Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
J Nutr Biochem ; 107: 109051, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35609853

RESUMEN

Metastasis is a devastating aspect of cancer. This study tested the hypothesis that metabolome of metastases differs from that of host organs by using the spontaneous metastasis model of Lewis lung carcinoma (LLC). In a 2 × 2 design, male C57BL/6 mice with or without a subcutaneous LLC inoculation were fed the standard AIN93G diet or a high-fat diet (HFD) for 12 weeks. Lung metastases from injected mice and the lungs from non-injected mice were harvested at the end of study for untargeted metabolomics of primary metabolism by using gas chromatography time-of-flight mass spectrometry. We identified 91 metabolites for metabolomic analysis. The analysis demonstrated that amino acid and energy metabolism were altered the most in LLC metastases compared to the lungs. A 60% decrease in glutamine and a 25-fold elevation in sorbitol were observed in metastases. Cholesterol and its metabolite dihydrocholesterol were 50% lower in metastases than in the lungs. The HFD elevated arachidonic acid and its precursor linoleic acid in the lungs from noncancer-bearing mice, reflecting the dietary fatty acid composition of the HFD. This elevation did not occur in metastases from HFD-fed LLC-bearing mice, suggesting alterations in lipid metabolism during LLC metastatic progression. Understanding the differences in metabolome between pulmonary LLC metastases and the normal healthy lungs can be useful in designing targeted studies for prevention and treatment of cancer spread using this LLC spontaneous metastasis model.


Asunto(s)
Carcinoma Pulmonar de Lewis , Neoplasias Pulmonares , Animales , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/secundario , Dieta Alta en Grasa/efectos adversos , Pulmón/metabolismo , Neoplasias Pulmonares/patología , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL
2.
Neurosci Lett ; 754: 135851, 2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-33781910

RESUMEN

Psychological stress is a common etiology among patients with lung cancer and serves as a potential indication of poor prognosis and advanced cancer clinical stage. Evidence indicates that depression is positively correlated with the evolvement of lung carcinoma. Nevertheless, the mechanisms underlying the effects of mental disorder on lung cancer have not been considerably and systemically explored. We hypothesized that mental disorder may affect the adjustment of the tumor microenvironment and immune cells. We used the chronic unpredictable mild stress (CUMS) procedure to induce depressed mice models and established tumor-bearing models of C57BL/6 J mice. Results revealed that the worsening of lung cancer was notably hastened in the CUMS + tumor group. Notably, the expression of PD-L1 in tumor issues increased in the tumor microenvironment, accompanied with a decline in the levels of CD8. On the basis of the date of tumor migration, our results indicated that MMPs and VEGF significantly increased after CUMS + tumor treatment. Thus, we demonstrated that modulation of the tumor microenvironment is pivotal for depression-promoted lung cancer migration.


Asunto(s)
Carcinoma Pulmonar de Lewis/secundario , Depresión/complicaciones , Neoplasias Pulmonares/patología , Estrés Psicológico/complicaciones , Microambiente Tumoral/inmunología , Animales , Antidepresivos/administración & dosificación , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/prevención & control , Carcinoma Pulmonar de Lewis/psicología , Línea Celular Tumoral , Depresión/tratamiento farmacológico , Depresión/inmunología , Depresión/psicología , Progresión de la Enfermedad , Humanos , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Psicológico/inmunología , Estrés Psicológico/psicología , Linfocitos T Citotóxicos , Microambiente Tumoral/efectos de los fármacos
3.
J Cell Mol Med ; 24(18): 10768-10784, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32750747

RESUMEN

Distant metastases occur when non-small cell lung cancer (NSCLC) is at late stages. Bone metastasis is one of the most frequent metastases of NSCLC and leads to poor prognosis. It has been reported that high expression of BMP2 in NSCLC correlates with poor survival, but whether BMP2 contributes to NSCLC bone metastasis remains largely unknown. The activation of BMP signalling is found in metastatic bone tumours of mice Lewis lung carcinoma and predicts poor survival in human NSCLC. BMP2 signalling activation can enhance bone metastasis of Lewis lung carcinoma. Moreover, BMP2 secreted by stroma fibroblasts can promote the migration and invasion of NSCLC cells. Besides, in combination with pre-osteoblast and LLCs, BMP2 could enhance the differentiation of macrophages into osteoclasts to play roles in the osteolytic mechanism of NSCLC bone metastasis. Interestingly, NSCLC cells can also enrich BMP2 to pre-osteoblasts to function in the osteoblastic mechanism. Our results firstly demonstrate the detailed mechanisms about what roles BMP2 signalling play in enhancing NSCLC bone metastases. These findings provide a new potential therapy choice for preventing bone metastases of NSCLC via the inhibition of BMP2 signalling.


Asunto(s)
Proteína Morfogenética Ósea 2/fisiología , Neoplasias Óseas/secundario , Carcinoma Pulmonar de Lewis/secundario , Carcinoma de Pulmón de Células no Pequeñas/secundario , Neoplasias Pulmonares/fisiopatología , Proteínas de Neoplasias/fisiología , Células A549 , Animales , Neoplasias Óseas/complicaciones , Neoplasias Óseas/fisiopatología , Carcinoma Pulmonar de Lewis/fisiopatología , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Movimiento Celular , Femenino , Fibroblastos/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/fisiopatología , Osteoblastos/patología , Osteólisis/etiología , Osteólisis/fisiopatología , Células RAW 264.7 , Transducción de Señal , Organismos Libres de Patógenos Específicos , Células del Estroma/metabolismo
4.
Clin Exp Metastasis ; 37(1): 133-144, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31489536

RESUMEN

Acidic extracellular pH (pHe) is an important microenvironment for cancer cells. This study assessed whether adaptation to acidic pHe enhances the metastatic phenotype of tumor cells. The low metastatic variant of Lewis lung carcinoma (LLCm1) cells were subjected to stepwise acidification, establishing acidic pHe-adapted (LLCm1A) cells growing exponentially at pH 6.2. These LLCm1A cells showed increased production of matrix metalloproteinases (MMPs), including MMP-2, -3, -9, and -13, and pulmonary metastasis following injection into mouse tail veins. Although LLCm1A cells exhibited a fibroblastic shape, keratin-5 expression was increased and α-smooth muscle actin expression was reduced. Despite serial passage of these cells at pH 7.4, high invasive activity through Matrigel® was sustained for at least 28 generations. Thus, adaptation to acidic pHe resulted in a more invasive phenotype, which was sustained during passage at pH 7.4, suggesting that an acidic microenvironment at the primary tumor site is important in the acquisition of a metastatic phenotype.


Asunto(s)
Carcinoma Pulmonar de Lewis/secundario , Espacio Extracelular/química , Concentración de Iones de Hidrógeno , Pulmón/patología , Microambiente Tumoral , Animales , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología
5.
Cell Death Dis ; 10(12): 941, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31819035

RESUMEN

Bone marrow mesenchymal stem cells (BMSCs) are multipotent stromal cells that can differentiate into a variety of cell types. BMSCs are chemotactically guided towards the cancer cells and contribute to the formation of a cancer microenvironment. The homing of BMSCs was affected by various factors. Disseminated tumour cells (DTCs) in distant organs, especially in the bone marrow, are the source of cancer metastasis and cancer relapse. DTC survival is also determined by the microenvironment. Here we aim to elucidate how cancer-educated BMSCs promote the survival of cancer cells at primary tumour sites and distant sites. We highlight the dynamic change by identifying different gene expression signatures in intratumoral BMSCs and in BMSCs that move back in the bone marrow. Intratumoral BMSCs acquire high mobility and displayed immunosuppressive effects. Intratumoral BMSCs that ultimately home to the bone marrow exhibit a strong immunosuppressive function. Cancer-educated BMSCs promote the survival of lung cancer cells via expansion of MDSCs in bone marrow, primary tumour sites and metastatic sites. These Ly6G+ MDSCs suppress proliferation of T cells. CXCL5, nitric oxide and GM-CSF produced by cancer-educated BMSCs contribute to the formation of malignant microenvironments. Treatment with CXCL5 antibody, the iNOS inhibitor 1400w and GM-CSF antibody reduced MDSC expansion in the bone marrow, primary tumour sites and metastatic sites, and promoted the efficiency of PD-L1 antibody. Our study reveals that cancer-educated BMSCs are the component of the niche for primary lung cancer cells and DTCs, and that they can be the target for immunotherapy.


Asunto(s)
Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Supervivencia Celular/genética , Células Madre Mesenquimatosas/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Células Madre Neoplásicas/metabolismo , Células A549 , Aloinjertos , Animales , Médula Ósea/patología , Carcinoma Pulmonar de Lewis/patología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/genética , Transfección , Proteína Fluorescente Roja
6.
Yakugaku Zasshi ; 139(12): 1495-1500, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-31787635

RESUMEN

The roles of chondroitin sulfate (CS) and dermatan sulfate (DS) have been demonstrated in various biological events such as the construction of the extracellular matrix, tissue development, and cell signaling through interactions with extracellular matrix components, morphogens, and growth factors. Human genetic diseases, including skeletal abnormalities, connective tissue diseases, and heart defects, were reported to be caused by mutations in the genes encoding glycosyltransferases, epimerases, and sulfotransferases that are responsible for the biosynthesis of CS and DS. Glycobiological approaches revealed that mutations in CS- and DS-biosynthetic enzymes led to reductions in their enzymatic activities and in the levels of CS and DS. Furthermore, CS at the surface of tumor cells plays a key role in pulmonary metastasis. A receptor for advanced glycation end-products (RAGE) was predominantly expressed in the lung, and was identified as a functional receptor for CS chains. CS and anti-RAGE antibodies inhibited the pulmonary metastasis of not only Lewis lung carcinoma but also B16 melanoma cells. Hence, RAGE and CS are potential targets of drug discovery for pulmonary metastasis and a number of other pathological conditions involving RAGE in the pathogenetic mechanism. This review provides an overview of glycobiological studies on characterized genetic disorders caused by the impaired biosynthesis of CS, as well as DS, and on the pulmonary metastasis of Lewis lung carcinoma cells involving CS and RAGE.


Asunto(s)
Enfermedades Óseas/etiología , Enfermedades Óseas/genética , Carcinoma Pulmonar de Lewis/etiología , Carcinoma Pulmonar de Lewis/secundario , Condroitín/biosíntesis , Dermatán Sulfato/biosíntesis , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/secundario , Receptor para Productos Finales de Glicación Avanzada , Enfermedades de la Piel/etiología , Enfermedades de la Piel/genética , Animales , Carcinoma Pulmonar de Lewis/genética , Humanos , Neoplasias Pulmonares/genética , Ratones , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo
7.
Anticancer Res ; 39(4): 1729-1738, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30952712

RESUMEN

AIM: Monocyte chemotactic protein-1 (MCP1) is a potent adipokine. This study tested the hypothesis that adipose-produced MCP1 contributes to metastasis. MATERIALS AND METHODS: In a spontaneous metastasis model of Lewis lung carcinoma (LLC), male adipose MCP1-deficient (Mcp1-/-) and wild-type (WT) mice were fed the AIN93G diet or a high-fat diet (HFD) for 11 weeks. Lung metastasis from a subcutaneous tumor was the primary endpoint. RESULTS: The adipose expression of MCP1 was lower in Mcp1-/- mice than in WT controls. The HFD increased the number of lung metastases in WT mice. The number of metastasis was significantly lower in the HFD-fed Mcp1-/- mice than in the HFD-fed WT mice. Compared to the WT mice, adipose MCP1 deficiency lowered plasma concentrations of insulin, proinflammatory adipokines (leptin, plasminogen activator inhibitor-1, and resistin), and angiogenic markers (vascular endothelial growth factor, hepatocyte growth factor, and angiopoietin-2). CONCLUSION: Adipose MCP1 deficiency attenuates HFD-enhanced pulmonary metastasis of LLC.


Asunto(s)
Tejido Adiposo/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Quimiocina CCL2/deficiencia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Adipoquinas/metabolismo , Adiposidad , Proteínas Angiogénicas/metabolismo , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/prevención & control , Quimiocina CCL2/genética , Dieta Alta en Grasa , Mediadores de Inflamación/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Transducción de Señal
8.
Anticancer Res ; 39(4): 1739-1748, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30952713

RESUMEN

BACKGROUND/AIM: Obesity is a risk factor for cancer. Disruption of the daily feeding and fasting rhythm can contribute to obesity. This study tested the hypothesis that time-restricted feeding (TRF) attenuates obesity-enhanced metastasis. MATERIALS AND METHODS: In a spontaneous metastasis model of Lewis lung carcinoma (LLC), male C57BL/6 mice were fed the standard AIN93G diet or a high-fat diet (HFD) with or without dark-phase restricted feeding (12 h per day) for 10 weeks. Pulmonary metastases from a subcutaneous tumor were quantified. RESULTS: The number and size of lung metastases were greater in the HFD group than in the AIN93G group, but did not differ between the TRF and AIN93G groups. TRF prevented HFD-induced increases in plasma concentrations of glucose, insulin, proinflammatory cytokines (leptin, monocyte chemotactic protein-1, plasminogen activator inhibitor-1), and angiogenic factors (angiopoietin-2, hepatic growth factor, vascular endothelial growth factor). CONCLUSION: TRF attenuates the HFD-enhanced spontaneous metastasis of LLC in mice.


Asunto(s)
Carcinoma Pulmonar de Lewis/prevención & control , Carcinoma Pulmonar de Lewis/secundario , Dieta Alta en Grasa/efectos adversos , Ayuno , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Obesidad/prevención & control , Adipoquinas/sangre , Adiposidad , Proteínas Angiogénicas/sangre , Animales , Glucemia/metabolismo , Carcinoma Pulmonar de Lewis/sangre , Carcinoma Pulmonar de Lewis/genética , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Citocinas/sangre , Mediadores de Inflamación/sangre , Insulina/sangre , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/genética , Masculino , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/etiología , Obesidad/genética , Factores de Tiempo , Aumento de Peso
9.
Cancer Lett ; 442: 15-20, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30401632

RESUMEN

The transient receptor potential vanilloid 4 (TRPV4) channel is a mechanosensor in endothelial cells (EC) that regulates cyclic strain-induced reorientation and flow-mediated nitric oxide production. We have recently demonstrated that TRPV4 expression is reduced in tumor EC and tumors grown in TRPV4KO mice exhibited enhanced growth and immature leaky vessels. However, the mechanism by which TRPV4 regulates tumor vascular integrity and metastasis is not known. Here, we demonstrate that VE-cadherin expression at the cell-cell contacts is significantly reduced in TRPV4-deficient tumor EC and TRPV4KO EC. In vivo angiogenesis assays with Matrigel of varying stiffness (700-900 Pa) revealed a significant stiffness-dependent reduction in VE-cadherin-positive vessels in Matrigel plugs from TRPV4KO mice compared with WT mice, despite an increase in vessel growth. Further, syngeneic Lewis Lung Carcinomatumor experiments demonstrated a significant decrease in VE-cadherin positive vessels in TRPV4KO tumors compared with WT. Functionally, enhanced tumor cell metastasis to the lung was observed in TRPV4KO mice. Our findings demonstrate that TRPV4 channels regulate tumor vessel integrity by maintaining VE-cadherin expression at cell-cell contacts and identifies TRPV4 as a novel target for metastasis.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Movimiento Celular , Células Endoteliales/metabolismo , Uniones Intercelulares/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Mecanotransducción Celular , Neovascularización Patológica , Canales Catiónicos TRPV/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Células Endoteliales/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Uniones Intercelulares/genética , Uniones Intercelulares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
10.
Mol Cancer Res ; 17(3): 783-793, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30552233

RESUMEN

Increased levels of the chemokine CCL2 in cancer patients are associated with poor prognosis. Experimental evidence suggests that CCL2 correlates with inflammatory monocyte recruitment and induction of vascular activation, but the functionality remains open. Here, we show that endothelial Ccr2 facilitates pulmonary metastasis using an endothelial-specific Ccr2-deficient mouse model (Ccr2ecKO). Similar levels of circulating monocytes and equal leukocyte recruitment to metastatic lesions of Ccr2ecKO and Ccr2fl/fl littermates were observed. The absence of endothelial Ccr2 strongly reduced pulmonary metastasis, while the primary tumor growth was unaffected. Despite a comparable cytokine milieu in Ccr2ecKO and Ccr2fl/fl littermates the absence of vascular permeability induction was observed only in Ccr2ecKO mice. CCL2 stimulation of pulmonary endothelial cells resulted in increased phosphorylation of MLC2, endothelial cell retraction, and vascular leakiness that was blocked by an addition of a CCR2 inhibitor. These data demonstrate that endothelial CCR2 expression is required for tumor cell extravasation and pulmonary metastasis. IMPLICATIONS: The findings provide mechanistic insight into how CCL2-CCR2 signaling in endothelial cells promotes their activation through myosin light chain phosphorylation, resulting in endothelial retraction and enhanced tumor cell migration and metastasis.


Asunto(s)
Carcinoma Pulmonar de Lewis/metabolismo , Quimiocina CCL2/metabolismo , Células Endoteliales/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores CCR2/metabolismo , Animales , Permeabilidad Capilar , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/secundario , Movimiento Celular/fisiología , Células Endoteliales/patología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Metástasis de la Neoplasia
11.
Exp Oncol ; 40(4): 275-281, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30593747

RESUMEN

AIM: To investigate the effect of chicken embryo proteins (CEP) as a prototype of xenogeneic vaccine on immune reactions in mice immunized after Lewis lung carcinoma (LLC) surgical removal. MATERIALS AND METHODS: C57Bl male mice were immunized on days 1, 8, and 15 after surgical removal of LLC. The immune response was assessed on days 7, 14, 21 and 28 after tumor resection. Cytotoxic activity of natural killer cells (NK) and cytotoxic T-lymphocytes as well as antibody dependent cellular cytotoxicity was estimated in MTT-assay; specific antibodies were detected in ELISA; lymphocyte proliferation was tested in reaction of in vitro blast transformation. RESULTS: None of the immunized mice developed LLC metastases. Immunization with CEP seems to prevent the potential decrease in NK cell cytotoxic activity and spontaneous blast transformation activity of lymphocytes following the surgically induced stress. Further research on improving immunization schedule and elucidating the mechanisms of NK modulation with CEP is needed.


Asunto(s)
Proteínas Aviares/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Neoplasias Pulmonares/inmunología , Activación de Linfocitos , Animales , Anticuerpos Heterófilos/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma Pulmonar de Lewis/secundario , Embrión de Pollo , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Inmunización , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología
12.
Cancer Lett ; 420: 109-115, 2018 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-29408339

RESUMEN

Electrochemotherapy is an evolving therapy which has recently been shown to induce an immunogenic form of cell death. It is hypothesized that the immunogenic cell death induced by electrochemotherapy may compliment the responses seen with anti-cancer immunotherapies. We therefore examined the effect of electrochemotherapy in combination with ICOS activation, which promotes the activity of previously activated T cells. In comparison to either monotherapy which resulted in no curative outcomes in any model, in a CT26 primary tumour 50% of mice were cured, with 100% of cured mice surviving tumour rechallenge. In a dual flank CT26 model mimicking secondary disease 20% of mice were cured, and 30% of mice were cured using an aggressively metastatic Lewis Lung Carcinoma model. We have shown the novel combination of electrochemotherapy with ICOS activation can inhibit local and distal tumour growth, including total tumour clearance with long lasting immunological memory.


Asunto(s)
Anticuerpos/administración & dosificación , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/secundario , Neoplasias del Colon/tratamiento farmacológico , Proteína Coestimuladora de Linfocitos T Inducibles/agonistas , Animales , Anticuerpos/farmacología , Carcinoma Pulmonar de Lewis/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/inmunología , Citocinas/metabolismo , Electroquimioterapia/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones , Trasplante de Neoplasias , Análisis de Supervivencia , Resultado del Tratamiento
13.
J Biol Chem ; 292(47): 19198-19208, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-28924047

RESUMEN

Tumors depend on their microenvironment for sustained growth, invasion, and metastasis. In this environment, endothelial cells (ECs) are an important stromal cell type interacting with malignant cells to facilitate tumor angiogenesis and cancer cell extravasation. Of note, lysosomal acid lipase (LAL) deficiency facilitates melanoma growth and metastasis. ECs from LAL-deficient (lal-/-) mice possess enhanced proliferation, migration, and permeability of inflammatory cells by activating the mammalian target of rapamycin (mTOR) pathway. Here we report that lal-/- ECs facilitated in vivo tumor angiogenesis, growth, and metastasis, largely by stimulating tumor cell proliferation, migration, adhesion, and transendothelial migration via increased expression of IL-6 and monocyte chemoattractant protein 1 (MCP-1). This prompted us to look for lysosomal proteins that are involved in lal-/- EC dysfunctions. We found that lal-/- ECs displayed increased expression of Rab7, a late endosome/lysosome-associated small GTPase. Moreover, Rab7 and mTOR were co-increased and co-localized to lysosomes and physically interacted in lal-/- ECs. Rab7 inhibition reversed lal-/- EC dysfunctions, including decreasing their enhanced migration and permeability of tumor-stimulatory myeloid cells, and suppressed EC-mediated stimulation of in vitro tumor cell transmigration, proliferation, and migration and in vivo tumor growth and metastasis. Finally, Rab7 inhibition reduced overproduction of reactive oxygen species and increased IL-6 and MCP-1 secretion in lal-/- ECs. Our results indicate that metabolic reprogramming resulting from LAL deficiency enhances the ability of ECs to stimulate tumor cell proliferation and metastasis through stimulation of lysosome-anchored Rab7 activity.


Asunto(s)
Carcinoma Pulmonar de Lewis/secundario , Endotelio Vascular/enzimología , Lisosomas/enzimología , Melanoma Experimental/secundario , Esterol Esterasa/fisiología , Enfermedad de Wolman/complicaciones , Proteínas de Unión al GTP rab/metabolismo , Animales , Apoptosis , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Movimiento Celular , Proliferación Celular , Endotelio Vascular/patología , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Noqueados , Células Mieloides/metabolismo , Células Mieloides/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Especies Reactivas de Oxígeno , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Migración Transendotelial y Transepitelial , Células Tumorales Cultivadas , Enfermedad de Wolman/fisiopatología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión a GTP rab7 , Enfermedad de Wolman
14.
Cancer Res ; 77(18): 4809-4822, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28754672

RESUMEN

Dissociation from epithelial sheets and invasion through the surrounding stroma are critical early events during epithelial cancer metastasis. Here we find that a lymphocyte lineage-restricted transcription factor, Spi-B, is frequently expressed in human lung cancer tissues. The Spi-B-expressing cancer cells coexpressed vimentin but repressed E-cadherin and exhibited invasive behavior. Increased Spi-B expression was associated with tumor grade, lymphatic metastasis, and short overall survival. Mechanistically, Spi-B disrupted intercellular junctions and enhanced invasiveness by reconfiguring the chromatin structure of the tight junction gene claudin-2 (CLDN2) and repressing its transcription. These data suggest that Spi-B participates in mesenchymal invasion, linking epithelial cancer metastasis with a lymphatic transcriptional program. Cancer Res; 77(18); 4809-22. ©2017 AACR.


Asunto(s)
Carcinoma Pulmonar de Lewis/secundario , Claudina-2/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/patología , Factores de Transcripción/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/secundario , Animales , Apoptosis , Biomarcadores de Tumor , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/secundario , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundario , Proliferación Celular , Claudina-2/genética , Proteínas de Unión al ADN/genética , Humanos , Uniones Intercelulares , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/secundario , Factores de Transcripción/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Lett ; 402: 43-51, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28549790

RESUMEN

Id1 promotes carcinogenesis and metastasis, and predicts prognosis of non-small cell lung cancer (NSCLC)-adenocarcionoma patients. We hypothesized that Id1 may play a critical role in lung cancer colonization of the liver by affecting both tumor cells and the microenvironment. Depleted levels of Id1 in LLC (Lewis lung carcinoma cells, LLC shId1) significantly reduced cell proliferation and migration in vitro. Genetic loss of Id1 in the host tissue (Id1-/- mice) impaired liver colonization and increased survival of Id1-/- animals. Histologically, the presence of Id1 in tumor cells of liver metastasis was responsible for liver colonization. Microarray analysis comparing liver tumor nodules from Id1+/+ mice and Id1-/- mice injected with LLC control cells revealed that Id1 loss reduces the levels of EMT-related proteins, such as vimentin. In tissue microarrays containing 532 NSCLC patients' samples, we found that Id1 significantly correlated with vimentin and other EMT-related proteins. Id1 loss decreased the levels of vimentin, integrinß1, TGFß1 and snail, both in vitro and in vivo. Therefore, Id1 enables both LLC and the host microenvironment for an effective liver colonization, and may represent a novel therapeutic target to avoid NSCLC liver metastasis.


Asunto(s)
Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Movimiento Celular , Transición Epitelial-Mesenquimal , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Microambiente Tumoral , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/secundario , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína 1 Inhibidora de la Diferenciación/efectos de los fármacos , Proteína 1 Inhibidora de la Diferenciación/genética , Integrina beta1/genética , Integrina beta1/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Carga Tumoral , Vimentina/genética , Vimentina/metabolismo
16.
J Ethnopharmacol ; 192: 406-412, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27649680

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: A fruit of Ginkgo biloba L. also known as Ginkgo biloba, can be used for the treatment of cancer in Chinese traditional medicine. The scientific name of succulent skin, which is the episperm of Ginkgo nuts, is exocarp. Experiment shows that Ginkgo biloba exocarp extracts (GBEE) has the effects of immune promotion, cancer inhibition and etc. AIM OF STUDY: Study on the activity of GBEE against Lewis lung cancer (LLC) angiogenesis and its partial molecular mechanism. MATERIALS AND METHODS: The effect of GBEE on proliferation of LLC cells was detected by MTT method in vitro. The metastasis model of LLC was set up. The C57BL/6J mice were randomly separated into normal control, model control, positive control and GBEE (50, 100, 200mg/kg) treatment groups, n=10. The mice in normal group and model group were both intragastric gavage (i.g.) normal saline (NS) in a volume of 0.1mL/10g (b.w.), positive group were intraperitoneal (i.p.) injection cyclophosphamide (CPA) at a dose of 20mg/kg (b.w.) , the GBEE treatment groups were respectively i.g. GBEE 50, 100, and 200mg/kg (b.w.), once a day for 20d. After treatment, we calculated the tumor inhibition rate and anti-metastasis rate. The microvessel density (MVD) was measured by immunohistochemistry method in transplanted tumor. The expression levels of vascular en-dothelial growth factor (VEGF) and VEGFR2 mRNA or Wnt3a, ß-catenin, VEGF, VEGFR2 and p-Akt/Akt protein expression were respectively tested by Quantitative Reverse transcription Polymerase chain reaction (qRT-PCR) or western blot in vitro and vivo. RESULTS: GBEE suppressed the growth of LLC cells in a dose-dependent way at the dose of 5, 10, 20, 40, 80 and 160µg/mL in vitro. It can suppressed Wnt3a and ß-catenin protein expression and the content of mRNA of VEGF and VEGFR2 in LLC cells significantly. In vivo, we discovered GBEE can retard the growth of LLC transplanted tumor in a dose-dependent way at the dose of 50, 100, 200mg/kg, suppressing tumor lung metastasis. The expression of CD34 was reduced, which means MVD was inhibited and so do ß-catenin, VEGF, VEGFR2 and p-AKT/AKT protein expression and VEGF and VEGFR2 mRNA expression levels in LLC transplanted tumor of C57BL/6 mice. CONCLUSIONS: GBEE played the effects of anti-tumor and anti-metastatic depending upon the inhibition of tumor angiogenesis, which may be closely relevant to its effect in blockage of Wnt /ß-catenin-VEGF signaling pathway in LLC.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/farmacología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Ginkgo biloba/química , Microvasos/efectos de los fármacos , Neovascularización Patológica , Extractos Vegetales/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Inhibidores de la Angiogénesis/aislamiento & purificación , Animales , Antineoplásicos Fitogénicos/aislamiento & purificación , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclofosfamida/farmacología , Relación Dosis-Respuesta a Droga , Ratones Endogámicos C57BL , Microvasos/metabolismo , Microvasos/patología , Fosforilación , Fitoterapia , Extractos Vegetales/aislamiento & purificación , Plantas Medicinales , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
17.
Oncotarget ; 7(32): 52104-52114, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27438143

RESUMEN

OBJECTIVE: In this study, we aim to find out the role of microRNA-93-5p (miR-93) and Smad5 in morphine tolerance in mouse models of bone cancer pain (BCP). RESULTS: At 7 days after injection of morphine, the PMWT showed no significant difference between the morphine model group and the saline model group (P < 0.05), suggesting that morphine tolerance had formed in the morphine model group. The morphine model group had higher miR-93 expression and lower Smad5 mRNA expression than the saline model group. Smad5 is a downstream target gene of miR-93. At 7, 9 and 14 days after injection of lentiviruses, the L/anti-miR-93 group had the lowest PMWTs, while the Smad5 shRNA group presented the highest PMWTs among these five groups (all P < 0.05). METHODS: We built mouse models of BCP and morphine tolerance and recorded 50% PMWT. After 6 days of modeling, we set saline control group, morphine control, saline model group and morphine model group (morphine tolerance emerged). We performed luciferase reporter gene assay to verify the relation between miR-93 and Smad5. After lentivirus transfection, the mice with morphine tolerance were assigned into L/anti-miR-93 group, Smad5 shRNA group, L/anti-miR-93 + Smad5 shRNA group, blank group and PBS control group. RT-qPCR, Western Blot assay and immumohistochemical staining were performed to observe the changes of miR-93 and Smad5. CONCLUSION: Up-regulation of miR-93 may contribute to the progression of morphine tolerance by targeting Smad5 in mouse model of BCP.


Asunto(s)
Neoplasias Óseas/complicaciones , Dolor en Cáncer , Tolerancia a Medicamentos/genética , MicroARNs/genética , Proteína Smad5/metabolismo , Analgésicos Opioides/farmacología , Animales , Neoplasias Óseas/secundario , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/genética , Dolor en Cáncer/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Ratones , Ratones Endogámicos C57BL , Morfina/farmacología
18.
Proc Natl Acad Sci U S A ; 113(8): 2223-8, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26858439

RESUMEN

Circulating cancer cells can putatively colonize distant organs to form metastases or to reinfiltrate primary tumors themselves through a process termed "tumor self-seeding." Here we exploit this biological attribute to deliver tumor necrosis factor alpha (TNF), a potent antitumor cytokine, directly to primary and metastatic tumors in a mechanism that we have defined as "tumor self-targeting." For this purpose, we genetically engineered mouse mammary adenocarcinoma (TSA), melanoma (B16-F10), and Lewis lung carcinoma cells to produce and release murine TNF. In a series of intervention trials, systemic administration of TNF-expressing tumor cells was associated with reduced growth of both primary tumors and metastatic colonies in immunocompetent mice. We show that these malignant cells home to tumors, locally release TNF, damage neovascular endothelium, and induce massive cancer cell apoptosis. We also demonstrate that such tumor-cell-mediated delivery avoids or minimizes common side effects often associated with TNF-based therapy, such as acute inflammation and weight loss. Our study provides proof of concept that genetically modified circulating tumor cells may serve as targeted vectors to deliver anticancer agents. In a clinical context, this unique paradigm represents a personalized approach to be translated into applications potentially using patient-derived circulating tumor cells as self-targeted vectors for drug delivery.


Asunto(s)
Neoplasias Experimentales/terapia , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Apoptosis , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/secundario , Carcinoma Pulmonar de Lewis/terapia , Ingeniería Celular , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Sistemas de Liberación de Medicamentos , Endotelio Vascular/patología , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/secundario , Neoplasias Mamarias Experimentales/terapia , Melanoma Experimental/patología , Melanoma Experimental/secundario , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/patología , Neoplasias Experimentales/secundario , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Transducción Genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Oncotarget ; 7(2): 1529-43, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26625314

RESUMEN

Lysosomal acid lipase (LAL) is a key enzyme controlling neutral lipid metabolic signaling in myeloid-derived suppressor cells (MDSCs). MDSCs from LAL-deficient (lal-/-) mice directly stimulate cancer cell proliferation. PPARγ ligand treatment inhibited lal-/- MDSCs stimulation of tumor cell growth and metastasis in vivo, and tumor cell proliferation and migration in vitro. In addition, PPARγ ligand treatment impaired lal-/- MDSCs transendothelial migration, and differentiation from lineage-negative cells. The corrective effects of PPARγ ligand on lal-/- MDSCs functions were mediated by regulating the mammalian target of rapamycin (mTOR) pathway, and subsequently blocking MDSCs ROS overproduction. Furthermore, in the myeloid-specific dominant-negative PPARγ (dnPPARγ) overexpression bitransgenic mouse model, tumor growth and metastasis were enhanced, and MDSCs from these mice stimulated tumor cell proliferation and migration. MDSCs with dnPPARγ overexpression showed increased transendothelial migration, overactivation of the mTOR pathway, and ROS overproduction. These results indicate that PPARγ plays a critical role in neutral lipid metabolic signaling controlled by LAL, which provides a mechanistic basis for clinically targeting MDSCs to reduce the risk of cancer proliferation, growth and metastasis.


Asunto(s)
Carcinoma Pulmonar de Lewis/metabolismo , Comunicación Celular , Movimiento Celular , Proliferación Celular , Melanoma Experimental/metabolismo , Células Mieloides/metabolismo , PPAR gamma/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Antígenos Ly/metabolismo , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/secundario , Comunicación Celular/efectos de los fármacos , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Predisposición Genética a la Enfermedad , Ácidos Linoleicos Conjugados/farmacología , Melanoma Experimental/genética , Melanoma Experimental/secundario , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/efectos de los fármacos , PPAR gamma/agonistas , PPAR gamma/genética , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Esterol Esterasa/deficiencia , Esterol Esterasa/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Migración Transendotelial y Transepitelial , Transfección , Carga Tumoral
20.
Bull Exp Biol Med ; 160(1): 49-52, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26593414
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...