Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(3)2023 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-36768147

RESUMEN

Telomerase reverse transcriptase (TERT) plays a key role in the maintenance of telomere DNA length. The rs10069690 single nucleotide variant, located in intron 4 of TERT, was found to be associated with telomere length and the risk of estrogen receptor-negative but not-positive breast cancer. This study aimed at analysis of the association of rs10069690 genotype and TERT expression with the risk, age at onset, prognosis, and clinically and molecularly relevant subtypes of breast cancer. Accordingly, rs10069690 was genotyped in a hospital-based case-control study of 403 female breast cancer patients and 246 female controls of a Central European (Austrian) study population, and the mRNA levels of TERT were quantified in 106 primary breast tumors using qRT-PCR. We found that in triple-negative breast cancer patients, the minor rs10069690 TT genotype tended to be associated with an increased breast cancer risk (OR, 1.87; 95% CI, 0.75-4.71; p = 0.155) and was significantly associated with 11.7 years younger age at breast cancer onset (p = 0.0002), whereas the CC genotype was associated with a poor brain metastasis-free survival (p = 0.009). Overall, our data show that the rs10069690 CC genotype and a high TERT expression tended to be associated with each other and with a poor prognosis. Our findings indicate a key role of rs10069690 in triple-negative breast cancer.


Asunto(s)
Telomerasa , Neoplasias de la Mama Triple Negativas , Femenino , Humanos , Edad de Inicio , Estudios de Casos y Controles , Predisposición Genética a la Enfermedad , Genotipo , Polimorfismo de Nucleótido Simple , Telomerasa/genética , Neoplasias de la Mama Triple Negativas/genética
2.
Basic Res Cardiol ; 117(1): 42, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-36008727

RESUMEN

Sympathetic nerve denervation after myocardial infarction (MI) predicts risk of sudden cardiac death. Therefore, therapeutic approaches limit infarct size, improving adverse remodeling and restores sympathetic innervation have a great clinical potential. Remote ischemic perconditioning (RIPerc) could markedly attenuate MI-reperfusion (MIR) injury. In this study, we aimed to assess its effects on cardiac sympathetic innervation and metabolism. Transient myocardial ischemia is induced by ligature of the left anterior descending coronary artery (LAD) in male Sprague-Dawley rats, and in vivo cardiac 2-[18F]FDG and [11C]mHED PET scans were performed at 14-15 days after ischemia. RIPerc was induced by three cycles of 5-min-long unilateral hind limb ischemia and intermittent 5 min of reperfusion during LAD occlusion period. The PET quantitative parameters were quantified in parametric polar maps. This standardized format facilitates the regional radioactive quantification in deficit regions to remote areas. The ex vivo radionuclide distribution was additionally identified using autoradiography. Myocardial neuron density (tyrosine hydroxylase positive staining) and chondroitin sulfate proteoglycans (CSPG, inhibiting neuron regeneration) expression were assessed by immunohistochemistry. There was no significant difference in the mean hypometabolism 2-[18F]FDG uptake ratio (44.6 ± 4.8% vs. 45.4 ± 4.4%) between MIR rats and MIR + RIPerc rats (P > 0.05). However, the mean [11C]mHED nervous activity of denervated myocardium was significantly elevated in MIR + RIPerc rats compared to the MIR rats (35.9 ± 7.1% vs. 28.9 ± 2.3%, P < 0.05), coupled with reduced denervated myocardium area (19.5 ± 5.3% vs. 27.8 ± 6.6%, P < 0.05), which were associated with preserved left-ventricular systolic function, a less reduction in neuron density, and a significant reduction in CSPG and CD68 expression in the myocardium. RIPerc presented a positive effect on cardiac sympathetic-nerve innervation following ischemia, but showed no significant effect on myocardial metabolism.


Asunto(s)
Infarto del Miocardio , Daño por Reperfusión Miocárdica , Animales , Fluorodesoxiglucosa F18 , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley
3.
Int J Mol Sci ; 17(10)2016 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-27669225

RESUMEN

Neuroblastoma (NB) is the most common extracranial solid tumor of childhood and is a rapidly growing, highly-vascularized cancer. NBs frequently express angiogenic factors and high tumor angiogenesis has been associated with poor outcomes. Placental growth factor (PlGF) is an angiogenic protein belonging to the vascular endothelial growth factor (VEGF) family and is up-regulated mainly in pathologic conditions. Recently, PlGF was identified as a member of a gene expression signature characterizing highly malignant NB stem cells drawing attention as a potential therapeutic target in NB. In the present study, we sought to investigate the expression of PlGF in NB patients and the effect of PlGF inhibition on high-risk MYCN-non-amplified SK-N-AS NB xenografts. Human SK-N-AS cells, which are poorly differentiated and express PlGF and VEGF-A, were implanted subcutaneously in athymic nude mice. Treatment was done by intratumoral injection of replication-incompetent adenoviruses (Ad) expressing PlGF- or VEGF-specific short hairpin (sh)RNA, or soluble (s)VEGF receptor 2 (VEGFR2). The effect on tumor growth and angiogenesis was analyzed. High PlGF expression levels were observed in human advanced-stage NBs. Down-regulating PlGF significantly reduced NB growth in established NB xenografts by reducing cancer cell proliferation but did not suppress angiogenesis. In contrast, blocking VEGF by administration of Ad(sh)VEGF and Ad(s)VEGFR2 reduced tumor growth associated with decreased tumor vasculature. These findings suggest that PlGF and VEGF-A modulate MYCN-non-amplified NB tumors by different mechanisms and support a role for PlGF in NB biology.


Asunto(s)
Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/patología , Factor de Crecimiento Placentario/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adenoviridae/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Preescolar , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Lactante , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica , Neuroblastoma/metabolismo , Neuroblastoma/prevención & control , Factor de Crecimiento Placentario/antagonistas & inhibidores , Factor de Crecimiento Placentario/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , 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
4.
Int J Mol Sci ; 16(12): 29643-53, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26690424

RESUMEN

Like other RECQ helicases, WRN/RECQL2 plays a crucial role in DNA replication and the maintenance of genome stability. Inactivating mutations in RECQL2 lead to Werner syndrome, a rare autosomal disease associated with premature aging and an increased susceptibility to multiple cancer types. We analyzed the association of two coding single-nucleotide polymorphisms in WRN, Cys1367Arg (rs1346044), and Arg834Cys (rs3087425), with the risk, age at onset, and clinical subclasses of breast cancer in a hospital-based case-control study of an Austrian population of 272 breast cancer patients and 254 controls. Here we report that the rare homozygous CC genotype of rs1346044 was associated with an approximately two-fold elevated breast cancer risk. Moreover, patients with the CC genotype exhibited a significantly increased risk of developing breast cancer under the age of 55 in both recessive and log-additive genetic models. CC patients developed breast cancer at a mean age of 55.2 ± 13.3 years and TT patients at 60.2 ± 14.7 years. Consistently, the risk of breast cancer was increased in pre-menopausal patients in the recessive model. These findings suggest that the CC genotype of WRN rs1346044 may contribute to an increased risk and a premature onset of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Exodesoxirribonucleasas/genética , RecQ Helicasas/genética , Edad de Inicio , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Riesgo , Síndrome de Werner/genética , Helicasa del Síndrome de Werner
5.
Int J Mol Sci ; 15(1): 1538-53, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24451137

RESUMEN

The early growth response transcription factor Egr-1 controls cell specific responses to proliferation, differentiation and apoptosis. Expression of Egr-1 and downstream transcription is closely controlled and cell specific upregulation induced by processes such as hypoxia and ischemia has been previously linked to multiple aspects of cardiovascular injury. In this study, we showed constitutive expression of Egr-1 in cultured human ventricular cardiac fibroblasts, used adenoviral mediated gene transfer to study the effects of continuous Egr-1 overexpression and studied downstream transcription by Western blotting, immunohistochemistry and siRNA transfection. Apoptosis was assessed by fluorescence microscopy and flow cytometry in the presence of caspase inhibitors. Overexpression of Egr-1 directly induced apoptosis associated with caspase activation in human cardiac fibroblast cultures in vitro assessed by fluorescence microscopy and flow cytometry. Apoptotic induction was associated with a caspase activation associated loss of mitochondrial membrane potential and transient downstream transcriptional up-regulation of the pro-apoptotic gene product Siva-1. Suppression of Siva-1 induction by siRNA partially reversed Egr-1 mediated loss of cell viability. These findings suggest a previously unknown role for Egr-1 and transcriptional regulation of Siva-1 in the control of cardiac accessory cell death.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Fibroblastos/metabolismo , Regulación hacia Arriba , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ventrículos Cardíacos/citología , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
Int J Mol Sci ; 15(1): 712-24, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24402127

RESUMEN

The CYP19 gene encodes aromatase, an enzyme catalyzing the conversion of androgens to estrogens. Studies analyzing associations between single nucleotide polymorphisms in CYP19 and breast cancer risk have shown inconsistent results. The rs10046 polymorphism is located in the 3' untranslated region of the CYP19 gene, but the influence of this polymorphism on breast cancer risk is unclear. In this study, we investigated the impact of rs10046 SNP on breast cancer risk, age at onset and association with clinical characteristics in an Austrian population of 274 breast cancer patients and 253 controls. The results show that a significantly increased fraction of patients with the TT genotype of rs10046 develop breast cancer under the age of 50 (41.8% of TT patients, compared to 26.6% of C carriers; p = 0.018, Chi-square test). No rs10046 genotypes were significantly associated with increased breast cancer risk or patient characteristics other than age at onset. These results suggest that the rs10046 polymorphism in the CYP19 gene may have an effect on breast cancer susceptibility at an age under 50 in the investigated population.


Asunto(s)
Aromatasa/genética , Neoplasias de la Mama/genética , Polimorfismo de Nucleótido Simple , Regiones no Traducidas 3' , Edad de Inicio , Alelos , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/patología , Femenino , Genotipo , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Oportunidad Relativa , Premenopausia , Receptor ErbB-2/metabolismo , Factores de Riesgo
7.
J Transl Med ; 11: 295, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24279335

RESUMEN

BACKGROUND: Rho GTPases play important roles in cytoskeleton organization, cell cycle progression and are key regulators of tumor progression. Strategies to modulate increased Rho GTPase activities during cancer progression could have therapeutic potential. METHODS: We report here the characterization of a Cdc42-selective small-molecule inhibitor AZA197 for the treatment of colon cancer that was developed based on structural information known from previously developed compounds affecting Rho GTPase activation. We investigated the effects of AZA197 treatment on RhoA, Rac1 and Cdc42 activities and associated molecular mechanisms in colon cancer cells in vitro. Therapeutic effects of AZA197 were examined in vivo using a xenograft mouse model of SW620 human colon cancer cells. After treatment, tumors were excised and processed for Ki-67 staining, TUNEL assays and Western blotting to evaluate proliferative and apoptotic effects induced by AZA197. RESULTS: In SW620 and HT-29 human colon cancer cells, AZA197 demonstrated selectivity for Cdc42 without inhibition of Rac1 or RhoA GTPases from the same family. AZA197 suppressed colon cancer cell proliferation, cell migration and invasion and increased apoptosis associated with down-regulation of the PAK1 and ERK signaling pathways in vitro. Furthermore, systemic AZA197 treatment reduced tumor growth in vivo and significantly increased mouse survival in SW620 tumor xenografts. Ki-67 staining and tissue TUNEL assays showed that both inhibition of cell proliferation and induction of apoptosis associated with reduced PAK/ERK activation contributed to the AZA197-induced therapeutic effects in vivo. CONCLUSIONS: These data indicate the therapeutic potential of the small-molecule inhibitor AZA197 based on targeting Cdc42 GTPase activity to modulate colorectal cancer growth.


Asunto(s)
Neoplasias del Colon/patología , Regulación hacia Abajo/efectos de los fármacos , Indoles/farmacología , Terapia Molecular Dirigida , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Quinasas p21 Activadas/metabolismo , Células 3T3 , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Indoles/química , Indoles/uso terapéutico , L-Lactato Deshidrogenasa/metabolismo , Ratones , Invasividad Neoplásica , Unión Proteica/efectos de los fármacos , Pirimidinas/química , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Análisis de Supervivencia , Proteína de Unión al GTP cdc42/metabolismo
8.
Int J Mol Sci ; 14(9): 17958-71, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-24005860

RESUMEN

The growth and vascularization of prostate cancer is dependent on interactions between cancer cells and supporting stromal cells. The primary stromal cell type found in prostate tumors is the carcinoma-associated fibroblast, which produces placental growth factor (PlGF). PlGF is a member of the vascular endothelial growth factor (VEGF) family of angiogenic molecules and PlGF mRNA levels increase after androgen deprivation therapy in prostate cancer. In this study, we show that PlGF has a direct dose-dependent proliferative effect on human PC-3 prostate cancer cells in vitro and fibroblast-derived PlGF increases PC-3 proliferation in co-culture. In xenograft tumor models, intratumoral administration of murine PlGF siRNA reduced stromal-derived PlGF expression, reduced tumor burden and decreased the number of Ki-67 positive proliferating cells associated with reduced vascular density. These data show that targeting stromal PlGF expression may represent a therapeutic target for the treatment of prostate cancer.


Asunto(s)
Proteínas Gestacionales/metabolismo , Neoplasias de la Próstata/metabolismo , Células del Estroma/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Ratones , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
JACC Basic Transl Sci ; 8(9): 1160-1176, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37791301

RESUMEN

Chronic kidney disease is a global health problem affecting 10% to 12% of the population. Uremic cardiomyopathy is often characterized by left ventricular hypertrophy, fibrosis, and diastolic dysfunction. Dysregulation of neuregulin-1ß signaling in the heart is a known contributor to heart failure. The systemically administered recombinant human neuregulin-1ß for 10 days in our 5/6 nephrectomy-induced model of chronic kidney disease alleviated the progression of uremic cardiomyopathy and kidney dysfunction in type 4 cardiorenal syndrome. The currently presented positive preclinical data warrant clinical studies to confirm the beneficial effects of recombinant human neuregulin-1ß in patients with chronic kidney disease.

10.
Int J Cancer ; 126(6): 1339-52, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19711348

RESUMEN

The molecular mechanisms of tumor-host interactions that render neuroblastoma (NB) cells highly invasive are unclear. Cancer cells upregulate host stromal cell colony-stimulating factor-1 (CSF-1) production to recruit tumor-associated macrophages (TAMs) and accelerate tumor growth by affecting extracellular matrix remodeling and angiogenesis. By coculturing NB with stromal cells in vitro, we showed the importance of host CSF-1 expression for macrophage recruitment to NB cells. To examine this interaction in NB in vivo, mice bearing human CSF-1-expressing SK-N-AS and CSF-1-negative SK-N-DZ NB xenografts were treated with intratumoral injections of small interfering RNAs directed against mouse CSF-1. Significant suppression of both SK-N-AS and SK-N-DZ NB growth by these treatments was associated with decreased TAM infiltration, matrix metalloprotease (MMP)-12 levels and angiogenesis compared to controls, while expression of tissue inhibitors of MMPs increased following mouse CSF-1 blockade. Furthermore, Tie-2-positive and -negative TAMs recruited by host CSF-1 were identified in NB tumor tissue by confocal microscopy and flow cytometry. However, host-CSF-1 blockade prolonged survival only in CSF-1-negative SK-N-DZ NB. These studies demonstrated that increased CSF-1 production by host cells enhances TAM recruitment and NB growth and that the CSF-1 phenotype of NB tumor cells adversely affects survival.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/metabolismo , Neuroblastoma/patología , Interferencia de ARN , Células del Estroma/metabolismo , Animales , Western Blotting , Comunicación Celular , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Técnicas de Cocultivo , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Microscopía Confocal , Neuroblastoma/genética , Neuroblastoma/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología , Análisis de Supervivencia , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Trasplante Heterólogo
11.
Stem Cells ; 27(9): 2342-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19522014

RESUMEN

Prostate cancer tumor growth and neovascularization is promoted by an interplay between migratory tumor stromal cells such as specialized tumor-associated macrophages (TAMs) and circulating endothelial precursor cells (CEPs). As vehicles for tumor therapy, human CEPs are relatively easy to isolate from peripheral blood, are able to proliferate long-term in vitro, are amenable to viral manipulation, and preferentially home to regions of ischemia found in growing tumors. We show here that human peripheral blood CEPs expanded ex vivo migrate to prostate cancer cells in vitro and efficiently home to human prostate tumor xenografts in vivo. Infection of precursors ex vivo with an adenovirus constructed to secrete a soluble form of the colony-stimulating factor-1 receptor CD115 that inhibits macrophage viability and migration in vitro significantly decreases the number of TAMs in xenografts (p < .05), reduces proliferation (p < .01) and vascular density (p < .03), and suppresses the growth of xenografts (p < .03). These data show for the first time that targeting stromal cell processes with cellular therapy has the potential to retard prostate tumor growth.


Asunto(s)
Adenoviridae/genética , Células Endoteliales/metabolismo , Neoplasias de la Próstata/terapia , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Endoteliales/citología , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Trasplante de Células Madre/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancers (Basel) ; 12(2)2020 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-32028699

RESUMEN

Macrophages form a major component of the leukocyte infiltrate in solid tumors and it has become increasingly clear that tumor-associated macrophages (TAMs) have tumor-promoting effects within the stroma [1]. Renal cell carcinoma (RCC) solid tumors are comprised of a heterogeneous microenvironment of both malignant and normal stromal cells containing large numbers of macrophages [2].We read with interest the paper by Suguru Kadomoto et al. entitled "Tumor-associated macrophages induce migration of renal cell carcinoma cells via activation of the CCL20-CCR6 axis", published in Cancers [3], in which they report that the CCL20-CCR6 axis induces migration and epithelial-mesenchymal transition (EMT) of ACHN and Caki-1 RCC cells in co-cultures with THP-1/U937-derived tumor conditioned macrophages.[...].

13.
Methods Mol Biol ; 2115: 289-325, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32006408

RESUMEN

Tumor-associated macrophages (TAMs) are representing a major leukocyte population in solid tumors. Macrophages are very heterogeneous and plastic cells and can acquire distinct functional phenotypes ranging from antitumorigenic to immunosuppressive tumor-promoting M2-like TAMs, depending on the local tissue microenvironment (TME). TAMs express cytokines, chemokines, growth factors, and extracellular matrix (ECM) modifying factors, and the cross talk with the TME regulates pathways involved in the recruitment, polarization, and metabolism of TAMs during tumor progression. Due to their crucial role in tumor growth and metastasis, selective targeting of TAM for the treatment of cancer with therapeutic agents that promote phagocytosis or suppress survival, proliferation, trafficking, or polarization of TAMs may prove to be beneficial in cancer therapy. In this chapter, we will discuss TAM biology and current strategies for the targeting of TAMs using small interfering RNA (siRNA)-based drugs. In the past few years, advances in the field of nanomedicine pave the way for the development of siRNA-based drugs as an additional class of personalized cancer immuno-nanomedicines. Fundamental challenges associated with this group of therapeutics include the development process, delivery system, and clinical translation for siRNA-based drugs.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/terapia , Interferencia de ARN , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia/métodos , Macrófagos Asociados a Tumores/metabolismo , Animales , Humanos , Nanomedicina/métodos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , ARN Interferente Pequeño/administración & dosificación , Microambiente Tumoral , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/patología
14.
Cancer Res ; 67(3): 1038-45, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17283136

RESUMEN

The interplay between malignant and stromal cells is essential in tumorigenesis. We have previously shown that colony-stimulating factor (CSF)-1, matrix metalloprotease (MMP)-2, and vascular endothelial growth factor (VEGF)-A production by stromal cells is enhanced by CSF-1-negative SW620 colon cancer cells. In the present study, the mechanisms by which colon cancer cells up-regulate host factors to promote tumorigenesis were investigated. Profiling of tumor cell cytokine expression in SW620 tumor xenografts in nude mice showed increased human tumor necrosis factor (TNF)-alpha mRNA expression with tumor growth. Incubation of macrophages with small interfering (si) RNAs directed against TNF-alpha or TNF-alpha-depleted SW620 cell conditioned medium versus SW620 cell conditioned medium failed to support mouse macrophage proliferation, migration, and expression of CSF-1, VEGF-A, and MMP-2 mRNAs. Consistent with these results, human TNF-alpha gene silencing decreased mouse macrophage TNF-alpha, CSF-1, MMP-2, and VEGF-A mRNA expression in macrophages cocultured with human cancer cells. In addition, inhibition of human TNF-alpha or mouse CSF-1 expression by siRNA reduced tumor growth in SW620 tumor xenografts in mice. These results suggest that colon cancer cell-derived TNF-alpha stimulates TNF-alpha and CSF-1 production by macrophages, and that CSF-1, in turn, induces macrophage VEGF-A and MMP-2 in an autocrine manner. Thus, interrupting tumor cell-macrophage communication by targeting TNF-alpha may provide an alternative therapeutic approach for the treatment of colon cancer.


Asunto(s)
Neoplasias del Colon/metabolismo , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Expresión Génica , Humanos , Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/patología , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
15.
Cardiovasc Res ; 79(3): 395-404, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18436538

RESUMEN

AIMS: Skeletal myoblasts are used in repair of ischaemic myocardium. However, a large fraction of grafted myoblasts degenerate upon engraftment. Colony-stimulating factor-1 (CSF-1) accelerates myoblast proliferation and angiogenesis. We hypothesized that CSF-1 overexpression improves myoblast survival and cardiac function in ischaemia-induced heart failure. METHODS AND RESULTS: Three weeks following myocardial infarction, rats developed heart failure and received intramyocardial injections of mouse CSF-1-transfected or untransfected primary autologous rat myoblasts, recombinant human CSF-1, mouse CSF-1 expressing plasmids, or culture medium. Tissue gene and protein expression was measured by quantitative RT-PCR (reverse transcription-polymerase chain reaction) and western blotting. Fluorescence imaging and immunocytochemistry were used to analyse myoblasts, endothelial cells, macrophages, and infarct wall thickening. Electrocardiograms were recorded online using a telemetry system. Left ventricular function was assessed by echocardiography over time, and improved significantly only in the CSF-1-overexpressing myoblast group. CSF-1-overexpression enhanced myoblast numbers and was associated with an increased infarct wall thickness, enhanced angiogenesis, increased macrophage recruitment and upregulated matrix metalloproteases (MMP)-2 and -12 in the zone bordering the infarction. Transplantation of CSF-1-overexpressing myoblasts did not result in major arrhythmias. CONCLUSION: Autologous intramyocardial transplantation of CSF-1 overexpressing myoblasts might be a novel strategy in the treatment of ischaemia-induced heart failure.


Asunto(s)
Terapia Genética/métodos , Insuficiencia Cardíaca/terapia , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Mioblastos Esqueléticos/trasplante , Isquemia Miocárdica/terapia , Miocardio/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/metabolismo , Masculino , Metaloproteinasas de la Matriz Secretadas/metabolismo , Ratones , Mioblastos Esqueléticos/metabolismo , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Miocardio/enzimología , Miocardio/patología , Neovascularización Fisiológica , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/biosíntesis , Factores de Tiempo , Transfección , Trasplante Autólogo , Función Ventricular Izquierda , Remodelación Ventricular
16.
Front Biosci ; 13: 5571-9, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508606

RESUMEN

Tumor cells can stimulate matrix metalloproteinase (MMP) production by stromal cells through cell-cell interactions mediated by cell adhesion molecules such as extracellular matrix metalloproteinase inducer (human CD147/EMMPRIN, mouse CD147/Basigin). This study sought to characterize whether specific tumor-stromal cell interactions mediated by CD147 promote colon cancer growth by utilizing small interfering (si)RNAs directed against human CD147/EMMPRIN or mouse CD147/Basigin in co-cultures of cancer cells with macrophages and fibroblasts and established human SW620 colon cancer xenograft models in immune deficient mice. We show that blockade of host (mouse) CD147/Basigin expression, but not cancer cell-derived CD147/EMMPRIN, suppresses tumor growth in human colon cancer xenografts. Experiments in vitro indicated that colon cancer cell-stromal cell interactions mediated by CD147 lead to increased MMP-2 expression in fibroblasts but not macrophages. Furthermore, expression of host VEGF-A in both fibroblasts and macrophages is independent of CD147 in vitro and in vivo. Interestingly, inhibition of cancer cell-derived EMMPRIN leads to increased MMP-9 levels in vivo. Our findings provide new insights into CD147-mediated tumor-host interactions mediating colon cancer growth.


Asunto(s)
Basigina/genética , Neoplasias del Colon/patología , ARN Interferente Pequeño/genética , Células 3T3 , Animales , División Celular , ADN Complementario/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Macrófagos/fisiología , Ratones , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/fisiología , Transfección , Trasplante Heterólogo
17.
Oncotarget ; 9(33): 23126-23148, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29796177

RESUMEN

Interleukin-34 (IL-34) is a ligand for the CSF-1R and has also two additional receptors, PTPRZ1 and syndecan-1. IL-34 plays a role in innate immunity, inflammation, and cancer. However, the role of IL-34 in breast cancer is still ill-defined. We analyzed IL-34 mRNA expression in breast cancer cell lines and breast cancer patients and applied established computational approaches (CIBERSORT, ESTIMATE, TIMER, TCIA), to analyze gene expression data from The Cancer Genome Atlas (TCGA). Expression of IL-34 was associated with a favorable prognosis in luminal and HER2 but not basal breast cancer patients. Gene expression of CSF-1 and CSF-1R was strongly associated with myeloid cell infiltration, while we found no or only weak correlations between IL-34, PTPRZ1, syndecan-1 and myeloid cells. In vitro experiments showed that tyrosine phosphorylation of CSF-1R, ERK, and FAK and cell migration are differentially regulated by IL-34 and CSF-1 in breast cancer cell lines. Collectively, our data suggest that correlation of IL-34 gene expression with survival is dependent on the molecular breast cancer subtype. Furthermore, IL-34 is not associated with myeloid cell infiltration and directly regulates breast cancer cell migration and signaling.

18.
Cancer Res ; 64(15): 5378-84, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15289345

RESUMEN

Colony-stimulating factor (CSF)-1 is the primary regulator of tissue macrophage production. CSF-1 expression is correlated with poor prognosis in breast cancer and is believed to enhance mammary tumor progression and metastasis through the recruitment and regulation of tumor-associated macrophages. Macrophages produce matrix metalloproteases (MMPs) and vascular endothelial growth factor, which are crucial for tumor invasion and angiogenesis. Given the important role of CSF-1, we hypothesized that blockade of CSF-1 or the CSF-1 receptor (the product of the c-fms proto-oncogene) would suppress macrophage infiltration and mammary tumor growth. Human MCF-7 mammary carcinoma cell xenografts in mice were treated with either mouse CSF-1 antisense oligonucleotide for 2 weeks or five intratumoral injections of either CSF-1 small interfering RNAs or c-fms small interfering RNAs. These treatments suppressed mammary tumor growth by 50%, 45%, and 40%, respectively, and selectively down-regulated target protein expression in tumor lysates. Host macrophage infiltration; host MMP-12, MMP-2, and vascular endothelial growth factor A expression; and endothelial cell proliferation within tumors of treated mice were decreased compared with tumors in control mice. In addition, mouse survival significantly increased after CSF-1 blockade. These studies demonstrate that CSF-1 and CSF-1 receptor are potential therapeutic targets for the treatment of mammary cancer.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Neoplasias Mamarias Animales/prevención & control , Oligonucleótidos Antisentido/uso terapéutico , ARN Interferente Pequeño/uso terapéutico , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Animales , División Celular , Regulación hacia Abajo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Metaloproteinasa 12 de la Matriz , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proto-Oncogenes Mas , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Oncotarget ; 7(29): 46187-46202, 2016 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-27323822

RESUMEN

Frizzled2 (FZD2) is a receptor for Wnts and may activate both canonical and non-canonical Wnt signaling pathways in cancer. However, no studies have reported an association between FZD2 signaling and high-risk NB so far. Here we report that FZD2 signaling pathways are critical to NB growth in MYCN-single copy SK-N-AS and MYCN-amplified SK-N-DZ high-risk NB cells. We demonstrate that stimulation of FZD2 by Wnt3a and Wnt5a regulates ß-catenin-dependent and -independent Wnt signaling factors. FZD2 blockade suppressed ß-catenin-dependent signaling activity and increased phosphorylation of PKC, AKT and ERK in vitro, consistent with upregulation of ß-catenin-independent signaling activity. Finally, FZD2 small interfering RNA knockdown suppressed tumor growth in murine NB xenograft models associated with suppressed ß-catenin-dependent signaling and a less vascularized phenotype in both NB xenografts. Together, our study suggests a role for FZD2 in high-risk NB cell growth and provides a potential candidate for therapeutic inhibition in FZD2-expressing NB patients.


Asunto(s)
Receptores Frizzled/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Vía de Señalización Wnt/fisiología , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Xenoinjertos , Humanos , Ratones , Ratones Desnudos
20.
Cardiovasc Res ; 109(2): 331-43, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26676850

RESUMEN

AIMS: Shock wave therapy (SWT) represents a clinically widely used angiogenic and thus regenerative approach for the treatment of ischaemic heart or limb disease. Despite promising results in preclinical and clinical trials, the exact mechanism of action remains unknown. Toll-like receptor 3, which is part of the innate immunity, is activated by binding double-stranded (ds) RNA. It plays a key role in inflammation, a process that is needed also for angiogenesis. We hypothesize that SWT causes cellular cavitation without damaging the target cells, thus liberating cytoplasmic RNA that in turn activates TLR3. METHODS AND RESULTS: SWT induces TLR3 and IFN-ß1 gene expression as well as RNA liberation from endothelial cells in a time-dependant manner. Conditioned medium from SWT-treated HUVECs induced TLR3 signalling in reporter cells. The response was lost when the medium was treated with RNase III to abolish dsRNAs or when TLR3 was silenced using siRNAs. In a mouse hind limb ischaemia model using wt and TLR3(-/-) mice (n = 6), SWT induced angiogenesis and arteriogenesis only in wt animals. These effects were accompanied by improved blood perfusion of treated limbs. Analysis of main molecules of the TLR3 pathways confirmed TLR3 signalling in vivo following SWT. CONCLUSION: Our data reveal a central role of the innate immune system, namely Toll-like receptor 3, to mediate angiogenesis upon release of cytoplasmic RNAs by mechanotransduction of SWT.


Asunto(s)
Células Endoteliales/metabolismo , Inmunidad Innata/inmunología , Inflamación/metabolismo , Mecanotransducción Celular/fisiología , Neovascularización Patológica/metabolismo , Transducción de Señal , Animales , Isquemia/metabolismo , Masculino , Ratones Endogámicos C57BL , ARN Bicatenario/metabolismo , Receptor Toll-Like 3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA