenotype. The extent to which SS-TAMs contribute to lymphoma growth through responses generated by interactions with apoptotic tumor cells is unknown. Here, we demonstrate a role for the receptor tyrosine kinase, MERTK, in the oncogenic activity of SS-TAMs. We show that MERTK expression is largely restricted to the macrophages of human BL and of murine models of SS B-cell lymphoma and that it is upregulated in SS-TAMs as compared to the germinal center or paracortical macrophages of normal lymph nodes. Our results further demonstrate that MERTK is active in the phagocytosis of apoptotic lymphoma cells by macrophages and, most significantly, that SS lymphoma growth is markedly inhibited in Mertk-/- mice. These results point toward the MERTK apoptotic-cell clearance/response pathway playing a key role in growth of aggressive B-cell lymphoma and identifies MERTK as a novel potential antilymphoma target.


Asunto(s)
Apoptosis , Linfoma de Burkitt/enzimología , Fagocitosis , Macrófagos Asociados a Tumores/enzimología , Tirosina Quinasa c-Mer/metabolismo , Animales , Linfoma de Burkitt/genética , Linfoma de Burkitt/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Células THP-1 , Carga Tumoral , Tirosina Quinasa c-Mer/genética
10.
Int J Mol Sci ; 21(16)2020 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-32823915

RESUMEN

Carbonic anhydrases (CAs) are acid-base regulatory proteins that modulate a variety of physiological functions. Recent findings have shown that CAIX is particularly upregulated in glioblastoma multiforme (GBM) and is associated with a poor patient outcome and survival rate. An analysis of the GSE4290 dataset of patients with gliomas showed that CAIX was highly expressed in GBM and was negatively associated with prognosis. The expression of CAIX under hypoxic conditions in GBM significantly increased in protein, mRNA, and transcriptional activity. Importantly, CAIX upregulation also regulated GBM motility, monocyte adhesion to GBM, and the polarization of tumor-associated monocytes/macrophages (TAM). Furthermore, the overexpression of CAIX was observed in intracranial GBM cells. Additionally, epidermal growth factor receptor/signal transducer and activator of transcription 3 regulated CAIX expression under hypoxic conditions by affecting the stability of hypoxia-inducible factor 1α. In contrast, the knockdown of CAIX dramatically abrogated the change in GBM motility and monocyte adhesion to GBM under hypoxic conditions. Our results provide a comprehensive understanding of the mechanisms of CAIX in the GBM microenvironment. Hence, novel therapeutic targets of GBM progression are possibly developed.


Asunto(s)
Anhidrasa Carbónica IX/metabolismo , Movimiento Celular , Receptores ErbB/metabolismo , Glioblastoma/enzimología , Glioblastoma/patología , Factor de Transcripción STAT3/metabolismo , Hipoxia Tumoral , Macrófagos Asociados a Tumores/patología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Adhesión Celular , Línea Celular Tumoral , Polaridad Celular , Humanos , Concentración de Iones de Hidrógeno , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Monocitos/patología , Microambiente Tumoral , Macrófagos Asociados a Tumores/enzimología
11.
Biomed Pharmacother ; 129: 110440, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32768942

RESUMEN

Jian-pi-yang-zheng Decoction (JPYZ) is a traditional Chinese medicine that is used for the treatment of advanced gastric cancer, and it shows good efficacy in patients. A previous study indicated that JPYZ inhibited the progression of gastric cancer via the regulation of tumor-associated macrophages (TAMs), but the underlying molecular target of JPYZ regulation of TAMs has not been determined. The present study used modified-JPYZ (mJPYZ) to extend our investigation of gastric cancer. Our results showed that mJPYZ inhibited gastric cancer progression in vivo and in vitro. We found that mJPYZ decreased the activity of PI3-kinase γ (PI3Kγ) in TAMs, reduced the anti-inflammatory factor IL-10 and increased the expression of pro-inflammatory cytokines, such as TNF-α and IL-1ß, which ultimately promoted the conversion of TAMs from M2 to M1. Our findings also indicated that mJPYZ inhibited the growth and metastasis of gastric cancer by alleviating the unfavorable differentiation of TAMs via the PI3Kγ signaling cascades. In conclusion, the present findings indicated that mJPYZ inhibited gastric cancer cell EMT via PI3Kγ-dependent TAM reprogramming, which eventually suppressed gastric cancer growth and metastasis. Our study provides an underlying mechanism of a Chinese medicine in the treatment of gastric cancer via PI3Kγ in macrophages.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Medicamentos Herbarios Chinos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Macrófagos Asociados a Tumores/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Citocinas/metabolismo , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Metástasis de la Neoplasia , Fenotipo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Células THP-1 , Microambiente Tumoral , Macrófagos Asociados a Tumores/enzimología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/patología
12.
Theranostics ; 10(7): 3049-3063, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194854

RESUMEN

Simultaneously targeted treatment of tumor cells and their surrounding growth-supporting immune cells is a promising strategy to reshape immunosuppressive tumor microenvironment (TME) and potentiate host innate and adaptive antitumor immune responses. Methods: We designed a series of melittin-(RADA)n hybrid peptide sequences with varying self-assembling motifs of RADA and screened out a melittin-(RADA)6 peptide that has an optimal gel-formation ability and in vitro antitumor activity. Results: The formed melittin-(RADA)6 (MR52) hydrogel scaffold could be loaded with a specific Ca2+/calmodulin-dependent protein kinase II (CAMKII) inhibitor, KN93, originally found to have both direct tumoricidal activity and macrophages-reprogramming ability, for potent immunotherapy against melanoma and hepatoma ascites in mice models. Our MR52 hydrogel has an interweaving nanofiber-like structure, possesses direct antitumor and controlled drug release properties, and promotes the enhanced intracellular uptake of loaded cargo. Compared to free KN93, the MR52-KN93 hydrogel (MRK) improved the killing effects and levels of immunogenic cell death (ICD) on tumor cells significantly. Due to the dual role of KN93, the injection of the MRK hydrogel retarded the growth of subcutaneous melanoma tumors dramatically and resulted in a high number of mature dendritic cells of draining lymph nodes, significantly enhancing the portion of cytotoxic T cells and reduced number of M2-like tumor-associated macrophages (TAMs) in tumors. Using a mouse model of malignant ascites (MAs), where traditional therapy was ineffective, we demonstrated that the MRK hydrogel treatment offered a significantly prolonged survival compared to controls. Following treatment with the MRK hydrogel, macrophages had elevated programmed cell death protein ligand-1 (PD-L1) expression, promising follow-up combined anti-PD-1 therapy that confers a cure rate of approximately 30% against MAs in mice models. Conclusion: Thus, the MRK hydrogel may serve as a prospective platform for antitumor applications.


Asunto(s)
Antineoplásicos/uso terapéutico , Ascitis/terapia , Bencilaminas/uso terapéutico , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Hidrogeles/administración & dosificación , Inmunoterapia/métodos , Neoplasias Hepáticas Experimentales/terapia , Melanoma Experimental/terapia , Meliteno/administración & dosificación , Terapia Molecular Dirigida/métodos , Proteínas de Neoplasias/antagonistas & inhibidores , Oligopéptidos/administración & dosificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Sulfonamidas/uso terapéutico , Macrófagos Asociados a Tumores/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antineoplásicos/administración & dosificación , Ascitis/etiología , Ascitis/inmunología , Antígeno B7-H1/biosíntesis , Bencilaminas/administración & dosificación , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Técnicas de Reprogramación Celular , Composición de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Inyecciones Intraperitoneales , Neoplasias Hepáticas Experimentales/complicaciones , Neoplasias Hepáticas Experimentales/inmunología , Activación de Macrófagos , Masculino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas de Neoplasias/fisiología , Inhibidores de Proteínas Quinasas/administración & dosificación , Distribución Aleatoria , Proteínas Recombinantes de Fusión/administración & dosificación , Sulfonamidas/administración & dosificación , Escape del Tumor/efectos de los fármacos , Macrófagos Asociados a Tumores/clasificación , Macrófagos Asociados a Tumores/enzimología
13.
Front Immunol ; 11: 607891, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33708191

RESUMEN

Chronic inflammation increases the risk for colorectal cancer through a variety of mechanisms involving the tumor microenvironment. MAPK-activated protein kinase 2 (MK2), a major effector of the p38 MAPK stress and DNA damage response signaling pathway, and a critical regulator of pro-inflammatory cytokine production, has been identified as a key contributor to colon tumorigenesis under conditions of chronic inflammation. We have previously described how genetic inactivation of MK2 in an inflammatory model of colon cancer results in delayed tumor progression, decreased tumor angiogenesis, and impaired macrophage differentiation into a pro-tumorigenic M2-like state. The molecular mechanism responsible for the impaired angiogenesis and tumor progression, however, has remained contentious and poorly defined. Here, using RNA expression analysis, assays of angiogenesis factors, genetic models, in vivo macrophage depletion and reconstitution of macrophage MK2 function using adoptive cell transfer, we demonstrate that MK2 activity in macrophages is necessary and sufficient for tumor angiogenesis during inflammation-induced cancer progression. We identify a critical and previously unappreciated role for MK2-dependent regulation of the well-known pro-angiogenesis factor CXCL-12/SDF-1 secreted by tumor associated-macrophages, in addition to MK2-dependent regulation of Serpin-E1/PAI-1 by several cell types within the tumor microenvironment.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Neoplasias Asociadas a Colitis/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neovascularización Patológica , Proteínas Serina-Treonina Quinasas/metabolismo , Macrófagos Asociados a Tumores/enzimología , Traslado Adoptivo , Proteínas Angiogénicas/genética , Animales , Células Cultivadas , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Transcripción Genética , Microambiente Tumoral , Macrófagos Asociados a Tumores/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...