Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 31(1): 78-89, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36045587

RESUMEN

Androgen receptor signaling inhibitors (ARSIs) are standard of care for advanced prostate cancer (PCa) patients. Eventual resistance to ARSIs can include the expression of androgen receptor (AR) splice variant, AR-V7, expression as a recognized means of ligand-independent androgen signaling. We demonstrated that interleukin (IL)-6-mediated AR-V7 expression requires bone morphogenic protein (BMP) and CD105 receptor activity in both PCa and associated fibroblasts. Chromatin immunoprecipitation supported CD105-dependent ID1- and E2F-mediated expression of RBM38. Further, RNA immune precipitation demonstrated RBM38 binds the AR-cryptic exon 3 to enable AR-V7 generation. The forced expression of AR-V7 by primary prostatic fibroblasts diminished PCa sensitivity to ARSI. Conversely, downregulation of AR-V7 expression in cancer epithelia and associated fibroblasts was achieved by a CD105-neutralizing antibody, carotuximab. These compelling pre-clinical findings initiated an interventional study in PCa patients developing ARSI resistance. The combination of carotuximab and ARSI (i.e., enzalutamide or abiraterone) provided disease stabilization in four of nine assessable ARSI-refractory patients. Circulating tumor cell evaluation showed AR-V7 downregulation in the responsive subjects on combination treatment and revealed a three-gene panel that was predictive of response. The systemic antagonism of BMP/CD105 signaling can support ARSI re-sensitization in pre-clinical models and subjects that have otherwise developed resistance due to AR-V7 expression.


Asunto(s)
Antagonistas de Receptores Androgénicos , Endoglina , Neoplasias de la Próstata Resistentes a la Castración , Receptores Androgénicos , Humanos , Masculino , Resistencia a Antineoplásicos , Células Neoplásicas Circulantes/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Isoformas de Proteínas , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Proteínas de Unión al ARN , Endoglina/antagonistas & inhibidores , Antagonistas de Receptores Androgénicos/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico
2.
J Med Chem ; 63(18): 10433-10459, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32865411

RESUMEN

The adenosinergic pathway represents an attractive new therapeutic approach in cancer immunotherapy. In this pathway, ecto-5-nucleotidase CD73 has the unique function of regulating production of immunosuppressive adenosine (ADO) through the hydrolysis of AMP. CD73 is overexpressed in many cancers, resulting in elevated levels of ADO that correspond to poor patient prognosis. Therefore, reducing the level of ADO via inhibition of CD73 is a potential strategy for treating cancers. Based on the binding mode of adenosine 5'-(α,ß-methylene)diphosphate (AOPCP) with human CD73, we designed a series of novel monophosphonate small-molecule CD73 inhibitors. Among them, OP-5244 (35) proved to be a highly potent and orally bioavailable CD73 inhibitor. In preclinical studies, 35 completely inhibited ADO production in both human cancer cells and CD8+ T cells. Furthermore, 35 lowered the ratio of ADO/AMP significantly and reversed immunosuppression in mouse models, indicating its potential as an in vivo tool compound for further development.


Asunto(s)
5'-Nucleotidasa/antagonistas & inhibidores , Adenosina/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Factores Inmunológicos/farmacología , Nucleósidos/farmacología , Organofosfonatos/farmacología , Administración Oral , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular Tumoral , Perros , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Proteínas Ligadas a GPI/antagonistas & inhibidores , Humanos , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/síntesis química , Factores Inmunológicos/farmacocinética , Macaca fascicularis , Ratones Endogámicos BALB C , Estructura Molecular , Nucleósidos/administración & dosificación , Nucleósidos/síntesis química , Nucleósidos/farmacocinética , Organofosfonatos/administración & dosificación , Organofosfonatos/síntesis química , Organofosfonatos/farmacocinética , Ratas , Estereoisomerismo , Relación Estructura-Actividad
3.
Proc Natl Acad Sci U S A ; 117(15): 8515-8523, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32238563

RESUMEN

Stromal-epithelial interactions dictate cancer progression and therapeutic response. Prostate cancer (PCa) cells were identified to secrete greater concentration of mitochondrial DNA (mtDNA) compared to noncancer epithelia. Based on the recognized coevolution of cancer-associated fibroblasts (CAF) with tumor progression, we tested the role of cancer-derived mtDNA in a mechanism of paracrine signaling. We found that prostatic CAF expressed DEC205, which was not expressed by normal tissue-associated fibroblasts. DEC205 is a transmembrane protein that bound mtDNA and contributed to pattern recognition by Toll-like receptor 9 (TLR9). Complement C3 was the dominant gene targeted by TLR9-induced NF-κB signaling in CAF. The subsequent maturation complement C3 maturation to anaphylatoxin C3a was dependent on PCa epithelial inhibition of catalase in CAF. In a syngeneic tissue recombination model of PCa and associated fibroblast, the antagonism of the C3a receptor and the fibroblastic knockout of TLR9 similarly resulted in immune suppression with a significant reduction in tumor progression, compared to saline-treated tumors associated with wild-type prostatic fibroblasts. Interestingly, docetaxel, a common therapy for advanced PCa, further promoted mtDNA secretion in cultured epithelia, mice, and PCa patients. The antiapoptotic signaling downstream of anaphylatoxin C3a signaling in tumor cells contributed to docetaxel resistance. The inhibition of C3a receptor sensitized PCa epithelia to docetaxel in a synergistic manner. Tumor models of human PCa epithelia with CAF expanded similarly in mice in the presence or absence of docetaxel. The combination therapy of docetaxel and C3 receptor antagonist disrupted the mtDNA/C3a paracrine loop and restored docetaxel sensitivity.


Asunto(s)
Anafilatoxinas/metabolismo , Fibroblastos Asociados al Cáncer/patología , ADN Mitocondrial/metabolismo , Docetaxel/farmacología , Resistencia a Antineoplásicos , Epitelio/patología , Neoplasias de la Próstata/patología , Animales , Antineoplásicos/farmacología , Apoptosis , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Comunicación Paracrina , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptor Toll-Like 9/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Anticancer Res ; 40(3): 1459-1462, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32132043

RESUMEN

BACKGROUND/AIM: We have previously found elevated levels of endoglin (CD105) in the prostate cancer (PC) tissue of men with poor prognosis, compared to men with indolent disease. Herein, we examined whether plasma levels of the soluble form of CD105 (sCD105) differ according to the PC grade at diagnosis. PATIENTS AND METHODS: We measured sCD105 in 73 subjects with biopsy-confirmed PC at the Durham, North Carolina, Veteran Affairs Health System. The association between sCD105 and intermediate/high-grade PC risk [Gleason Group (GG) 2-5 vs. 1] was examined using regression models. RESULTS: Of 73 men, 27 had low-grade PC and 46 high-grade PC. Higher GG was linked to lower sCD105 (GG1: 6938 pg/ml, GG2-3: 6150 pg/ml, GG4-5: 5554 pg/ml; p=0.012). On multivariable analysis, lower sCD105 was associated with increased high-grade PC risk (ORper 1000 units=1.33, p=0.028). CONCLUSION: Lower sCD105 levels were associated with intermediate and high-risk PC. Further investigation is warranted in a larger PC cohort.


Asunto(s)
Endoglina/sangre , Neoplasias de la Próstata/sangre , Anciano , Biomarcadores de Tumor/sangre , Estudios de Cohortes , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Neoplasias de la Próstata/patología
5.
Endocr Relat Cancer ; 27(1): 1-9, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31648185

RESUMEN

While the overall 5-year survival rate for prostate cancer is near 100%, up to 35% of patients will develop recurrent disease. At the time of prostatectomy, prostate-specific antigen (PSA) is used to guide primary therapy with the goal of curative intervention. It can be valuable to know when primary therapy may not in fact be curative, so that subsequent adjuvant therapy can be administered at an early stage to limit progression. We examined prostate cancer patients with PSA ≤10 ng/mL that were all subjected to prostatectomy with at least 5 years of follow-up (n = 181). Based on data that endoglin (CD105) signaling in the tumor can contribute to prostate cancer progression, we examined the expression of soluble CD105 (sCD105) in the patient plasma. To determine the relation of plasma sCD105 measures to cellular CD105 in tissues, we tested an independent set of prostate cancer tissues and paired plasma (n = 31). Elevated sCD105 was found to be associated with recurrence-free survival of prostate cancer patients. Further, sCD105 levels in patient plasma were inversely correlated with cellular CD105 expression. This translational study supported preclinical data demonstrating the pro-tumorigenic capacity of cellular CD105 and provide a blood-based biomarker, sCD105, for prostate cancer recurrence in prostatectomy patients with PSA levels ≤10 ng/mL.


Asunto(s)
Endoglina/sangre , Recurrencia Local de Neoplasia/sangre , Neoplasias de la Próstata/sangre , Biomarcadores de Tumor/sangre , Progresión de la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Estadificación de Neoplasias , Prostatectomía/métodos , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Tasa de Supervivencia
6.
Oncogene ; 38(5): 716-730, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30177832

RESUMEN

Heterogeneous prostatic carcinoma-associated fibroblasts (CAF) contribute to tumor progression and resistance to androgen signaling deprivation therapy (ADT). CAF subjected to extended passaging, compared to low passage CAF, were found to lose tumor expansion potential and heterogeneity. Cell surface endoglin (CD105), known to be expressed on proliferative endothelia and mesenchymal stem cells, was diminished in high passage CAF. RNA-sequencing revealed SFRP1 to be distinctly expressed by tumor-inductive CAF, which was further demonstrated to occur in a CD105-dependent manner. Moreover, ADT resulted in further expansion of the CD105+ fibroblastic population and downstream SFRP1 in 3-dimensional cultures and patient-derived xenograft tissues. In patients, CD105+ fibroblasts were found to circumscribe epithelia with neuroendocrine differentiation. CAF-derived SFRP1, driven by CD105 signaling, was necessary and sufficient to induce prostate cancer neuroendocrine differentiation in a paracrine manner. A partially humanized CD105 neutralizing antibody, TRC105, inhibited fibroblastic SFRP1 expression and epithelial neuroendocrine differentiation. In a novel synthetic lethality paradigm, we found that simultaneously targeting the epithelia and its microenvironment with ADT and TRC105, respectively, reduced castrate-resistant tumor progression, in a model where either ADT or TRC105 alone had little effect.


Asunto(s)
Diferenciación Celular , Endoglina/metabolismo , Fibroblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Células Neuroendocrinas/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal , Línea Celular Tumoral , Endoglina/genética , Fibroblastos/patología , Humanos , Masculino , Proteínas de Neoplasias/genética , Células Neuroendocrinas/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología
7.
J Clin Invest ; 128(10): 4472-4484, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30047926

RESUMEN

Prostate cancer is an androgen-dependent disease subject to interactions between the tumor epithelium and its microenvironment. Here, we found that epigenetic changes in prostatic cancer-associated fibroblasts (CAF) initiated a cascade of stromal-epithelial interactions. This facilitated lethal prostate cancer growth and development of resistance to androgen signaling deprivation therapy (ADT). We identified a Ras inhibitor, RASAL3, as epigenetically silenced in human prostatic CAF, leading to oncogenic Ras activity driving macropinocytosis-mediated glutamine synthesis. Interestingly, ADT further promoted RASAL3 epigenetic silencing and glutamine secretion by prostatic fibroblasts. In an orthotopic xenograft model, subsequent inhibition of macropinocytosis and glutamine transport resulted in antitumor effects. Stromal glutamine served as a source of energy through anaplerosis and as a mediator of neuroendocrine differentiation for prostate adenocarcinoma. Antagonizing the uptake of glutamine restored sensitivity to ADT in a castration-resistant xenograft model. In validating these findings, we found that prostate cancer patients on ADT with therapeutic resistance had elevated blood glutamine levels compared with those with therapeutically responsive disease (odds ratio = 7.451, P = 0.02). Identification of epigenetic regulation of Ras activity in prostatic CAF revealed RASAL3 as a sensor for metabolic and neuroendocrine reprogramming in prostate cancer patients failing ADT.


Asunto(s)
Silenciador del Gen , Glutamina/metabolismo , Proteínas de Neoplasias/metabolismo , Tumores Neuroendocrinos/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Fibroblastos/metabolismo , Fibroblastos/patología , Glutamina/genética , Humanos , Masculino , Ratones , Proteínas de Neoplasias/genética , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Activadoras de ras GTPasa/genética
8.
Oncogene ; 37(32): 4385-4397, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29717261

RESUMEN

Radiation therapy is the primary intervention for nearly half of the patients with localized advanced prostate cancer and standard of care for recurrent disease following surgery. The development of radiation-resistant disease is an obstacle for nearly 30-50% of patients undergoing radiotherapy. A better understanding of mechanisms that lead to radiation resistance could aid in the development of sensitizing agents to improve outcome. Here we identified a radiation-resistance pathway mediated by CD105, downstream of BMP and TGF-ß signaling. Antagonizing CD105-dependent BMP signaling with a partially humanized monoclonal antibody, TRC105, resulted in a significant reduction in clonogenicity when combined with irradiation. In trying to better understand the mechanism for the radio-sensitization, we found that radiation-induced CD105/BMP signaling was sufficient and necessary for the upregulation of sirtuin 1 (SIRT1) in contributing to p53 stabilization and PGC-1α activation. Combining TRC105 with irradiation delayed DNA damage repair compared to irradiation alone. However, in the absence of p53 function, combining TRC105 and radiation resulted in no reduction in clonogenicity compared to radiation alone, despite similar reduction of DNA damage repair observed in p53-intact cells. This suggested DNA damage repair was not the sole determinant of CD105 radio-resistance. As cancer cells undergo an energy deficit following irradiation, due to the demands of DNA and organelle repair, we examined SIRT1's role on p53 and PGC-1α with respect to glycolysis and mitochondrial biogenesis, respectively. Consequently, blocking the CD105-SIRT1 axis was found to deplete the ATP stores of irradiated cells and cause G2 cell cycle arrest. Xenograft models supported these findings that combining TRC105 with irradiation significantly reduces tumor size over irradiation alone (p value = 10-9). We identified a novel synthetic lethality strategy of combining radiation and CD105 targeting to address the DNA repair and metabolic addiction induced by irradiation in p53-functional prostate cancers.


Asunto(s)
Endoglina/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Tolerancia a Radiación/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Fase G2/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Próstata/efectos de los fármacos , Próstata/metabolismo , Próstata/efectos de la radiación , Neoplasias de la Próstata/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
9.
Sci Rep ; 6: 39257, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27995963

RESUMEN

Hemorrhagic cystitis is an inflammatory and ulcerative bladder condition associated with systemic chemotherapeutics, like cyclophosphomide. Earlier, we reported reactive oxygen species resulting from cyclophosphamide metabolite, acrolein, causes global methylation followed by silencing of DNA damage repair genes. Ogg1 (8-oxoguanine DNA glycosylase) is one such silenced base excision repair enzyme that can restore DNA integrity. The accumulation of DNA damage results in subsequent inflammation associated with pyroptotic death of bladder smooth muscle cells. We hypothesized that reversing inflammasome-induced imprinting in the bladder smooth muscle could prevent the inflammatory phenotype. Elevated recruitment of Dnmt1 and Dnmt3b to the Ogg1 promoter in acrolein treated bladder muscle cells was validated by the pattern of CpG methylation revealed by bisulfite sequencing. Knockout of Ogg1 in detrusor cells resulted in accumulation of reactive oxygen mediated 8-Oxo-dG and spontaneous pyroptotic signaling. Histone deacetylase (HDAC) inhibitor, suberoylanilide hydroxamic acid (SAHA), restored Ogg1 expression in cells treated with acrolein and mice treated with cyclophosphamide superior to the standard of care, mesna or nicotinamide-induced DNA demethylation. SAHA restored cyclophosphamide-induced bladder pathology to that of untreated control mice. The observed epigenetic imprinting induced by inflammation suggests a new therapeutic target for the treatment of hemorrhagic cystitis.


Asunto(s)
Ciclofosfamida/toxicidad , Cistitis/etiología , ADN Glicosilasas/metabolismo , Reparación del ADN/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Acroleína/toxicidad , Animales , Células Cultivadas , Cistitis/inducido químicamente , Cistitis/metabolismo , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Daño del ADN/efectos de los fármacos , ADN Glicosilasas/deficiencia , ADN Glicosilasas/genética , Regulación hacia Abajo/efectos de los fármacos , Femenino , Mesna/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Niacinamida/farmacología , Especies Reactivas de Oxígeno/metabolismo , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Vorinostat , ADN Metiltransferasa 3B
10.
Biochim Biophys Acta ; 1853(5): 1087-95, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25721765

RESUMEN

Epidermal growth factor (EGF) receptor (EGFR) is the founding member of the ErbB family of growth factor receptors that modulate a complex network of intracellular signaling pathways controlling growth, proliferation, differentiation, and motility. Selenoprotein W (SEPW1) is a highly conserved, diet-regulated 9kDa thioredoxin-like protein required for normal cell cycle progression. We report here that SEPW1 is required for EGF-induced EGFR activation and that it functions by suppressing EGFR ubiquitination and receptor degradation. SEPW1 depletion inhibited EGF-dependent cell cycle entry in breast and prostate epithelial cells. In prostate cells, SEPW1 depletion decreased EGFR auto-phosphorylation, while SEPW1 overexpression increased EGFR auto-phosphorylation. SEPW1 depletion increased the rate of EGFR degradation, which decreased total and surface EGFR and suppressed EGF-dependent EGFR endocytosis, EGFR dimer formation, and activation of EGF-dependent pathways. EGFR ubiquitination was increased in SEPW1-depleted cells--in agreement with the increased rate of EGFR degradation, and suggests that SEPW1 suppresses EGFR ubiquitination. Ubiquitination-directed lysozomal degradation controls post-translational EGFR expression and is dysregulated in many cancers. Thus, suppression of EGFR ubiquitination by SEPW1 may be related to the putative increase in cancer risk associated with high selenium intakes. Knowledge of the mechanisms underlying SEPW1's regulation of EGFR ubiquitination may reveal new opportunities for nutritional cancer prevention or cancer drug development.


Asunto(s)
Membrana Celular/metabolismo , Receptores ErbB/metabolismo , Proteolisis , Selenoproteína W/metabolismo , Ubiquitinación , Mama/citología , Ciclo Celular/efectos de los fármacos , Línea Celular , Membrana Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Masculino , Fosforilación/efectos de los fármacos , Próstata/citología , Multimerización de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Ubiquitinación/efectos de los fármacos
11.
Metab Syndr Relat Disord ; 12(4): 227-30, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24593134

RESUMEN

BACKGROUND: There is much data supporting a role for adipokines in both obesity and metabolic syndrome. Insulin resistance and low-grade inflammation are crucial in the genesis of both disorders. Although data suggest that the ratio of leptin/adiponectin correlates with insulin resistance and predicts cardiovascular disease (CVD), there is scanty data on the relationship between the retinol-binding protein-4 (RBP4)/adiponectin ratio with insulin resistance and inflammation. We tested the relationship of both these ratios with measures of insulin resistance and inflammation. METHODS: In 72 individuals, including controls and patients with metabolic syndrome, we calculated the homeostasis model assessment of insulin resistance (HOMA-IR) and assayed high-sensitivity C-reactive protein (hsCRP) and the adipokines, adiponectin, leptin, and RBP4. RESULTS: Whereas both the leptin/adiponectin and RBP4/adiponectin ratios did not correlate with HOMA-IR, both correlated significantly with the prototypic biomarker of inflammation, hsCRP. Also in patients with metabolic syndrome following adjustment for adiposity, only the RBP4/adiponectin ratio was significantly increased. CONCLUSIONS: Hence it appears that whereas both the leptin/adiponectin and RBP4/adiponectin ratios correlate with inflammation, only the RBP4/adiponectin ratio was significantly increased in metabolic syndrome and would be more useful to predict CVD, especially in metabolic syndrome.


Asunto(s)
Adiponectina/sangre , Resistencia a la Insulina , Leptina/sangre , Síndrome Metabólico/sangre , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Adiponectina/metabolismo , Adulto , Anciano , Biomarcadores/metabolismo , Índice de Masa Corporal , Proteína C-Reactiva/metabolismo , Femenino , Humanos , Inflamación , Leptina/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...