Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Aging (Albany NY) ; 14(3): 1068-1086, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35158337

RESUMEN

Radiation therapy is a commonly used treatment modality for cancer. Although effective in providing local tumor control, radiation causes oxidative stress, inflammation, immunomodulatory and mitogenic cytokine production, extracellular matrix production, and premature senescence in lung parenchyma. The senescence associated secretory phenotype (SASP) can promote inflammation and stimulate alterations in the surrounding tissue. Therefore, we hypothesized that radiation-induced senescent parenchymal cells in irradiated lung would enhance tumor growth. Using a murine syngeneic tumor model of melanoma and non-small cell lung cancer lung metastasis, we demonstrate that radiation causes a significant increase in markers of premature senescence in lung parenchyma within 4 to 8 weeks. Further, injection of B16F0 (melanoma) or Lewis Lung carcinoma (epidermoid lung cancer) cells at these time points after radiation results in an increase in the number and size of pulmonary tumor nodules relative to unirradiated mice. Treatment of irradiated mice with a senolytic agent (ABT-737) or agents that prevent senescence (rapamycin, INK-128) was sufficient to reduce radiation-induced lung parenchymal senescence and to mitigate radiation-enhanced tumor growth. These agents abrogated radiation-induced expression of 12-Lipoxygenase (12-LOX), a molecule implicated in several deleterious effects of senescence. Deficiency of 12-LOX prevented radiation-enhanced tumor growth. Together, these data demonstrate the pro-tumorigenic role of radiation-induced senescence, introduces the dual TORC inhibitor INK-128 as an effective agent for prevention of radiation-induced normal tissue senescence, and identifies senescence-associated 12-LOX activity as an important component of the pro-tumorigenic irradiated tissue microenvironment. These studies suggest that combining senotherapeutic agents with radiotherapy may decrease post-therapy tumor growth.


Asunto(s)
Carcinoma Pulmonar de Lewis , Neoplasias Pulmonares , Melanoma Experimental , Animales , Araquidonato 12-Lipooxigenasa/farmacología , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/patología , Procesos de Crecimiento Celular , Senescencia Celular , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Melanoma Experimental/enzimología , Melanoma Experimental/patología , Ratones , Microambiente Tumoral
2.
J Immunol ; 207(6): 1662-1671, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34417261

RESUMEN

The 1858C>T allele of the tyrosine phosphatase PTPN22 (causing amino acid substitution R620W in encoded protein lymphoid tyrosine phosphatase) is present in 5-10% of the North American population and is strongly associated with numerous autoimmune diseases. Although much research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its proautoimmune allele have in tumor immunity is poorly defined. To interrogate the role this allele may have in the antitumor immune response, we used CRISPR/Cas9 to generate mice in which the ortholog of lymphoid tyrosine phosphatase, PEST domain-enriched protein (PEP), is mutated at position 619 to produce the relevant proautoimmune mutation (R619W). Results of this study show that mice homozygous for this alteration (PEP-619WW) resist tumor growth as compared with wild-type mice. Consistent with these results, tumors from PEP-619WW mice have more CD45 infiltrates containing more activated CD8 T cells and CD4 T cells. In addition, there are more conventional dendritic cell type 1 (cDC1) cells and fewer myeloid-derived suppressor cells in tumors from PEP-619WW mice. Interestingly, the tumor-infiltrating PEP-619WW cDC1 cells have decreased PD-L1 expression compared with cDC1 cells from PEP-wild-type mice. Taken together, our data show that the proautoimmune allele of Ptpn22 drives a strong antitumor response in innate and adaptive immune cells resulting in superior control of tumors.


Asunto(s)
Alelos , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/inmunología , Inmunidad , Melanoma Experimental/enzimología , Melanoma Experimental/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/inmunología , Animales , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Femenino , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias/métodos , Fenotipo , Neoplasias Cutáneas/patología
3.
Neoplasia ; 23(9): 966-978, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34343822

RESUMEN

Heparanase is highly implicated in tumor metastasis due to its capacity to cleave heparan sulfate and, consequently, remodel the extracellular matrix underlying epithelial and endothelial cells. In striking contrast, only little attention was given to its close homolog, heparanase 2 (Hpa2), possibly because it lacks heparan sulfate-degrading activity typical of heparanase. We subjected sections of gastric carcinoma to immunostaining and correlated Hpa2 immunoreactivity with clinical records, including tumor grade, stage and patients' status. We over-expressed Hpa2 in gastric carcinoma cell lines and examined their tumorigenic properties in vitro and in vivo. We also evaluated the expression of Hpa2 by gastric carcinoma cells following inhibition of the proteasome, leading to proteotoxic stress, and the resulting signaling responsible for Hpa2 gene regulation. Here, we report that gastric cancer patients exhibiting high levels of Hpa2 survive longer. Similarly, mice administrated with gastric carcinoma cells engineered to over-express Hpa2 produced smaller tumors and survived longer than mice administrated with control cells. This was associated with increased phosphorylation of AMP-activated protein kinase (AMPK), a kinase that is situated at the center of a tumor suppressor network. We also found that MG132, an inhibitor of the proteasome that results in proteotoxic stress, prominently enhances Hpa2 expression. Notably, Hpa2 induction by MG132 appeared to be mediated by AMPK, and AMPK was found to induce the expression of Hpa2, thus establishing a loop that feeds itself where Hpa2 enhances AMPK phosphorylation that, in turn, induces Hpa2 expression, leading to attenuation of gastric tumorigenesis. These results indicate that high levels of Hpa2 in some tumors are due to stress conditions that tumors often experience due to their high rates of cell proliferation and high metabolic demands. This increase in Hpa2 levels by the stressed tumors appears critically important for patient outcomes.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Glucuronidasa/biosíntesis , Neoplasias Gástricas/enzimología , Anciano , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Bortezomib/farmacología , Bortezomib/uso terapéutico , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Glucuronidasa/genética , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología
4.
Cell Death Dis ; 12(7): 680, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226519

RESUMEN

It has been recently reported that CD38 expressed on tumor cells of multiple murine and human origins could be upregulated in response to PD-L1 antibody therapy, which led to dysfunction of tumor-infiltrating CD8+ T immune cells due to increasing the production of adenosine. However, the role of tumor expressed-CD38 on neoplastic formation and progression remains elusive. In the present study, we aimed to delineate the molecular and biochemical function of the tumor-associated CD38 in lung adenocarcinoma progression. Our clinical data showed that the upregulation of tumor-originated CD38 was correlated with poor survival of lung cancer patients. Using multiple in vitro assays we found that the enzymatic activity of tumor expressed-CD38 facilitated lung cancer cell migration, proliferation, colony formation, and tumor development. Consistently, our in vivo results showed that inhibition of the enzymatic activity or antagonizing the enzymatic product of CD38 resulted in the similar inhibition of tumor proliferation and metastasis as CD38 gene knock-out or mutation. At biochemical level, we further identified that cADPR, the mainly hydrolytic product of CD38, was responsible for inducing the opening of TRPM2 iron channel leading to the influx of intracellular Ca2+ and then led to increasing levels of NRF2 while decreasing expression of KEAP1 in lung cancer cells. These findings suggested that malignant lung cancer cells were capable of using cADPR catalyzed by CD38 to facilitate tumor progression, and blocking the enzymatic activity of CD38 could be represented as an important strategy for preventing tumor progression.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Adenocarcinoma del Pulmón/enzimología , ADP-Ribosa Cíclica/metabolismo , Neoplasias Pulmonares/enzimología , Glicoproteínas de Membrana/metabolismo , Células A549 , ADP-Ribosil Ciclasa 1/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/secundario , Animales , Señalización del Calcio , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Movimiento Celular , Proliferación Celular , Bases de Datos Genéticas , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Glicoproteínas de Membrana/genética , Ratones Endogámicos C57BL , Persona de Mediana Edad , Factor 2 Relacionado con NF-E2/metabolismo , Invasividad Neoplásica , Canales Catiónicos TRPM/metabolismo
5.
Angiogenesis ; 24(3): 471-482, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33730293

RESUMEN

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is overexpressed in many cancer types and in vivo studies have shown that vascular endothelial cell FAK expression and FAK-phosphorylation at tyrosine (Y) 397, and subsequently FAK-Y861, are important in tumour angiogenesis. Pericytes also play a vital role in regulating tumour blood vessel stabilisation, but the specific involvement of pericyte FAK-Y397 and FAK-Y861 phosphorylation in tumour blood vessels is unknown. Using PdgfrßCre + ;FAKWT/WT, PdgfrßCre + ;FAKY397F/Y397F and PdgfrßCre + ;FAKY861F/Y861F mice, our data demonstrate that Lewis lung carcinoma tumour growth, tumour blood vessel density, blood vessel perfusion and pericyte coverage were affected only in late stage tumours in PdgfrßCre + ;FAKY861F/Y861F but not PdgfrßCre + ;FAKY397F/Y397F mice. Further examination indicates a dual role for pericyte FAK-Y861 phosphorylation in the regulation of tumour vessel regression and also in the control of pericyte derived signals that influence apoptosis in cancer cells. Overall this study identifies the role of pericyte FAK-Y861 in the regulation of tumour vessel regression and tumour growth control and that non-phosphorylatable FAK-Y861F in pericytes reduces tumour growth and blood vessel density.


Asunto(s)
Apoptosis , Carcinoma Pulmonar de Lewis , Quinasa 1 de Adhesión Focal , Mutación Missense , Proteínas de Neoplasias , Neovascularización Patológica , Pericitos/enzimología , Sustitución de Aminoácidos , Animales , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/genética , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética , Fosforilación
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.
Biosci Rep ; 40(6)2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32530032

RESUMEN

A homogeneous polysaccharide (GLP), with an average molecular weight of 4.44 × 104 Da, was isolated and purified from the fruiting bodies of Ganoderma lucidum. In this work, we examined the antitumor activities of GLP using a mouse Lewis lung cancer (LLC) model and explored possible molecular pathways involved in its immunomodulatory mechanism on tumor-host interaction. GLP administration (25 and 100 mg/kg) significantly inhibited tumor growth, as evidenced by the decreased tumor volume and tumor weight, as well as histological features of tumor tissues with concomitant down-regulation of proliferating cell nuclear antigen (PCNA) proliferative marker. Less myeloid-derived suppressor cells (MDSCs) were accumulated in both spleen and tumor tissues from GLP-treated mice. In contrast, the percentage of CD4+ and CD8+ T cells together with the production of Th1-type cytokines (IFN-γ and IL-12) was increased in the spleen of LLC-bearing mice following GLP administration. Furthermore, GLP administration reversed the attenuated expression of CARD9, p-Syk and p-p65, and increased indoleamine 2,3-dioxygenase (IDO) protein expression in MDSCs of LLC-bearing mice. Collectively, our data demonstrated the first time that GLP induced the differentiation of MDSCs and inhibited the accumulation of MDSCs via CARD9-NF-κB-IDO pathway, thus prevented lung cancer development.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Polisacáridos Fúngicos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Células Supresoras de Origen Mieloide/efectos de los fármacos , Reishi , Animales , Antineoplásicos/aislamiento & purificación , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Cuerpos Fructíferos de los Hongos , Polisacáridos Fúngicos/aislamiento & purificación , Masculino , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/enzimología , Células Supresoras de Origen Mieloide/inmunología , FN-kappa B/metabolismo , Reishi/química , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral
8.
Circ Res ; 127(6): 707-723, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32527198

RESUMEN

RATIONALE: How endothelial cells (ECs) migrate and form an immature vascular plexus has been extensively studied. Yet, mechanisms underlying vascular remodeling remain poorly established. A better understanding of these processes may lead to the design of novel therapeutic strategies complementary to current angiogenesis inhibitors. OBJECTIVE: Starting from our previous observations that PP2A (protein phosphatase 2) regulates the HIF (hypoxia-inducible factor)/PHD-2 (prolyl hydroxylase 2)-constituted oxygen machinery, we hypothesized that this axis could play an important role during blood vessel formation, tissue perfusion, and oxygen restoration. METHODS AND RESULTS: We show that the PP2A regulatory subunit B55α is at the crossroad between vessel pruning and vessel maturation. Blood vessels with high B55α counter cell stress conditions and thrive for stabilization and maturation. When B55α is inhibited, ECs cannot cope with cell stress and undergo apoptosis, leading to massive pruning of nascent blood vessels. Mechanistically, we found that the B55α/PP2A complex restrains PHD-2 activity, promoting EC survival in a HIF-dependent manner, and furthermore dephosphorylates p38, altogether protecting ECs against cell stress occurring, for example, during the onset of blood flow. In tumors, EC-specific B55α deficiency induces pruning of immature-like tumor blood vessels resulting in delayed tumor growth and metastasis, without affecting nonpathological vessels. Consistently, systemic administration of a pan-PP2A inhibitor disrupts vascular network formation and tumor progression in vivo without additional effects on B55α-deficient vessels. CONCLUSIONS: Our data underline a unique role of the B55α/PP2A phosphatase complex in vessel remodeling and suggest the use of PP2A-inhibitors as potent antiangiogenic drugs targeting specifically nascent blood vessels with a mode-of-action complementary to VEGF-R (vascular endothelial growth factor receptor)-targeted therapies. Graphical Abstract: A graphical abstract is available for this article.


Asunto(s)
Apoptosis , Neoplasias de la Mama/enzimología , Carcinoma Pulmonar de Lewis/enzimología , Células Endoteliales/enzimología , Neovascularización Patológica , Proteína Fosfatasa 2/metabolismo , Remodelación Vascular , Inhibidores de la Angiogénesis/farmacología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/genética , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Cancer Lett ; 483: 98-113, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32217106

RESUMEN

Liver metastases remain a major cause of death from gastrointestinal tract cancers and other malignancies, such as breast and lung carcinomas. Understanding the underlying biology is essential for the design of effective therapies. We previously identified the chemokine CCL7 and its receptor CCR3 as critical mediators of invasion and metastasis in lung and colon carcinoma cells. Here we show that the CCL7/CCR3 axis regulates a late stage in invadopodia genesis namely, the targeting of MMP-9 to the invadopodia complex, thereby promoting invadopodia maturation and collagen degradation. We show that this process could be blocked by overexpression of a dominant negative RhoA in highly invasive cells, while a constitutively active RhoA upregulated invadopodia maturation in CCL7-silenced and poorly invasive and metastatic cells and also enhanced their metastatic potential in vivo, collectively, implicating RhoA activation in signaling downstream of CCL7. Blockade of the ERK or PI3K pathways by chemical inhibitors also inhibited invadopodia formation, but affected the initiation stage of invadopodia genesis. Our data implicate CCL7/CCR3 signaling in invadopodia maturation and suggest that chemokine signaling acts in concert with extracellular matrix-initiated signals to promote invasion and liver metastasis.


Asunto(s)
Carcinoma Pulmonar de Lewis/enzimología , Movimiento Celular , Quimiocina CCL7/metabolismo , Colágeno/metabolismo , Neoplasias del Colon/enzimología , Neoplasias Hepáticas/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Podosomas/enzimología , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Quimiocina CCL7/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Metaloproteinasa 9 de la Matriz/genética , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Podosomas/genética , Podosomas/patología , Transporte de Proteínas , Proteolisis , Receptores CCR3/genética , Receptores CCR3/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
10.
Mol Cancer Ther ; 19(3): 755-764, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31974273

RESUMEN

Macrophages (MΦ) play a critical role in tumor growth, immunosuppression, and inhibition of adaptive immune responses in cancer. Hence, targeting signaling pathways in MΦs that promote tumor immunosuppression will provide therapeutic benefit. PI3Kγ has been recently established by our group and others as a novel immuno-oncology target. Herein, we report that an MΦ Syk-PI3K axis drives polarization of immunosuppressive MΦs that establish an immunosuppressive tumor microenvironment in in vivo syngeneic tumor models. Genetic or pharmacologic inhibition of Syk and/or PI3Kγ in MΦs promotes a proinflammatory MΦ phenotype, restores CD8+ T-cell activity, destabilizes HIF under hypoxia, and stimulates an antitumor immune response. Assay for transposase-accessible Chromatin using Sequencing (ATAC-seq) analyses on the bone marrow-derived macrophages (BMDM) show that inhibition of Syk kinase promotes activation and binding of NF-κB motif in SykMC-KO BMDMs, thus stimulating immunostimulatory transcriptional programming in MΦs to suppress tumor growth. Finally, we have developed in silico the "first-in-class" dual Syk/PI3K inhibitor, SRX3207, for the combinatorial inhibition of Syk and PI3K in one small molecule. This chemotype demonstrates efficacy in multiple tumor models and represents a novel combinatorial approach to activate antitumor immunity.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Fosfatidilinositol 3-Quinasa Clase Ib/química , Neoplasias del Colon/inmunología , Macrófagos/inmunología , Melanoma Experimental/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Quinasa Syk/antagonistas & inhibidores , Animales , Apoptosis , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/patología , Proliferación Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Citocinas/metabolismo , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/enzimología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Mol Sci ; 21(1)2019 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-31878088

RESUMEN

Tyrosyl-DNA phosphodiesterase 1 (Tdp1) is an important DNA repair enzyme in humans, and a current and promising inhibition target for the development of new chemosensitizing agents due to its ability to remove DNA damage caused by topoisomerase 1 (Top1) poisons such as topotecan and irinotecan. Herein, we report our work on the synthesis and characterization of new Tdp1 inhibitors that combine the arylcoumarin (neoflavonoid) and monoterpenoid moieties. Our results showed that they are potent Tdp1 inhibitors with IC50 values in the submicromolar range. In vivo experiments with mice revealed that compound 3ba (IC50 0.62 µM) induced a significant increase in the antitumor effect of topotecan on the Krebs-2 ascites tumor model. Our results further strengthen the argument that Tdp1 is a druggable target with the potential to be developed into a clinically-potent adjunct therapy in conjunction with Top1 poisons.


Asunto(s)
Carcinoma Krebs 2/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Monoterpenos , Proteínas de Neoplasias , Inhibidores de Fosfodiesterasa , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Carcinoma Krebs 2/enzimología , Carcinoma Krebs 2/patología , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/patología , Femenino , Humanos , Células MCF-7 , Masculino , Ratones , Monoterpenos/síntesis química , Monoterpenos/química , Monoterpenos/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Inhibidores de Fosfodiesterasa/síntesis química , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Relación Estructura-Actividad
12.
J Cell Biochem ; 120(10): 16978-16989, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31111562

RESUMEN

The effects of ethoxy-erianin phosphate (EBTP) on cell proliferation, mitotic cell arrest, migration, infiltration, and endothelial tubular structures were evaluated in this study. The antiproliferative activity of EBTP and combretastatin A-4P (CA4P) was analyzed on several tumor cells (including MCF-7, HeLa, 2LL, and 2LL-IDO) and on an endothelial cell (human umbilical vein endothelial cells [HUVECs]) as well as a human normal liver cell (L02). The results showed that EBTP possessed antiproliferative activity in the micromole range and was relatively less toxic than CA4P. Treating HUVECs with EBTP caused cell accumulation in the G2/M phase, and wound-healing assays indicated that EBTP inhibited cell migration. Furthermore, EBTP and CA4P destroyed the vasculature in endothelial cells and showed vascular disrupting activity of the chorioallantoic membrane in fertilized chicken eggs. In addition, we found that EBTP suppressed the expression of indoleamine 2,3-dioxygenase (IDO) and significantly inhibited IDO-induced migration and infiltration of 2LL-IDO cells. Administration of EBTP blocked vasculogenic mimicry in 2LL-IDO cells, which was typically observed in tube formation assays of 2LL-IDO cells. Moreover, the results of Lewis lung carcinoma in mice showed a high inhibition rate of EBTP. EBTP is an effective vascular disrupting agent that is superior to CA4P and may prevent and treat malignancy by inhibiting the expression of IDO.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/farmacología , Bibencilos/farmacología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Neovascularización Patológica/tratamiento farmacológico , Animales , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Células HeLa , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Estilbenos/farmacología , Carga Tumoral/efectos de los fármacos
13.
Proc Natl Acad Sci U S A ; 116(14): 6836-6841, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30890638

RESUMEN

Lysyl oxidase (LOX) and LOX-like (LOXL) proteins are copper-dependent metalloenzymes with well-documented roles in tumor metastasis and fibrotic diseases. The mechanism by which copper is delivered to these enzymes is poorly understood. In this study, we demonstrate that the copper transporter ATP7A is necessary for the activity of LOX and LOXL enzymes. Silencing of ATP7A inhibited LOX activity in the 4T1 mammary carcinoma cell line, resulting in a loss of LOX-dependent mechanisms of metastasis, including the phosphorylation of focal adhesion kinase and myeloid cell recruitment to the lungs, in an orthotopic mouse model of breast cancer. ATP7A silencing was also found to attenuate LOX activity and metastasis of Lewis lung carcinoma cells in mice. Meta-analysis of breast cancer patients found that high ATP7A expression was significantly correlated with reduced survival. Taken together, these results identify ATP7A as a therapeutic target for blocking LOX- and LOXL-dependent malignancies.


Asunto(s)
Carcinoma Pulmonar de Lewis/enzimología , ATPasas Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Neoplasias Mamarias Animales/enzimología , Proteínas de Neoplasias/metabolismo , Proteína-Lisina 6-Oxidasa/metabolismo , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , ATPasas Transportadoras de Cobre/genética , Femenino , Humanos , Transporte Iónico , Masculino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Metaanálisis como Asunto , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteína-Lisina 6-Oxidasa/genética
14.
J Integr Med ; 16(4): 283-289, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29752140

RESUMEN

OBJECTIVE: Escape from the body's immune response is a basic characteristic of lung cancer, and indoleamine-2,3-dioxygenase (IDO) plays a key role in mediating immune escape of non-small-cell lung cancer, which leads to recurrence and metastasis. Feiji Recipe, a compound Chinese herbal medicine, has the effect of stabilizing lesions and prolonging survival in patients with lung cancer. The purpose of this study was to investigate the mechanisms underlying the anticancer properties of Feiji Recipe. METHODS: An orthotopic transplant model of mouse Lewis lung cancer, with stable expression of IDO gene, was established in C57BL/6 mice. Optical imaging was used to observe the effects of Feiji Recipe in the treatment of lung cancer in vivo. The effects of Feiji Recipe on the proliferation of mouse Lewis lung cancer cell line 2LL, 2LL-enhanced green fluorescent protein (2LL-EGFP) and 2LL-EGFP-IDO were investigated, and the apoptosis of T-cells was examined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide using flow cytometry. Chemical composition of Feiji Recipe was validated by high-performance liquid chromatography. RESULTS: Compared to the control group, the survival of animals treated with Feiji Recipe was significantly prolonged (P = 0.0074), and the IDO protein level decreased (P = 0.0072); moreover, the percentages of CD4+CD25+ T-cells and Foxp3+ T-cells were significantly decreased (P < 0.05). The molecular mechanism of Feiji Recipe against lung cancer may relate to the regulation of immune cells, such as T-cells and regulatory T-cells. CONCLUSION: The molecular mechanism of Feiji Recipe in treatment of lung cancer is to restore the function of T-cells in the cancer microenvironment through interfering with the IDO pathway.


Asunto(s)
Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/administración & dosificación , Inhibidores de Crecimiento/administración & dosificación , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/fisiopatología , Modelos Animales de Enfermedad , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
15.
J Thorac Cardiovasc Surg ; 155(5): 2164-2175.e1, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29429629

RESUMEN

OBJECTIVE: Past studies are inconsistent with regard to the role of matrix metalloproteinase 12 in lung tumorigenesis. This is due, in part, to differential tumorigenesis based on tumor-derived versus immune-derived matrix metalloproteinase 12 expression. Our study aims to thoroughly dissect the role of matrix metalloproteinase 12 in lung tumorigenesis. METHODS: We tested matrix metalloproteinase 12 expression and the association with prognosis using a tissue array and a published non-small cell lung cancer gene expression database. In addition, we characterized the contribution of matrix metalloproteinase 12 to tumor propagation in the lung using a series of in vitro and in vivo studies. RESULTS: Tumor cells of a diverse set of human lung cancers stained positive for matrix metalloproteinase 12, and high matrix metalloproteinase 12 mRNA levels in the tumor were associated with reduced survival. The lung microenvironment stimulated endogenous production of matrix metalloproteinase 12 in lung cancer cells (human 460 lung cancer cell line, Lewis lung carcinoma). In vitro, matrix metalloproteinase 12 knockout Lewis lung carcinoma and Lewis lung carcinoma cells had the same proliferation rate, but Lewis lung carcinoma showed increased invasiveness. In vivo, deficiency of matrix metalloproteinase 12 in Lewis lung carcinoma cells, but not in the host, reduced tumor growth and invasiveness. CONCLUSIONS: We suggest that tumor cell-derived matrix metalloproteinase 12 promotes tumor propagation in the lung and that in the context of pulmonary malignancies matrix metalloproteinase 12 should further be tested as a potential novel therapeutic target.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Movimiento Celular , Neoplasias Pulmonares/enzimología , Metaloproteinasa 12 de la Matriz/metabolismo , Animales , Biomarcadores de Tumor/genética , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Metaloproteinasa 12 de la Matriz/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Transducción de Señal
16.
Biochem Biophys Res Commun ; 493(4): 1478-1484, 2017 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-28986261

RESUMEN

As an extracellular matrix protein, osteopontin (OPN) has been shown to play an important role in regulation of the immune response to tumors. Myeloid-derived suppressor cells (MDSCs), a heterogeneous population of myeloid progenitors, are major components of the immune suppressive tumor microenvironment and contribute to tumor evasion of the immune response. However, the specific regulating mechanisms underlying MDSCs expansion remain unclear. Here, we found that MDSCs accumulated in the spleen and tumors of 3LL tumor-bearing mice. Supernatant collected from 3LL cells was able to induce the expansion of MDSCs in peripheral blood mononuclear cell (PBMC) in vitro. Results of enzyme linked immunosorbent assay showed high levels of OPN and matrix metalloproteinase-9 (MMP-9) in this supernatant. Silencing OPN can effectively reduce MDSCs frequency in vivo and in vitro. Furthermore, a specific fragment of OPN, OPN-32 kDa cleaved by MMP-9 was detected in the supernatant from 3LL cells. Overexpression of OPN-32 kDa in 3LL cells induced MDSCs expansion. Inhibition of MMP-9 by monoclonal antibody and inhibitor (TIMP-1) reduced MDSCs expansion in vitro and in vivo. These findings suggest that the MMP-9-cleaved OPN fragment, OPN-32kDa, was responsible for MDSCs expansion, which may contribute to tumor's evasion of the immune response.


Asunto(s)
Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Osteopontina/inmunología , Osteopontina/metabolismo , Escape del Tumor/inmunología , Escape del Tumor/fisiología , Animales , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Supresoras de Origen Mieloide/patología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/fisiología
17.
Biomed Pharmacother ; 88: 521-528, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28129624

RESUMEN

Tumor angiogenesis is the key process in tumor growth and metastasis, and transfers essential nutrients for solid tumor. Inhibition of tumor angiogenesis has been recognized as a more effective anti-cancer strategy for NSCLC and has acquired certain therapeutic effects. IDO has non-immune functions including regulating tumor angiogenesis and IDO dysregulation in cancer pathogenesis has been valued. Erianin is a natural product isolated from Dendrobium chrysotoxum Lindl. The antitumor activity of erianin in many kinds of cancers had been demonstrated in previous studies. In this study, we demonstrated that IDO could promote the attachment of 2LL cells, the ability of migration, invasion and VM formation, as well as the tubules forming ability of HUVECs. We also find that erianin suppressed expression and enzyme ability of IDO and erianin could inhibit IDO-induced metastasis and invasion ability of 2LL cells significantly. Erianin not only blocked IDO-induced tube formation of HUVECs, but also suppressed VM formation of 2LL-IDO cells. What's more, we examined that Erianin might play its role in angiogenesis through down-regulating phosphorylation of JAK2/STAT3, inhibiting its downstream target genes MMP-2/-9 and some inflammatory mediators (COX-2, HIF-1α and IL-6), which were all induced by IDO. All these results indicated that erianin had anti-angiogenesis ability and could inhibit the expresison of IDO to prevent and treat the malignant tumors.


Asunto(s)
Bibencilos/farmacología , Bibencilos/uso terapéutico , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Animales , Calibración , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación/patología , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neovascularización Patológica/patología , Fenol , Reproducibilidad de los Resultados , Microambiente Tumoral/efectos de los fármacos
18.
Proc Natl Acad Sci U S A ; 113(48): E7808-E7817, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27849593

RESUMEN

The emerging role of heparanase in tumor initiation, growth, metastasis, and chemoresistance is well recognized and is encouraging the development of heparanase inhibitors as anticancer drugs. Unlike the function of heparanase in cancer cells, very little attention has been given to heparanase contributed by cells composing the tumor microenvironment. Here we used a genetic approach and examined the behavior and function of macrophages isolated from wild-type (WT) and heparanase-knockout (Hpa-KO) mice. Hpa-KO macrophages express lower levels of cytokines (e.g., TNFα, IL1-ß) and exhibit lower motility and phagocytic capacities. Intriguingly, inoculation of control monocytes together with Lewis lung carcinoma (LLC) cells into Hpa-KO mice resulted in nearly complete inhibition of tumor growth. In striking contrast, inoculating LLC cells together with monocytes isolated from Hpa-KO mice did not affect tumor growth, indicating that heparanase is critically required for activation and function of macrophages. Mechanistically, we describe a linear cascade by which heparanase activates Erk, p38, and JNK signaling in macrophages, leading to increased c-Fos levels and induction of cytokine expression in a manner that apparently does not require heparanase enzymatic activity. These results identify heparanase as a key mediator of macrophage activation and function in tumorigenesis and cross-talk with the tumor microenvironment.


Asunto(s)
Carcinoma Pulmonar de Lewis/enzimología , Glucuronidasa/fisiología , Activación de Macrófagos , Macrófagos/enzimología , Animales , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Línea Celular , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Transducción de Señal , Factores de Transcripción/metabolismo , Activación Transcripcional , Carga Tumoral , Microambiente Tumoral
19.
Oncol Rep ; 36(6): 3397-3404, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27779695

RESUMEN

Stereotactic radiosurgery has been recognized as an effective treatment approach for metastatic brain tumors. By increasing the sensitivity of the tumor to radiation and decreasing the marginal dose, it is possible to improve therapeutic efficacy and decrease side-effects. In radiation-induced cells, c-Jun N-terminal kinase (JNK) signaling mediates the phosphorylation of H2AX, which indicates DNA damage sensitivity and modulates the effect of radiation. Lewis lung cancer (LLC) and breast cancer (4T1) cells were irradiated with a Gamma Knife in cell culture tubes. To evaluate the relationship between radiosensitivity and JNK activity, clonogenic assay was performed. DNA damage response was estimated by γH2AX focus formation assay and apoptosis­related protein levels were assessed by western blotting. The mice were subcutaneously inoculated with LLC cells, and irradiated concomitantly with JNK inhibitor treatment. The effect of the JNK inhibitor was investigated by tumor volumetry and immunohistochemistry. γH2AX expression, which mediates repair of radiation­induced DNA damage, was reduced in the cancer cell group pretreated with the JNK inhibitor. This finding shows that JNK inhibition may increase the radiosensitivity in radiated lung and breast cancer cells. For the in vivo study, irradiated tumor growth was significantly delayed in the JNK inhibitor-treated mouse group. Blockade of JNK signaling decreased γH2AX expression and increased apoptosis in the radiation-induced cancer cells. JNK inhibitor may be useful for enhancing the radiosensitivity of lung and breast cancer cells and improving the treatment efficacy of radiosurgical approaches for metastatic brain tumors.


Asunto(s)
Antracenos/farmacología , Antineoplásicos/farmacología , Carcinoma Pulmonar de Lewis/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Carcinoma Pulmonar de Lewis/terapia , Línea Celular Tumoral , Quimioradioterapia , Relación Dosis-Respuesta en la Radiación , Femenino , Histonas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Fosforilación , Procesamiento Proteico-Postraduccional , Tolerancia a Radiación , Transducción de Señal
20.
Bull Exp Biol Med ; 161(3): 367-70, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27502536

RESUMEN

Mice with Lewis lung carcinoma were used to study the antitumor and antimetastatic effects of JAK3 inhibitor. The study revealed no effect of JAK3 inhibitor on the growth of primary tumor node, but found a pronounced inhibition of hematogenous spread of the pathologic process into the lungs. In vitro blockade of JAK3 in cultured Lewis lung carcinoma produced no effect on the count of the stem tumor cells and stimulated functions of committed elements. In addition, blockade of JAK3 significantly elevated maturation index of the tumor tissue.


Asunto(s)
Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/enzimología , Inhibidores Enzimáticos/uso terapéutico , Janus Quinasa 3/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/farmacología , Femenino , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA