Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 46(5): e259-e264, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38691086

RESUMEN

We evaluated whether socioeconomic status (SES), race/ethnicity, and their interaction were associated with the presentation of advanced stage at diagnosis in embryonal tumors. Children 0 to 19 years of age diagnosed with embryonal tumors between 2006 and 2018 were identified from the US Surveillance, Epidemiology, and End Results program database specialized with Census Tract SES/Rurality. SES quintile was derived from a composite index for census tracts. We performed logistic regression to estimate odds ratios (ORs) and 95% confidence intervals by SES and race/ethnicity, adjusting for sex, age, and diagnosis year. Overall, no significant associations were found between either SES or race/ethnicity and the risk of presenting with advanced stage at diagnosis, although patterns of risk reductions were observed in atypical teratoid/rhabdoid tumors and embryonal rhabdomyosarcoma with increasing SES. In the stratified analysis, decreased odds of presenting with advanced-stage embryonal rhabdomyosarcoma were observed for Hispanics with higher SES (OR: 0.24, 95% Confidence Interval: 0.08-0.75) compared with Hispanics with lower SES. Future studies incorporating individual-level SES, cancer-specific staging information, and potential demographic, clinical, epidemiological, and genetic risk factors are warranted to confirm our findings.


Asunto(s)
Neoplasias de Células Germinales y Embrionarias , Clase Social , Humanos , Lactante , Femenino , Masculino , Preescolar , Niño , Adolescente , Neoplasias de Células Germinales y Embrionarias/epidemiología , Neoplasias de Células Germinales y Embrionarias/diagnóstico , Neoplasias de Células Germinales y Embrionarias/patología , Neoplasias de Células Germinales y Embrionarias/etnología , Recién Nacido , Adulto Joven , Programa de VERF , Factores de Riesgo , Estadificación de Neoplasias , Etnicidad/estadística & datos numéricos , Estados Unidos/epidemiología
2.
Lab Invest ; 100(1): 38-51, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31409888

RESUMEN

Neuroblastoma (NB) is a pediatric tumor of the peripheral nervous system. Treatment of the disease represents an unsolved clinical problem, as survival of patients with aggressive form of NB remains below 50%. Despite recent identification of numerous potential therapeutic targets, clinical trials validating them are challenging due to the rarity of the disease and its high patient-to-patient heterogeneity. Hence, there is a need for the accurate preclinical models that would allow testing novel therapeutic approaches and prioritizing the clinical studies, preferentially in personalized way. Here, we propose using conditional reprogramming (CR) technology for rapid development of primary NB cell cultures that could become a new model for such tests. This newly established method allowed for indefinite propagation of normal and tumor cells of epithelial origin in an undifferentiated state by their culture in the presence of Rho-associated kinase (ROCK) inhibitor, Y-27632, and irradiated mouse feeder cells. Using a modification of this approach, we isolated cell lines from tumors arising in the TH-MYCN murine transgenic model of NB (CR-NB). The cells were positive for neuronal markers, including Phox2B and peripherin and consisted of two distinct populations: mesenchymal and adrenergic expressing corresponding markers of their specific lineage. This heterogeneity of the CR-NB cells mimicked the different tumor cell phenotypes in TH-MYCN tumor tissues. The CR-NB cells preserved anchorage-independent growth capability and were successfully passaged, frozen and biobanked. Further studies are required to determine the utility of this method for isolation of human NB cultures, which can become a novel model for basic, translational, and clinical research, including individualized drug testing.


Asunto(s)
Línea Celular Tumoral , Neuroblastoma/patología , Animales , Biomarcadores/metabolismo , Técnicas de Reprogramación Celular , Humanos , Ratones Transgénicos , Neoplasias Experimentales , Neuroblastoma/metabolismo , Fenotipo , Ratas
3.
Pathobiology ; 87(2): 87-99, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32045912

RESUMEN

The tumor microenvironment (TME) plays an essential role in the development and progression of neoplasms. TME consists of the extracellular matrix and numerous specialized cells interacting with cancer cells by paracrine and autocrine mechanisms. Tumor axonogenesis and neoneurogenesis constitute a developing area of investigation. Prostate cancer (PC) is one of the most common malignancies in men worldwide. During the past years, more and more studies have shown that mechanisms leading to the development of PC are not confined only to the epithelial cancer cell, but also involve the tumor stroma. Different nerve types and neurotransmitters present within the TME are thought to be important factors in PC biology. Moreover, perineural invasion, which is a common way of PC spreading, in parallel creates the neural niche for malignant cells. Cancer neurobiology seems to have become a new discipline to explore the contribution of neoplastic cell interactions with the nervous system and the neural TME component, also to search for potential therapeutic targets in malignant tumors such as PC.


Asunto(s)
Sistema Nervioso/patología , Neoplasias de la Próstata/patología , Microambiente Tumoral , Animales , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Neoplasias de la Próstata/terapia , Transducción de Señal
4.
Am J Pathol ; 186(11): 3040-3053, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27743558

RESUMEN

Neuroblastoma (NB) is a pediatric malignant neoplasm of sympathoadrenal origin. Challenges in its management include stratification of this heterogeneous disease and a lack of both adequate treatments for high-risk patients and noninvasive biomarkers of disease progression. Our previous studies have identified neuropeptide Y (NPY), a sympathetic neurotransmitter expressed in NB, as a potential therapeutic target for these tumors by virtue of its Y5 receptor (Y5R)-mediated chemoresistance and Y2 receptor (Y2R)-mediated proliferative and angiogenic activities. The goal of this study was to determine the clinical relevance and utility of these findings. Expression of NPY and its receptors was evaluated in corresponding samples of tumor RNA, tissues, and sera from 87 patients with neuroblastic tumors and in tumor tissues from the TH-MYCN NB mouse model. Elevated serum NPY levels correlated with an adverse clinical presentation, poor survival, metastasis, and relapse, whereas strong Y5R immunoreactivity was a marker of angioinvasive tumor cells. In NB tissues from TH-MYCN mice, high immunoreactivity of both NPY and Y5R marked angioinvasive NB cells. Y2R was uniformly expressed in undifferentiated tumor cells, which supports its previously reported role in NB cell proliferation. Our findings validate NPY as a therapeutic target for advanced NB and implicate the NPY/Y5R axis in disease dissemination. The correlation between elevated systemic NPY and NB progression identifies serum NPY as a novel NB biomarker.


Asunto(s)
Neuroblastoma/metabolismo , Neuropéptido Y/metabolismo , Adolescente , Animales , Biomarcadores/metabolismo , Proliferación Celular , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Neuropéptido Y/genética , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo
5.
Cancer ; 121(5): 697-707, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25387699

RESUMEN

BACKGROUND: Ewing sarcoma (ES) is driven by fusion of the Ewing sarcoma breakpoint region 1 gene (EWSR1) with an E26 transformation-specific (ETS) transcription factor (EWS-ETS), most often the Friend leukemia integration 1 transcription factor (FLI1). Neuropeptide Y (NPY) is an EWS-FLI1 transcriptional target; it is highly expressed in ES and exerts opposing effects, ranging from ES cell death to angiogenesis and cancer stem cell propagation. The functions of NPY are regulated by dipeptidyl peptidase IV (DPPIV), a hypoxia-inducible enzyme that cleaves the peptide and activates its growth-promoting actions. The objective of this study was to determine the clinically relevant functions of NPY by identifying the associations between patients' ES phenotype and their NPY concentrations and DPP activity. METHODS: NPY concentrations and DPP activity were measured in serum samples from 223 patients with localized ES and 9 patients with metastatic ES provided by the Children's Oncology Group. RESULTS: Serum NPY levels were elevated in ES patients compared with the levels in a healthy control group and an osteosarcoma patient population, and the elevated levels were independent of EWS-ETS translocation type. Significantly higher NPY concentrations were detected in patients with ES who had tumors of pelvic and bone origin. A similar trend was observed in patients with metastatic ES. There was no effect of NPY on survival in patients with localized ES. DPP activity in sera from patients with ES did not differ significantly from that in healthy controls and patients with osteosarcoma. However, high DPP levels were associated with improved survival. CONCLUSIONS: Systemic NPY levels are elevated in patients with ES, and these high levels are associated with unfavorable disease features. DPPIV in serum samples from patients with ES is derived from nontumor sources, and its high activity is correlated with improved survival.


Asunto(s)
Neoplasias Óseas/sangre , Dipeptidil Peptidasa 4/sangre , Neuropéptido Y/sangre , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/sangre , Adolescente , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/mortalidad , Línea Celular Tumoral , Niño , Dipeptidil Peptidasa 4/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Osteosarcoma/sangre , Osteosarcoma/genética , ARN Mensajero/genética , Receptores de Neuropéptido Y/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/mortalidad , Trasplante Heterólogo
6.
FASEB J ; 27(6): 2244-55, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23457218

RESUMEN

We previously reported that the sympathetic neurotransmitter neuropeptide Y (NPY) is potently angiogenic, primarily through its Y2 receptor, and that endogenous NPY is crucial for capillary angiogenesis in rodent hindlimb ischemia. Here we sought to identify the source of NPY responsible for revascularization and its mechanisms of action. At d 3, NPY(-/-) mice demonstrated delayed recovery of blood flow and limb function, consistent with impaired collateral conductance, while ischemic capillary angiogenesis was reduced (~70%) at d 14. This biphasic temporal response was confirmed by 2 peaks of NPY activation in rats: a transient early increase in neuronally derived plasma NPY and increase in platelet NPY during late-phase recovery. Compared to NPY-null platelets, collagen-activated NPY-rich platelets were more mitogenic (~2-fold vs. ~1.6-fold increase) for human microvascular endothelial cells, and Y2/Y5 receptor antagonists ablated this difference in proliferation. In NPY(+/+) mice, ischemic angiogenesis was prevented by platelet depletion and then restored by transfusion of platelets from NPY(+/+) mice, but not NPY(-/-) mice. In thrombocytopenic NPY(-/-) mice, transfusion of wild-type platelets fully restored ischemia-induced angiogenesis. These findings suggest that neuronally derived NPY accelerates the early response to femoral artery ligation by promoting collateral conductance, while platelet-derived NPY is critical for sustained capillary angiogenesis.


Asunto(s)
Plaquetas/metabolismo , Isquemia/sangre , Neovascularización Fisiológica , Neuropéptido Y/fisiología , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Miembro Posterior , Humanos , Isquemia/genética , Isquemia/fisiopatología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Neovascularización Fisiológica/genética , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Ratas , Ratas Wistar
7.
Nat Med ; 13(7): 803-11, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17603492

RESUMEN

The relationship between stress and obesity remains elusive. In response to stress, some people lose weight, whereas others gain. Here we report that stress exaggerates diet-induced obesity through a peripheral mechanism in the abdominal white adipose tissue that is mediated by neuropeptide Y (NPY). Stressors such as exposure to cold or aggression lead to the release of NPY from sympathetic nerves, which in turn upregulates NPY and its Y2 receptors (NPY2R) in a glucocorticoid-dependent manner in the abdominal fat. This positive feedback response by NPY leads to the growth of abdominal fat. Release of NPY and activation of NPY2R stimulates fat angiogenesis, macrophage infiltration, and the proliferation and differentiation of new adipocytes, resulting in abdominal obesity and a metabolic syndrome-like condition. NPY, like stress, stimulates mouse and human fat growth, whereas pharmacological inhibition or fat-targeted knockdown of NPY2R is anti-angiogenic and anti-adipogenic, while reducing abdominal obesity and metabolic abnormalities. Thus, manipulations of NPY2R activity within fat tissue offer new ways to remodel fat and treat obesity and metabolic syndrome.


Asunto(s)
Dieta , Neuropéptido Y/genética , Obesidad/metabolismo , Estrés Fisiológico/metabolismo , Células 3T3-L1 , Tejido Adiposo Blanco/metabolismo , Animales , Frío , Grasas de la Dieta , Eliminación de Gen , Regulación de la Expresión Génica , Masculino , Síndrome Metabólico , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/genética , Regulación hacia Arriba
8.
FASEB J ; 26(8): 3528-36, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22539639

RESUMEN

Neuropeptide Y (NPY) mediates stress-induced obesity in adult male mice by activating its Y2 receptor (Y2R) in visceral adipose tissue (VAT). Here, we studied whether the NPY-Y2R system is also activated by maternal low-protein diet (LPD) and linked to obesity in offspring. Prenatal LPD offspring had lower birth weights compared to normal-protein diet (NPD) offspring. Female prenatal and lactation stress (PLS) offspring from mothers fed an LPD developed abdominal adiposity and glucose intolerance associated with a 5-fold up-regulation of NPY mRNA and a 6-fold up-regulation of Y2R mRNA specifically in VAT, in addition to elevated platelet-rich-plasma (PRP) NPY, compared to control females fed a high-fat diet (HFD). Conversely, PLS male offspring showed lower NPY in PRP, a 10-fold decrease of Y2R mRNA in VAT, lower adiposity, and improved glucose tolerance compared to control males. Interestingly, prenatal LPD offspring cross-fostered to control lactating mothers had completely inverse metabolic and NPY phenotypes. Taken together, these findings suggested that maternal LPD activates the VAT NPY-Y2R system and increases abdominal adiposity and glucose intolerance in a sex- and time-specific fashion, suggesting that the peripheral NPY system is a potential mediator of programming for the offspring's vulnerability to obesity and metabolic syndrome.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Intolerancia a la Glucosa/genética , Grasa Intraabdominal/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Neuropéptido Y/metabolismo , Obesidad Abdominal/etiología , Receptores de Neuropéptido Y/biosíntesis , Animales , Dieta Alta en Grasa , Femenino , Lactancia/fisiología , Masculino , Síndrome Metabólico/genética , Ratones , Embarazo , Regulación hacia Arriba
9.
Cancers (Basel) ; 15(13)2023 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-37444423

RESUMEN

Preclinical in vitro and in vivo models remain indispensable tools in cancer research. These classic models, including two- and three-dimensional cell culture techniques and animal models, are crucial for basic and translational studies. However, each model has its own limitations and typically does not fully recapitulate the course of the human disease. Therefore, there is an urgent need for the development of novel, advanced systems that can allow for efficient evaluation of the mechanisms underlying cancer development and progression, more accurately reflect the disease pathophysiology and complexity, and effectively inform therapeutic decisions for patients. Preclinical models are especially important for rare cancers, such as neuroblastoma, where the availability of patient-derived specimens that could be used for potential therapy evaluation and screening is limited. Neuroblastoma modeling is further complicated by the disease heterogeneity. In this review, we present the current status of preclinical models for neuroblastoma research, discuss their development and characteristics emphasizing strengths and limitations, and describe the necessity of the development of novel, more advanced and clinically relevant approaches.

10.
J Cancer Res Clin Oncol ; 149(9): 5803-5822, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36583743

RESUMEN

PURPOSE: Neuropeptide Y (NPY) is a pleiotropic peptide, which is involved in many biological mechanisms important in regulation of cell growth and survival. The aim of this study was a comprehensive analysis of the NPY system in prostate pathology. METHODS: The study was based on immunohistochemical analysis of NPY and its receptors, Y1R, Y2R and Y5R, in tissue samples from benign prostate (BP), primary prostate cancer (PCa) and PCa bone metastases. Tissue microarray (TMA) technique was employed, with analysis of multiple cores from each specimen. Intensity of the immunoreactivity and expression index (EI), as well as distribution of the immunostaining in neoplastic cells and stromal elements were evaluated. Perineural invasion (PNI) and extraprostatic extension (EPE) were areas of special interests. Moreover, a transwell migration assay on the LNCaP PCa cell line was used to assess the chemotactic properties of NPY. RESULTS: Morphological analysis revealed homogeneous membrane and cytoplasmic pattern of NPY staining in cancer cells and its membrane localization with apical accentuation in BP glands. All elements of the NPY system were upregulated in pre-invasive prostate intraepithelial neoplasia, PCa and metastases. EI and staining intensity of NPY receptors were significantly higher in PCa then in BP with correlation between Y2R and Y5R. The strength of expression of the NPY system was further increased in the PNI and EPE areas. In bone metastases, Y1R and Y5R presented high expression scores. CONCLUSION: The results of our study suggest that the NPY system is involved in PCa, starting from early stages of its development to disseminated states of the disease, and participates in the invasion of PCa into the auto and paracrine matter.


Asunto(s)
Neuropéptido Y , Neoplasias de la Próstata , Masculino , Humanos , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/metabolismo , Proliferación Celular
11.
J Biol Chem ; 286(31): 27494-505, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21680731

RESUMEN

Ewing sarcoma family of tumors (ESFT) is a group of aggressive pediatric malignancies driven by the EWS-FLI1 fusion protein, an aberrant transcription factor up-regulating specific target genes, such as neuropeptide Y (NPY) and its Y1 and Y5 receptors (Y5Rs). Previously, we have shown that both exogenous NPY and endogenous NPY stimulate ESFT cell death via its Y1 and Y5Rs. Here, we demonstrate that this effect is prevented by dipeptidyl peptidases (DPPs), which cleave NPY to its shorter form, NPY(3-36), not active at Y1Rs. We have shown that NPY-induced cell death can be abolished by overexpression of DPPs and enhanced by their down-regulation. Both NPY treatment and DPP blockade activated the same cell death pathway mediated by poly(ADP-ribose) polymerase (PARP-1) and apoptosis-inducing factor (AIF). Moreover, the decrease in cell survival induced by DPP inhibition was blocked by Y1 and Y5R antagonists, confirming its dependence on endogenous NPY. Interestingly, similar levels of NPY-driven cell death were achieved by blocking membrane DPPIV and cytosolic DPP8 and DPP9. Thus, this is the first evidence of these intracellular DPPs cleaving releasable peptides, such as NPY, in live cells. In contrast, another membrane DPP, fibroblast activation protein (FAP), did not affect NPY actions. In conclusion, DPPs act as survival factors for ESFT cells and protect them from cell death induced by endogenous NPY. This is the first demonstration that intracellular DPPs are involved in regulation of ESFT growth and may become potential therapeutic targets for these tumors.


Asunto(s)
Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Sarcoma de Ewing/metabolismo , Línea Celular Tumoral , Humanos , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Ewing/enzimología , Sarcoma de Ewing/patología
12.
Nat Commun ; 13(1): 2323, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35484119

RESUMEN

Adverse prognosis in Ewing sarcoma (ES) is associated with the presence of metastases, particularly in bone, tumor hypoxia and chromosomal instability (CIN). Yet, a mechanistic link between these factors remains unknown. We demonstrate that in ES, tumor hypoxia selectively exacerbates bone metastasis. This process is triggered by hypoxia-induced stimulation of the neuropeptide Y (NPY)/Y5 receptor (Y5R) pathway, which leads to RhoA over-activation and cytokinesis failure. These mitotic defects result in the formation of polyploid ES cells, the progeny of which exhibit high CIN, an ability to invade and colonize bone, and a resistance to chemotherapy. Blocking Y5R in hypoxic ES tumors prevents polyploidization and bone metastasis. Our findings provide evidence for the role of the hypoxia-inducible NPY/Y5R/RhoA axis in promoting genomic changes and subsequent osseous dissemination in ES, and suggest that targeting this pathway may prevent CIN and disease progression in ES and other cancers rich in NPY and Y5R.


Asunto(s)
Neoplasias Óseas , Sarcoma de Ewing , Neoplasias Óseas/genética , Inestabilidad Cromosómica , Humanos , Hipoxia , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo , Sarcoma de Ewing/patología , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
13.
Front Cell Dev Biol ; 8: 627090, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33681186

RESUMEN

Neuropeptide Y (NPY) has been implicated in the regulation of cellular motility under various physiological and pathological conditions, including cancer dissemination. Yet, the exact signaling pathways leading to these effects remain unknown. In a pediatric malignancy, neuroblastoma (NB), high NPY release from tumor tissue associates with metastatic disease. Here, we have shown that NPY stimulates NB cell motility and invasiveness and acts as a chemotactic factor for NB cells. We have also identified the Y5 receptor (Y5R) as the main NPY receptor mediating these actions. In NB tissues and cell cultures, Y5R is highly expressed in migratory cells and accumulates in regions of high RhoA activity and dynamic cytoskeleton remodeling. Y5R stimulation activates RhoA and results in Y5R/RhoA-GTP interactions, as shown by pull-down and proximity ligation assays, respectively. This is the first demonstration of the role for the NPY/Y5R axis in RhoA activation and the subsequent cytoskeleton remodeling facilitating cell movement. These findings implicate Y5R as a target in anti-metastatic therapies for NB and other cancers expressing this receptor.

14.
Neuropeptides ; 73: 11-24, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30503694

RESUMEN

Neuropeptide Y (NPY) is a multifunctional neurotransmitter acting via G protein-coupled receptors - Y1R, Y2R and Y5R. NPY activities, such as its proliferative effects, are mediated by multiple receptors, which have the ability to dimerize. However, the role of this receptor interplay in NPY functions remains unclear. The goal of the current study was to identify NPY receptor interactions, focusing on the ligand-binding fraction, and determine their impact on the mitogenic activity of the peptide. Y1R, Y2R and Y5R expressed in CHO-K1 cells formed homodimers detectable on the cell surface by cross-linking. Moreover, Y1R and Y5R heterodimerized, while no Y2R/Y5R heterodimers were detected. Nevertheless, Y5R failed to block internalization of its cognate receptor in both Y1R/Y5R and Y2R/Y5R transfectants, indicating Y5R transactivation upon stimulation of the co-expressed receptor. These receptor interactions correlated with an augmented mitogenic response to NPY. In Y1R/Y5R and Y2R/Y5R transfectants, the proliferative response started at picomolar NPY concentrations, while nanomolar concentrations were needed to trigger proliferation in cells transfected with single receptors. Thus, our data identify direct and indirect heterotypic NPY receptor interactions as the mechanism amplifying its activity. Understanding these processes is crucial for the design of treatments targeting the NPY system.


Asunto(s)
Proliferación Celular/fisiología , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Células CHO , Cricetulus , Multimerización de Proteína/fisiología
15.
Front Oncol ; 13: 1223296, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37434976
16.
J Clin Invest ; 111(12): 1853-62, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12813021

RESUMEN

Previously we showed that neuropeptide Y (NPY), a sympathetic vasoconstrictor neurotransmitter, stimulates endothelial cell migration, proliferation, and differentiation in vitro. Here, we report on NPY's actions, receptors, and mediators in ischemic angiogenesis. In rats, hindlimb ischemia stimulates sympathetic NPY release (attenuated by lumbar sympathectomy) and upregulates NPY-Y2 (Y2) receptor and a peptidase forming Y2/Y5-selective agonist. Exogenous NPY at physiological concentrations also induces Y5 receptor, stimulates neovascularization, and restores ischemic muscle blood flow and performance. NPY-mediated ischemic angiogenesis is not prevented by a selective Y1 receptor antagonist but is reduced in Y2(-/-) mice. Nonischemic muscle vascularity is also lower in Y2(-/-) mice, whereas it is increased in NPY-overexpressing rats compared with their WT controls. Ex vivo, NPY-induced aortic sprouting is markedly reduced in Y2(-/-) aortas and spontaneous sprouting is severely impaired in NPY(-/-) mice. NPY-mediated aortic sprouting, but not cell migration/proliferation, is blocked by an antifetal liver kinase 1 antibody and abolished in mice null for eNOS. Thus, NPY mediates neurogenic ischemic angiogenesis at physiological concentrations by activating Y2/Y5 receptors and eNOS, in part due to release of VEGF. NPY's effectiveness in revascularization and restoring function of ischemic tissue suggests its therapeutic potential in ischemic conditions.


Asunto(s)
Isquemia/tratamiento farmacológico , Músculo Esquelético/irrigación sanguínea , Neovascularización Patológica/inducido químicamente , Neuropéptido Y/farmacología , Neuropéptido Y/fisiología , Animales , Dipeptidil Peptidasa 4/fisiología , Factores de Crecimiento Endotelial/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Isquemia/patología , Isquemia/fisiopatología , Linfocinas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/deficiencia , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/deficiencia , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/fisiología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
17.
Cancer Lett ; 245(1-2): 293-302, 2007 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-16513255

RESUMEN

Neuropeptide Y (NPY) is a sympathetic neurotransmitter recently found to be a potent growth and angiogenic factor. The peptide and its receptors are abundant in neural crest-derived tumors, such as sympathetic neuroblastomas and pheochromocytomas, as well as parasympathetic Ewing's sarcoma family of tumors. NPY regulates their growth directly, by an autocrine activation of tumor cell proliferation or apoptosis, and indirectly, by its angiogenic activity. The overall effect of the peptide on tumor growth depends on a balance between these processes and the type of receptors expressed in the tumor cells. Thus, NPY and its receptors may become targets for the treatment of neural tumors, directed against both tumor cell proliferation and angiogenesis.


Asunto(s)
Neoplasias de Tejido Nervioso/fisiopatología , Cresta Neural/patología , Neuropéptido Y/fisiología , Animales , Proliferación Celular , Humanos , Modelos Biológicos , Neoplasias de Tejido Nervioso/metabolismo , Neoplasias de Tejido Nervioso/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Cresta Neural/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuroblastoma/fisiopatología , Neuropéptido Y/biosíntesis , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Sarcoma de Ewing/fisiopatología
18.
Peptides ; 28(2): 405-12, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17229489

RESUMEN

Neuroblastomas are pediatric tumors of sympathetic origin, expressing neuronal markers, such as NPY and its receptors. Due to this, neuroblastomas are often associated with elevated plasma levels of NPY, which correlates with poor clinical outcome of the disease. This clinical data corroborates the recent discovery of growth-promoting actions of NPY in neuroblastomas. The peptide has been shown to stimulate proliferation of neuroblastoma cells in an autocrine manner and induce tumor vascularization. Since both processes are mediated by the same Y2 and Y5 receptors, targeting this pathway may be a potential bidirectional therapy for these children's tumors.


Asunto(s)
División Celular/fisiología , Neovascularización Patológica , Neuroblastoma/patología , Neuropéptido Y/fisiología , Humanos , Neuroblastoma/irrigación sanguínea
19.
Cancer Res ; 65(5): 1719-28, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15753367

RESUMEN

Neuropeptide Y (NPY) is a sympathetic neurotransmitter recently found to be potently angiogenic and growth promoting for endothelial, vascular smooth muscle and neuronal cells. NPY and its cognate receptors, Y1, Y2 and Y5, are expressed in neural crest-derived tumors; however, their role in regulation of growth is unknown. The effect of NPY on the growth and vascularization of neuroendocrine tumors was tested using three types of cells: neuroblastoma, pheochromocytoma, and Ewing's sarcoma family of tumors (ESFT). The tumors varied in expression of NPY receptors, which was linked to differential functions of the peptide. NPY stimulated proliferation of neuroblastoma cells via Y2/Y5Rs and inhibited ESFT cell growth by Y1/Y5-mediated apoptosis. In both tumor types, NPY receptor antagonists altered basal growth levels, indicating a regulatory role of autocrine NPY. In addition, the peptide released from the tumor cells stimulated endothelial cell proliferation, which suggests its paracrine angiogenic effects. In nude mice xenografts, exogenous NPY stimulated growth of neuroblastoma tumors, whereas it increased apoptosis and reduced growth of ESFT. However, in both tumors, NPY treatment led to an increase in tumor vascularization. Taken together, this is the first report of NPY being a growth-regulatory factor for neuroendocrine tumors, acting both by autocrine activation of tumor cell proliferation or apoptosis and by angiogenesis. NPY and its receptors may become targets for novel approaches in the treatment of these diseases, directed against both tumor cell proliferation and angiogenesis.


Asunto(s)
Apoptosis , Neovascularización Patológica , Neuroblastoma/patología , Neuropéptido Y/metabolismo , Feocromocitoma/patología , Receptores de Neuropéptido Y/fisiología , Animales , Ciclo Celular , Proliferación Celular , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuroblastoma/metabolismo , Feocromocitoma/metabolismo , Ratas , Receptores de Neuropéptido Y/clasificación , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA