Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci Alliance ; 7(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38458648

RESUMEN

Plexiform neurofibromas (PNFs) are nerve tumors caused by loss of NF1 and dysregulation of RAS-MAPK signaling in Schwann cells. Most PNFs shrink in response to MEK inhibition, but targets with increased and durable effects are needed. We identified the anaphylatoxin C5a as increased in PNFs and expressed largely by PNF m acrophages. We defined pharmacokinetic and immunomodulatory properties of a C5aR1/2 antagonist and tested if peptide antagonists augment the effects of MEK inhibition. MEK inhibition recruited C5AR1 to the macrophage surface; short-term inhibition of C5aR elevated macrophage apoptosis and Schwann cell death, without affecting MEK-induced tumor shrinkage. PNF macrophages lacking C5aR1 increased the engulfment of dying Schwann cells, allowing their visualization. Halting combination therapy resulted in altered T-cell distribution, elevated Iba1+ and CD169+ immunoreactivity, and profoundly altered cytokine expression, but not sustained trumor shrinkage. Thus, C5aRA inhibition independently induces macrophage cell death and causes sustained and durable effects on the PNF microenvironment.


Asunto(s)
Citofagocitosis , Neurofibroma Plexiforme , Humanos , Macrófagos/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos , Neurofibroma Plexiforme/patología , Transducción de Señal , Microambiente Tumoral
2.
Proteomics ; 22(1-2): e2100063, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34648240

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive, nerve-associated tumors and the main cause of death amongst neurofibromatosis type I (NF1) patients. Schwann cells (SCs) are the pathogenic cell type in MPNST, however the secretome of human MPNST -derived SCs is poorly defined. In this study, a comprehensive proteomic analysis of the proteins secreted by the sNF96.2 human SC line, derived from a patient with MPNST, was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of 17,354 unique peptides corresponding to 1538 individual proteins were identified. Among them, 995 proteins were confirmed as secreted using various bioinformatics tools including SignalP, SecretomeP, Vertebrate Secretome Database (VerSeDa), and Ingenuity Pathway Analysis (IPA). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were conducted to assign protein localization and function, and to define enriched pathways. Protein binding was the most enriched molecular function, and the most enriched biological process was cell-cell adhesion. Metabolic pathways showed the highest levels of enrichment. In addition, 13 of the identified proteins were validated in Western blotting. This comprehensive secretome map constitutes a reference library providing a new molecular insight into MPNST.


Asunto(s)
Neoplasias de la Vaina del Nervio , Neurofibrosarcoma , Línea Celular Tumoral , Cromatografía Liquida , Humanos , Proteómica , Células de Schwann , Secretoma , Espectrometría de Masas en Tándem
3.
Oncogene ; 40(24): 4229-4241, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34079083

RESUMEN

Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft-tissue sarcomas that cause significant mortality in adults with neurofibromatosis type 1. We compared gene expression of growth factors in normal human nerves to MPNST and normal human Schwann cells to MPNST cell lines. We identified WNT5A as the most significantly upregulated ligand-coding gene and verified its protein expression in MPNST cell lines and tumors. In many contexts WNT5A acts as an oncogene. However, inhibiting WNT5A expression using shRNA did not alter MPNST cell proliferation, invasion, migration, or survival in vitro. Rather, shWNT5A-treated MPNST cells upregulated mRNAs associated with the remodeling of extracellular matrix and with immune cell communication. In addition, these cells secreted increased amounts of the proinflammatory cytokines CXCL1, CCL2, IL6, CXCL8, and ICAM1. Versus controls, shWNT5A-expressing MPNST cells formed larger tumors in vivo. Grafted tumors contained elevated macrophage/stromal cells, larger and more numerous blood vessels, and increased levels of Mmp9, Cxcl13, Lipocalin-1, and Ccl12. In some MPNST settings, these effects were mimicked by targeting the WNT5A receptor ROR2. These data suggest that the non-canonical Wnt ligand WNT5A inhibits MPNST tumor formation by modulating the MPNST microenvironment, so that blocking WNT5A accelerates tumor growth in vivo.


Asunto(s)
Proliferación Celular/genética , Neoplasias de la Vaina del Nervio/genética , Microambiente Tumoral/genética , Proteína Wnt-5a/genética , Línea Celular Tumoral , Movimiento Celular/genética , Matriz Extracelular/genética , Humanos , Neoplasias de la Vaina del Nervio/patología , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibrosarcoma/genética , Neurofibrosarcoma/patología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Células de Schwann/patología
4.
Neurooncol Adv ; 2(Suppl 1): i23-i32, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32642730

RESUMEN

Plexiform neurofibromas (PNF) are peripheral nerve tumors caused by bi-allelic loss of NF1 in the Schwann cell (SC) lineage. PNF are common in individuals with Neurofibromatosis type I (NF1) and can cause significant patient morbidity, spurring research into potential therapies. Immune cells are rare in peripheral nerve, whereas in PNF 30% of the cells are monocytes/macrophages. Mast cells, T cells, and dendritic cells (DCs) are also present. NF1 mutant neurofibroma SCs with elevated Ras-GTP signaling resemble injury-induced repair SCs, in producing growth factors and cytokines not normally present in SCs. This provides a cytokine-rich environment facilitating PNF immune cell recruitment and fibrosis. We propose a model based on genetic and pharmacologic evidence in which, after loss of Nf1 in the SC lineage, a lag occurs. Then, mast cells and macrophages are recruited to nerve. Later, T cell/DC recruitment through CXCL10/CXCR3 drives neurofibroma initiation and sustains PNF macrophages and tumor growth. Stat3 signaling is an additional critical mediator of neurofibroma initiation, cytokine production, and PNF growth. At each stage of PNF development therapeutic benefit should be achievable through pharmacologic modulation of leukocyte recruitment and function.

5.
Urol Oncol ; 38(4): 150-173, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31937423

RESUMEN

Prostate cancer is the most common solid organ cancer in men, and the second most common cause of male cancer-related mortality. It has few effective therapies, and is difficult to diagnose accurately. Prostate-specific antigen (PSA), which is currently the most effective diagnostic tool available, cannot reliably discriminate between different pathologies, and in fact only around 30% of patients found to have elevated levels of PSA are subsequently confirmed to actually have prostate cancer. As such, there is a desperate need for more reliable diagnostic tools that will allow the early detection of prostate cancer so that the appropriate interventions can be applied. Nuclear magnetic resonance (NMR) spectroscopy and magnetic resonance spectroscopy (MRS) are 2 high throughput, noninvasive analytical procedures that have the potential to enable differentiation of prostate cancer from other pathologies using metabolomics, by focusing specifically on certain metabolites which are associated with the development of prostate cancer cells and its progression. The value that this type of approach has for the early detection, diagnosis, prognosis, and personalized treatment of prostate cancer is becoming increasingly apparent. Recent years have seen many promising developments in the fields of NMR spectroscopy and MRS, with improvements having been made to hardware as well as to techniques associated with the acquisition, processing, and analysis of related data. This review focuses firstly on proton NMR spectroscopy of blood serum, urine, and expressed prostatic secretions in vitro, and then on 1- and 2-dimensional proton MRS of the prostate in vivo. Major advances in these fields and methodological principles of data collection, acquisition, processing, and analysis are described along with some discussion of related challenges, before prospects that proton MRS has for future improvements to the clinical management of prostate cancer are considered.


Asunto(s)
Líquidos Corporales/diagnóstico por imagen , Espectroscopía de Resonancia Magnética/métodos , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/diagnóstico , Humanos , Masculino , Neoplasias de la Próstata/terapia
6.
JCI Insight ; 4(3)2019 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-30728335

RESUMEN

Plexiform neurofibroma is a major contributor to morbidity in patients with neurofibromatosis type I (NF1). Macrophages and mast cells infiltrate neurofibroma, and data from mouse models implicate these leukocytes in neurofibroma development. Antiinflammatory therapy targeting these cell populations has been suggested as a means to prevent neurofibroma development. Here, we compare gene expression in Nf1-mutant nerves, which invariably form neurofibroma, and show disruption of neuron-glial cell interactions and immune cell infiltration to mouse models, which rarely progresses to neurofibroma with or without disruption of neuron-glial cell interactions. We find that the chemokine Cxcl10 is uniquely upregulated in NF1 mice that invariably develop neurofibroma. Global deletion of the CXCL10 receptor Cxcr3 prevented neurofibroma development in these neurofibroma-prone mice, and an anti-Cxcr3 antibody somewhat reduced tumor numbers. Cxcr3 expression localized to T cells and DCs in both inflamed nerves and neurofibromas, and Cxcr3 expression was necessary to sustain elevated macrophage numbers in Nf1-mutant nerves. To our knowledge, these data support a heretofore-unappreciated role for T cells and DCs in neurofibroma initiation.

7.
Oncotarget ; 7(19): 28488-97, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27074571

RESUMEN

The precursor for nerve growth factor (proNGF) is expressed in some cancers but its clinicopathological significance is unclear. The present study aimed to define the clinicopathological significance of proNGF in thyroid cancer. ProNGF expression was analysed by immunohistochemistry in two cohorts of cancer versus benign tumors (adenoma) and normal thyroid tissues. In the first cohort (40 thyroid cancers, 40 thyroid adenomas and 80 normal thyroid tissues), proNGF was found overexpressed in cancers compared to adenomas and normal samples (p<0.0001). The area under the receiver-operating characteristic (ROC) curve was 0.84 (95% CI 0.75-0.93, p<0.0001) for cancers versus adenomas, and 0.99 (95% CI 0.98-1.00, p<0.0001) for cancers versus normal tissues. ProNGF overexpression was confirmed in a second cohort (127 cancers of various histological types and 55 normal thyroid tissues) and using a different antibody (p<0.0001). ProNGF staining intensity was highest in papillary carcinomas compared to other histological types (p<0.0001) and there was no significant association with age, gender, tumor size, stage and lymph node status. In conclusion, proNGF is increased in thyroid cancer and should be considered as a new potential diagnostic biomarker.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma/diagnóstico , Factor de Crecimiento Nervioso/biosíntesis , Precursores de Proteínas/biosíntesis , Neoplasias de la Tiroides/diagnóstico , Adenoma/diagnóstico , Adulto , Anciano , Área Bajo la Curva , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factor de Crecimiento Nervioso/análisis , Precursores de Proteínas/análisis , Curva ROC , Sensibilidad y Especificidad
8.
Mol Neurodegener ; 10: 57, 2015 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-26520394

RESUMEN

BACKGROUND: Amyotrophic Lateral Sclerosis is characterized by a focal onset of symptoms followed by a progressive spread of pathology that has been likened to transmission of infectious prions. Cell-to-cell transmission of SOD1 protein aggregates is dependent on fluid-phase endocytosis pathways, although the precise molecular mechanisms remain to be elucidated. RESULTS: We demonstrate in this paper that SOD1 aggregates interact with the cell surface triggering activation of Rac1 and subsequent membrane ruffling permitting aggregate uptake via stimulated macropinocytosis. In addition, other protein aggregates, including those associated with neurodegenerative diseases (TDP-43, Httex146Q, α-synuclein) also trigger membrane ruffling to gain entry into the cell. Aggregates are able to rupture unstructured macropinosomes to enter the cytosol allowing propagation of aggregation to proceed. CONCLUSION: Thus, we conclude that in addition to basic proteostasis mechanisms, pathways involved in the activation of macropinocytosis are key determinants in the spread of pathology in these misfolding diseases.


Asunto(s)
Neuronas Motoras/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Pinocitosis/fisiología , Agregado de Proteínas/fisiología , Superóxido Dismutasa/metabolismo , Animales , Línea Celular , Ratones , Neuronas Motoras/patología , Mutación/genética , Enfermedades Neurodegenerativas/patología , Pliegue de Proteína , Superóxido Dismutasa-1
9.
Cell Mol Life Sci ; 72(23): 4653-69, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26104008

RESUMEN

The Hippo pathway is emerging as a critical nexus that balances self-renewal of progenitors against differentiation; however, upstream elements in vertebrate Hippo signalling are poorly understood. High expression of Fat1 cadherin within the developing neuroepithelium and the manifestation of severe neurological phenotypes in Fat1-knockout mice suggest roles in neurogenesis. Using the SH-SY5Y model of neuronal differentiation and employing gene silencing techniques, we show that FAT1 acts to control neurite outgrowth, also driving cells towards terminal differentiation via inhibitory effects on proliferation. FAT1 actions were shown to be mediated through Hippo signalling where it activated core Hippo kinase components and antagonised functions of the Hippo effector TAZ. Suppression of FAT1 promoted the nucleocytoplasmic shuttling of TAZ leading to enhanced transcription of the Hippo target gene CTGF together with accompanying increases in nuclear levels of Smad3. Silencing of TAZ reversed the effects of FAT1 depletion thus connecting inactivation of TAZ-TGFbeta signalling with Hippo signalling mediated through FAT1. These findings establish FAT1 as a new upstream Hippo element regulating early stages of differentiation in neuronal cells.


Asunto(s)
Cadherinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuronas/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte Activo de Núcleo Celular , Cadherinas/genética , Diferenciación Celular , Línea Celular , Proliferación Celular , Técnicas de Silenciamiento del Gen , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Neuritas/fisiología , Neuronas/fisiología , Transducción de Señal , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
10.
Mol Oncol ; 9(8): 1626-35, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26009480

RESUMEN

Infiltration of the tumor microenvironment by nerve fibers is an understudied aspect of breast carcinogenesis. In this study, the presence of nerve fibers was investigated in a cohort of 369 primary breast cancers (ductal carcinomas in situ, invasive ductal and lobular carcinomas) by immunohistochemistry for the neuronal marker PGP9.5. Isolated nerve fibers (axons) were detected in 28% of invasive ductal carcinomas as compared to only 12% of invasive lobular carcinomas and 8% of ductal carcinomas in situ (p = 0.0003). In invasive breast cancers, the presence of nerve fibers was observed in 15% of lymph node negative tumors and 28% of lymph node positive tumors (p = 0.0031), indicating a relationship with the metastatic potential. In addition, there was an association between the presence of nerve fibers and the expression of nerve growth factor (NGF) in cancer cells (p = 0.0001). In vitro, breast cancer cells were able to induce neurite outgrowth in PC12 cells, and this neurotrophic activity was partially inhibited by anti-NGF blocking antibodies. In conclusion, infiltration by nerve fibers is a feature of the tumor microenvironment that is associated with aggressiveness and involves NGF production by cancer cells. The potential participation of nerve fibers in breast cancer progression needs to be further considered.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Fibras Nerviosas/fisiología , Factores de Crecimiento Nervioso/metabolismo , Microambiente Tumoral , Adulto , Anciano , Anciano de 80 o más Años , Animales , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patología , Línea Celular Tumoral , Femenino , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Células MCF-7 , Persona de Mediana Edad , Fibras Nerviosas/patología , Células PC12 , Ratas
11.
Oncotarget ; 6(12): 10473-86, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25871389

RESUMEN

The neuronal membrane protein sortilin has been reported in a few cancer cell lines, but its expression and impact in human tumors is unclear. In this study, sortilin was analyzed by immunohistochemistry in a series of 318 clinically annotated breast cancers and 53 normal breast tissues. Sortilin was detected in epithelial cells, with increased levels in cancers, as compared to normal tissues (p = 0.0088). It was found in 79% of invasive ductal carcinomas and 54% of invasive lobular carcinomas (p < 0.0001). There was an association between sortilin expression and lymph node involvement (p = 0.0093), suggesting a relationship with metastatic potential. In cell culture, sortilin levels were higher in cancer cell lines compared to non-tumorigenic breast epithelial cells and siRNA knockdown of sortilin inhibited cancer cell adhesion, while proliferation and apoptosis were not affected. Breast cancer cell migration and invasion were also inhibited by sortilin knockdown, with a decrease in focal adhesion kinase and SRC phosphorylation. In conclusion, sortilin participates in breast tumor aggressiveness and may constitute a new therapeutic target against tumor cell invasion.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Neoplasias de la Mama/metabolismo , Apoptosis/fisiología , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Células MCF-7 , Persona de Mediana Edad , Invasividad Neoplásica , Transfección
12.
Cancer Res ; 75(9): 1777-81, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25795709

RESUMEN

Recent studies have revealed the essential role played by nerves in tumor progression. Nerves have been shown to infiltrate the tumor microenvironment and actively stimulate cancer cell growth and dissemination. This mechanism involves the release of neurotransmitters, such as catecholamines and acetylcholine, directly into the vicinity of cancer and stromal cells to activate corresponding membrane receptors. Conversely, the secretion of neurotrophic growth factors by cancer cells drives the outgrowth of nerves in solid tumors. This reciprocal interaction between nerves and cancer cells provides new insights into the cellular and molecular bases of tumorigenesis and points to the potential utility of antineurogenic therapies. This review will discuss our evolving understanding of the cross-talk between nerves and cancer cells.


Asunto(s)
Carcinogénesis/patología , Neoplasias/patología , Neuronas/fisiología , Animales , Progresión de la Enfermedad , Humanos , Neoplasias/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Microambiente Tumoral/fisiología
13.
Adv Biol Regul ; 58: 16-27, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25491371

RESUMEN

Nerve growth factor (NGF) and its precursor (proNGF) are primarily considered as regulators of neuronal function that induce their responses via the tyrosine kinase receptor TrkA and the pan-neurotrophin receptor p75NTR. It has been generally held that NGF exerts its effects primarily through TrkA, inducing a cascade of tyrosine kinase-initiated responses, while proNGF binds more strongly to p75NTR. When this latter entity interacts with a third receptor, sortilin, apoptotic responses are induced in contrast to the survival/differentiation associated with the other two. Recent studies have outlined portions of the downstream phosphoproteome of TrkA in the neuronal PC12 cells and have clarified the contribution of individual docking sites in the TrkA endodomain. The patterns observed showed a similarity with the profile induced by the epidermal growth factor receptor, which is extensively associated with oncogenesis. Indeed, as with other neurotrophic factors, the distribution of TrkA and p75NTR is not limited to neuronal tissue, thus providing an array of targets outside the nervous systems. One such source is breast cancer cells, in which NGF and proNGF stimulate breast cancer cell survival/growth and enhance cell invasion, respectively. This latter activity is exerted via TrkA (as opposed to p75NTR) in conjunction with sortilin. Another tissue overexpressing proNGF is prostate cancer and here the ability of cancer cells to induce neuritogenesis has been implicated in cancer progression. These studies show that the non-neuronal functions of proNGF/NGF are likely integrated with their neuronal activities and point to the clinical utility of these growth factors and their receptors as biomarkers and therapeutic targets for metastasis and cancer pain.


Asunto(s)
Neoplasias de la Mama/genética , Factor de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Neoplasias de la Próstata/genética , Precursores de Proteínas/genética , Receptor trkA/genética , Receptores de Factor de Crecimiento Nervioso/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Precursores de Proteínas/metabolismo , Ratas , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal
14.
Am J Pathol ; 184(12): 3156-62, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25285721

RESUMEN

Nerve infiltration is essential to prostate cancer progression, but the mechanism by which nerves are attracted to prostate tumors remains unknown. We report that the precursor of nerve growth factor (proNGF) is overexpressed in prostate cancer and involved in the ability of prostate cancer cells to induce axonogenesis. A series of 120 prostate cancer and benign prostate hyperplasia (BPH) samples were analyzed by IHC for proNGF. ProNGF was mainly localized in the cytoplasm of epithelial cells, with marked expression in cancer compared with BPH. Importantly, the proNGF level positively correlated with the Gleason score (n = 104, τB = 0.51). A higher level of proNGF was observed in tumors with a Gleason score of ≥8 compared with a Gleason score of 7 and 6 (P < 0.001). In vitro, proNGF was detected in LNCaP, DU145, and PC-3 prostate cancer cells and BPH-1 cells but not in RWPE-1 immortalized nontumorigenic prostate epithelial cells or primary normal prostate epithelial cells. Co-culture of PC12 neuronal-like cells or 50B11 neurons with PC-3 cells resulted in neurite outgrowth in neuronal cells that was inhibited by blocking antibodies against proNGF, indicating that prostate cancer cells can induce axonogenesis via secretion of proNGF. These data reveal that ProNGF is a biomarker associated with high-risk prostate cancers and a potential driver of infiltration by nerves.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Próstata/inervación , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/metabolismo , Axones/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Citoplasma/metabolismo , Células Epiteliales/metabolismo , Humanos , Inmunohistoquímica , Masculino , Clasificación del Tumor , Neuronas/metabolismo , Hiperplasia Prostática/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...