Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Respir Res ; 24(1): 52, 2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36788537

RESUMEN

INTRODUCTION: Lung cancer remains the leading cause of death from cancer, worldwide. Developing early detection diagnostic methods, especially non-invasive methods, is a critical component to raising the overall survival rate and prognosis for lung cancer. The purpose of this study is to evaluate two protocols of a novel in vitro cellular immune response test to detect lung cancer. The test specifically quantifies the glycolysis metabolism pathway, which is a biomarker for the activation level of immune cells. It summarizes the results of two clinical trials, where each deploys a different protocol's version of this test for the detection of lung cancer. In the later clinical trial, an improved test protocol is applied. METHOD: The test platform is based on changes in the metabolic pathways of the immune cells following their activation by antigenic stimuli associated with Lung cancer. Peripheral Blood Mononuclear Cells are loaded on a multiwell plate together with various lung tumor associated antigens and a fluorescent probe that exhibits a pH-dependent absorption shift. The acidification process in the extracellular fluid is monitored by a commercial fluorescence plate reader device in continuous reading for 3 h at 37 °C to document the fluorescent signal received from each well. RESULTS: In the later clinical trial, an improved test protocol was applied and resulted in increased test accuracy. Specificity of the test increased to 94.0% and test sensitivity increased to 97.3% in lung cancer stage I, by using the improved protocol. CONCLUSION: The improved protocol of the novel cellular immune metabolic response based test detects stage I and stage II of lung cancer with high specificity and sensitivity, with low material costs and fast results.


Asunto(s)
Leucocitos Mononucleares , Neoplasias Pulmonares , Humanos , Inmunidad Celular , Biopsia Líquida , Neoplasias Pulmonares/diagnóstico , Pronóstico
2.
Handb Exp Pharmacol ; 233: 355-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25903410

RESUMEN

Obesity is associated with multiple metabolic disorders that drive cardiovascular disease, T2D and cancer. The doubling in the number of obese adults over the past 3 decades led to the recognition of obesity as a "disease". With over 42 million children obese or overweight, this epidemic is rapidly growing worldwide. Obesity and T2D are both associated together and independently with an increased risk for cancer and a worse prognosis. Accumulating evidence from epidemiological studies revealed potential factors that may explain the association between obesity-linked metabolic disorders and cancer risk. Studies based on the insulin resistance MKR mice, highlighted the roe of the insulin receptor and its downstream signaling proteins in mediating hyperinsulinemia's mitogenic effects. Hypercholesterolemia was also shown to promote the formation of larger tumors and enhancement in metastasis. Furthermore, the conversion of cholesterol into 27-Hydroxycholesterol was found to link high fat diet-induced hypercholesterolemia with cancer pathophysiology. Alteration in circulating adipokines and cytokines are commonly found in obesity and T2D. Adipokines are involved in tumor growth through multiple mechanisms including mTOR, VEGF and cyclins. In addition, adipose tissues are known to recruit and alter macrophage phenotype; these macrophages can promote cancer progression by secreting inflammatory cytokines such as TNF-α and IL-6. Better characterization on the above factors and their downstream effects is required in order to translate the current knowledge into the clinic, but more importantly is to understand which are the key factors that drive cancer in each patient. Until we reach this point, policies and activities toward healthy diets and physical activities remain the best medicine.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Síndrome Metabólico/complicaciones , Neoplasias/etiología , Adipoquinas/fisiología , Animales , Citocinas/fisiología , Diabetes Mellitus Tipo 2/epidemiología , Modelos Animales de Enfermedad , Humanos , Obesidad/complicaciones , Obesidad/epidemiología
3.
Breast Cancer Res ; 17: 78, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26040280

RESUMEN

INTRODUCTION: Breast tumors are comprised of distinct cancer cell populations which differ in their tumorigenic and metastatic capacity. Characterization of cell surface markers enables investigators to distinguish between cancer stem cells and their counterparts. CD24 is a well-known cell surface marker for mammary epithelial cells isolation, recently it was suggested as a potential prognostic marker in a wide variety of malignancies. Here, we demonstrate that CD24(+) cells create intra-tumor heterogeneity, and display highly metastatic properties. METHODS: The mammary carcinoma Mvt1 cells were sorted into CD24(-) and CD24(+) cells. Both subsets were morphologically and phenotypically characterized, and tumorigenic capacity was assessed via orthotopic inoculation of each subset into the mammary fat pad of wild-type and MKR mice. The metastatic capacity of each subset was determined with the tail vein metastasis assay. The role of CD24 in tumorigenesis was further examined with shRNA technology. GFP-labeled cells were monitored in vivo for differentiation. The genetic profile of each subset was analyzed using RNA sequencing. RESULTS: CD24(+) cells displayed a more spindle-like cytoplasm. The cells formed mammospheres in high efficiency and CD24(+) tumors displayed rapid growth in both WT and MKR mice, and were more metastatic than CD24- cells. Interestingly, CD24-KD in CD24+ cells had no effect both in vitro and in vivo on the various parameters studied. Moreover, CD24(+) cells gave rise in vivo to the CD24(-) that comprised the bulk of the tumor. RNA-seq analysis revealed enrichment of genes and pathways of the extracellular matrix in the CD24(+) cells. CONCLUSION: CD24(+) cells account for heterogeneity in mammary tumors. CD24 expression at early stages of the cancer process is an indication of a highly invasive tumor. However, CD24 is not a suitable therapeutic target; instead we suggest here new potential targets accounting for early differentiated cancer cells tumorigenic capacity.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Antígeno CD24/metabolismo , Animales , Biomarcadores , Neoplasias de la Mama/genética , Antígeno CD24/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Inmunofenotipificación , Ratones , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Fenotipo , Carga Tumoral
4.
Diabetes Metab Res Rev ; 31(5): 492-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25529355

RESUMEN

BACKGROUND: Type 1 diabetes is an autoimmune disease, characterized by a loss of pancreatic ß-cell mass and function, which results in dramatic reductions in insulin secretion and circulating insulin levels. Patients with type 1 diabetes are traditionally treated with insulin injections and insulin pumps ex vivo or undergo transplantation. Growth hormone (GH) has been shown to be involved in ß-cell function and survival in culture. METHODS: Twelve-week-old female C57BL/6 mice were treated with streptozotocin and monitored for their weight and blood glucose levels. Fourteen days post-initial injection, these mice were separated into two groups at random. One group was treated with GH while the other treated with vehicle for up to 3 weeks. These mice were compared with mice not treated with streptozotocin. RESULTS: Under our experimental conditions, we observed that mice treated with GH had larger islets and higher serum insulin levels than streptozotocin-treated mice treated with saline (0.288 vs. 0.073 ng/mL, p < 0.01). CONCLUSIONS: Our data demonstrate that GH may rescue islets and therefore may possess therapeutic potential in the treatment of type 1 diabetes, although consideration should be made regarding GH's effect on insulin resistance.


Asunto(s)
Diabetes Mellitus Experimental/sangre , Hormona de Crecimiento Humana/farmacología , Insulina/sangre , Islotes Pancreáticos/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/patología , Femenino , Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Proteínas Recombinantes
5.
J Endocrinol ; 227(3): 143-151, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26383532

RESUMEN

Obesity and type 2 diabetes (T2D) are associated with an increased risk of breast cancer incidence and mortality. Common features of obesity and T2D are insulin resistance and hyperinsulinemia. A mammary tumor promoting effect of insulin resistance and hyperinsulinemia was demonstrated in the transgenic female MKR mouse model of pre-diabetes inoculated with mammary cancer cells. Interestingly, in MKR mice, as well as in other diabetic mouse models, males exhibit severe hyperglycemia, while females display insulin resistance and hyperinsulinemia with only a mild increase in blood glucose levels. This gender-specific protection from hyperglycemia may be attributed to estradiol, a key player in the regulation of the metabolic state, including obesity, glucose homeostasis, insulin resistance, and lipid profile. The aim of this study was to investigate the effects of ovariectomy (including the removal of endogenous estradiol) on the metabolic state of MKR female mice and subsequently on the growth of Mvt-1 mammary cancer cells, inoculated into the mammary fat pad of ovariectomized mice, compared with sham-operated mice. The results showed an increase in body weight, accompanied by increased fat mass, elevated blood glucose levels, and hypercholesterolemia, in ovariectomized MKR mice. In addition, mammary tumor growth was significantly higher in these mice. The results suggest that ovarian hormone deficiency may promote impaired metabolic homeostasis in the hyperinsulinemic MKR female mice, which in turn is associated with an increased growth of mammary tumors.


Asunto(s)
Neoplasias Mamarias Experimentales/etiología , Neoplasias Mamarias Experimentales/patología , Ovariectomía/efectos adversos , Estado Prediabético/patología , Animales , Glucemia/metabolismo , Femenino , Resistencia a la Insulina , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Estado Prediabético/genética
6.
Mol Cell Endocrinol ; 374(1-2): 101-7, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23643895

RESUMEN

Hyperlipidemia and hypercholesterolemia have been found to be important factors in cancer development and metastasis. However, the metabolic mechanism and downstream cellular processes following cholesterol stimulation are still unknown. Here we tested the effect of cholesterol on MC-38 colon cancer cells. Using Illumina gene array technology we found a number of genes that were differentially expressed following short term (20-40 min) and longer term (between 2 and 5h) cholesterol stimulation. Three genes were consistently increased at these time points; c-Jun, Jun-B and the chemokine CXCL-1. We have previously shown that cholesterol stimulation leads to PI3K/Akt phosphorylation, and now demonstrated that cholesterol inhibits ERK1/2 phosphorylation; both effects reversed when cholesterol is depleted from lipid rafts using methyl-ß-cyclodextrin (MBCD). In addition, vanadate, an inhibitor of phosphatases, reversed the cholesterol inhibition of ERK1/2 phosphorylation. Specific inhibition of p-Akt by wortmannin did not affect cholesterol's stimulation of the expression of c-Jun and Jun-B, however the vanadate effect of increasing p-ERK1/2, inhibited c-Jun expression, specifically, and the MBCD effect of increasing p-ERK and inhibiting p-Akt reduced c-Jun expression. In contrast MBCD and vanadate both enhanced Jun-B gene expression in the presence of cholesterol and elevation of ERK phosphorylation. Thus there is apparently, a differential signaling pathway whereby cholesterol enhances gene expression of the Jun family members.


Asunto(s)
Colesterol/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Microdominios de Membrana/metabolismo , Transducción de Señal/efectos de los fármacos , Androstadienos/farmacología , Animales , Línea Celular Tumoral , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Colesterol/metabolismo , Colon/metabolismo , Colon/patología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/genética , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vanadatos/farmacología , Wortmanina , beta-Ciclodextrinas/farmacología
7.
Vitam Horm ; 93: 51-98, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23810003

RESUMEN

The worldwide epidemic of obesity is associated with increasing rates of the metabolic syndrome and type 2 diabetes. Epidemiological studies have reported that these conditions are linked to increased rates of cancer incidence and mortality. Obesity, particularly abdominal obesity, is associated with insulin resistance and the development of dyslipidemia, hyperglycemia, and ultimately type 2 diabetes. Although many metabolic abnormalities occur with obesity and type 2 diabetes, insulin resistance and hyperinsulinemia appear to be central to these conditions and may contribute to dyslipidemia and altered levels of circulating estrogens and androgens. In this review, we will discuss the epidemiological and molecular links between obesity, type 2 diabetes, and cancer, and how hyperinsulinemia and dyslipidemia may contribute to cancer development. We will discuss how these metabolic abnormalities may interact with estrogen signaling in breast cancer growth. Finally, we will discuss the effects of type 2 diabetes medications on cancer risk.


Asunto(s)
Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/metabolismo , Neoplasias/epidemiología , Neoplasias/metabolismo , Obesidad/epidemiología , Obesidad/metabolismo , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/etiología , Humanos , Neoplasias/complicaciones , Neoplasias/etiología , Obesidad/complicaciones , Obesidad/fisiopatología
9.
Rambam Maimonides Med J ; 2(2): e0043, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23908801

RESUMEN

Patients with type 2 diabetes (T2D) are at increased risk of developing cancer. This evidence arises from numerous epidemiologic studies that relate a positive association between T2D and cancer. In-vitro and several in-vivo experiments have attempted to discern the potential mechanistic factors involved in this relationship. Candidates include hyperinsulinemia, insulin-like growth factor-1 (IGF-1), and insulin-like growth factor-2 (IGF-2) signaling. These studies demonstrated that increased insulin, IGF-1, and IGF-2 signaling through the insulin receptor and IGF-1 receptor can induce cancer development and progression.

10.
J Biol Chem ; 284(5): 3037-3048, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19056736

RESUMEN

GATA3 is a critical transcription factor for many developmental processes. During T helper (Th) cell differentiation, GATA3 induces the Th2 and suppresses the Th1 pathway. Stimulation of the T cell receptor (TCR) of naive Th cells in the presence of interleukin 4 (IL-4) induces robust expression of GATA3; however, it is unclear where these signals integrate. Gata3 encodes two transcripts that differ in their alternative, untranslated first exons. We show here the involvement of the TCR-inducible transcription factor NFAT1 in the transcriptional regulation of both Gata3 transcripts following TCR stimulation of naive and differentiated Th2 cells. We also show that IL-4 is important for the initiation and establishment of Gata3 transcription in developing Th2 cells, especially from the distal promoter. The early function of IL-4 can be STAT6 dependent or independent. However, the establishment of the activity of the distal promoter is totally dependent on STAT6, whereas it is likely that the proximal promoter has additional activation mechanisms that are STAT6 independent. Our findings suggest that different combinations of transcription factors downstream of the IL-4 receptor (IL-4R) and TCR finely modulate Gata3 gene expression from its two promoters for optimal Th2 differentiation.


Asunto(s)
Factor de Transcripción GATA3/genética , Regulación de la Expresión Génica , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-4/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Calcineurina/metabolismo , Inmunoprecipitación de Cromatina , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Factores de Transcripción NFATC/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , ARN Mensajero/genética , Factor de Transcripción STAT6/metabolismo , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA