Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Immunol ; 24(4): 664-675, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36849745

RESUMEN

Antigen-specific CD8+ T cell accumulation in tumors is a prerequisite for effective immunotherapy, and yet the mechanisms of lymphocyte transit are not well defined. Here we show that tumor-associated lymphatic vessels control T cell exit from tumors via the chemokine CXCL12, and intratumoral antigen encounter tunes CXCR4 expression by effector CD8+ T cells. Only high-affinity antigen downregulates CXCR4 and upregulates the CXCL12 decoy receptor, ACKR3, thereby reducing CXCL12 sensitivity and promoting T cell retention. A diverse repertoire of functional tumor-specific CD8+ T cells, therefore, exit the tumor, which limits the pool of CD8+ T cells available to exert tumor control. CXCR4 inhibition or loss of lymphatic-specific CXCL12 boosts T cell retention and enhances tumor control. These data indicate that strategies to limit T cell egress might be an approach to boost the quantity and quality of intratumoral T cells and thereby response to immunotherapy.


Asunto(s)
Vasos Linfáticos , Neoplasias , Humanos , Linfocitos T CD8-positivos , Receptores CXCR4/metabolismo , Neoplasias/terapia , Neoplasias/patología , Vasos Linfáticos/metabolismo , Inmunoterapia
3.
J Immunol ; 206(2): 264-272, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33397740

RESUMEN

Lymphatic vessels provide an anatomical framework for immune surveillance and adaptive immune responses. Although appreciated as the route for Ag and dendritic cell transport, peripheral lymphatic vessels are often not considered active players in immune surveillance. Lymphatic vessels, however, integrate contextual cues that directly regulate transport, including changes in intrinsic pumping and capillary remodeling, and express a dynamic repertoire of inflammatory chemokines and adhesion molecules that facilitates leukocyte egress out of inflamed tissue. These mechanisms together contribute to the course of peripheral tissue immunity. In this review, we focus on context-dependent mechanisms that regulate fluid and cellular transport out of peripheral nonlymphoid tissues to provide a framework for understanding the effects of afferent lymphatic transport on immune surveillance, peripheral tissue inflammation, and adaptive immunity.


Asunto(s)
Inflamación/inmunología , Vasos Linfáticos/inmunología , Inmunidad Adaptativa , Animales , Movimiento Celular , Microambiente Celular , Humanos , Vigilancia Inmunológica
4.
Cancer Immunol Immunother ; 67(3): 445-457, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29204701

RESUMEN

A substantial body of evidence suggests the existence of MUC1-specific antibodies and cytotoxic T cell activities in pancreatic cancer patients. However, tumor-induced immunosuppression renders these responses ineffective. The current study explores a novel therapeutic combination wherein tumor-bearing hosts can be immunologically primed with their own antigen, through opsonization with a tumor antigen-targeted antibody, mAb-AR20.5. We evaluated the efficacy of immunization with this antibody in combination with PolyICLC and anti-PD-L1. The therapeutic combination of mAb-AR20.5 + anti-PD-L1 + PolyICLC induced rejection of human MUC1 expressing tumors and provided a long-lasting, MUC1-specific cellular immune response, which could be adoptively transferred and shown to provide protection against tumor challenge in human MUC1 transgenic (MUC.Tg) mice. Furthermore, antibody depletion studies revealed that CD8 cells were effectors for the MUC1-specific immune response generated by the mAb-AR20.5 + anti-PD-L1 + PolyICLC combination. Multichromatic flow cytometry data analysis demonstrated a significant increase over time in circulating, activated CD8 T cells, CD3+CD4-CD8-(DN) T cells, and mature dendritic cells in mAb-AR20.5 + anti-PD-L1 + PolyICLC combination-treated, tumor-bearing mice, as compared to saline-treated control counterparts. Our study provides a proof of principle that an effective and long-lasting anti-tumor cellular immunity can be achieved in pancreatic tumor-bearing hosts against their own antigen (MUC1), which can be further potentiated using a vaccine adjuvant and an immune checkpoint inhibitor.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antígeno B7-H1/antagonistas & inhibidores , Carboximetilcelulosa de Sodio/análogos & derivados , Desoxicitidina/análogos & derivados , Mucina-1/genética , Neoplasias Pancreáticas/mortalidad , Poli I-C/administración & dosificación , Polilisina/análogos & derivados , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Carboximetilcelulosa de Sodio/administración & dosificación , Citotoxicidad Inmunológica , Desoxicitidina/administración & dosificación , Humanos , Inmunidad Celular , Ratones , Ratones Transgénicos , Mucina-1/química , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/prevención & control , Polilisina/administración & dosificación , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
5.
Front Immunol ; 15: 1328602, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38361951

RESUMEN

Introduction: Quantitative, multiplexed imaging is revealing complex spatial relationships between phenotypically diverse tumor infiltrating leukocyte populations and their prognostic implications. The underlying mechanisms and tissue structures that determine leukocyte distribution within and around tumor nests, however, remain poorly understood. While presumed players in metastatic dissemination, new preclinical data demonstrates that blood and lymphatic vessels (lymphovasculature) also dictate leukocyte trafficking within tumor microenvironments and thereby impact anti-tumor immunity. Here we interrogate these relationships in primary human cutaneous melanoma. Methods: We established a quantitative, multiplexed imaging platform to simultaneously detect immune infiltrates and tumor-associated vessels in formalin-fixed paraffin embedded patient samples. We performed a discovery, retrospective analysis of 28 treatment-naïve, primary cutaneous melanomas. Results: Here we find that the lymphvasculature and immune infiltrate is heterogenous across patients in treatment naïve, primary melanoma. We categorized five lymphovascular subtypes that differ by functionality and morphology and mapped their localization in and around primary tumors. Interestingly, the localization of specific vessel subtypes, but not overall vessel density, significantly associated with the presence of lymphoid aggregates, regional progression, and intratumoral T cell infiltrates. Discussion: We describe a quantitative platform to enable simultaneous lymphovascular and immune infiltrate analysis and map their spatial relationships in primary melanoma. Our data indicate that tumor-associated vessels exist in different states and that their localization may determine potential for metastasis or immune infiltration. This platform will support future efforts to map tumor-associated lymphovascular evolution across stage, assess its prognostic value, and stratify patients for adjuvant therapy.


Asunto(s)
Vasos Linfáticos , Melanoma , Neoplasias Cutáneas , Humanos , Estudios Retrospectivos , Inmunohistoquímica , Vasos Linfáticos/patología , Microambiente Tumoral
6.
bioRxiv ; 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36824892

RESUMEN

Tissue resident memory T cells (TRM) provide important protection against infection, and yet the interstitial signals necessary for their formation and persistence remain incompletely understood. Here we show that antigen-dependent induction of the chemokine receptor, CXCR6, is a conserved requirement for TRM formation in peripheral tissue after viral infection. CXCR6 was dispensable for the early accumulation of antigen-specific CD8+ T cells in skin and did not restrain their exit. Single cell sequencing indicated that CXCR6-/- CD8+ T cells were also competent to acquire a transcriptional program of residence but exhibited deficiency in multiple pathways that converged on survival and metabolic signals necessary for memory. As such, CXCR6-/- CD8+ T cells exhibited increased rates of apoptosis relative to controls in the dermis, leading to inefficient TRM formation. CXCR6 expression may therefore represent a common mechanism across peripheral non-lymphoid tissues and inflammatory states that increases the probability of long-term residence.

7.
J Exp Med ; 219(5)2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35353138

RESUMEN

Lymphatic vessels are often considered passive conduits that flush antigenic material, pathogens, and cells to draining lymph nodes. Recent evidence, however, suggests that lymphatic vessels actively regulate diverse processes from antigen transport to leukocyte trafficking and dietary lipid absorption. Here we tested the hypothesis that infection-induced changes in lymphatic transport actively contribute to innate host defense. We demonstrate that cutaneous vaccinia virus infection by scarification activates dermal lymphatic capillary junction tightening (zippering) and lymph node lymphangiogenesis, which are associated with reduced fluid transport and cutaneous viral sequestration. Lymphatic-specific deletion of VEGFR2 prevented infection-induced lymphatic capillary zippering, increased fluid flux out of tissue, and allowed lymphatic dissemination of virus. Further, a reduction in dendritic cell migration to lymph nodes in the absence of lymphatic VEGFR2 associated with reduced antiviral CD8+ T cell expansion. These data indicate that VEGFR2-driven lymphatic remodeling is a context-dependent, active mechanism of innate host defense that limits viral dissemination and facilitates protective, antiviral CD8+ T cell responses.


Asunto(s)
Vasos Linfáticos , Ganglios Linfáticos , Linfangiogénesis , Sistema Linfático , Piel
8.
Lab Invest ; 91(11): 1643-51, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21863060

RESUMEN

The fate of newly synthesized lymphatic vessels induced by inflammation is poorly understood. To address this question, we designed experiments to determine the morphologic, phenotypic, and functional differences in regressing lymphatic vessels in the context of corneal recovery after an inflammatory response. A suture removal modification was used to induce corneal recovery after suture induced inflammation. We identified an increase in markers of corneal inflammation in sutured cornea that resolved in 14 days after suture removal. Sprouting newly synthesized lymphatic vessels trafficking MHC-II-positive leukocytes were visualized in sutured cornea. Following suture removal and recovery, the visualized lymphatic vessels were thin and fragmented, had bulbous termini, discontinuous expression of CD31 and VE-cadherin, and excluded MHC-II-positive leukocytes. VEGF-A, VEGF-C, and TGF-ß mRNA levels were increased during corneal recovery, suggesting a complex interaction between lymphangiogenic factors and the mechanisms that regulate corneal recovery. The balance of lymphatic vessel growth and regression is likely to have a central role in the pathogenesis of corneal inflammatory diseases.


Asunto(s)
Córnea/irrigación sanguínea , Córnea/fisiopatología , Inflamación/fisiopatología , Linfangiogénesis/fisiología , Vasos Linfáticos/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Córnea/cirugía , Neovascularización de la Córnea , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Inflamación/etiología , Ratones , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suturas/efectos adversos
9.
J Vis Exp ; (143)2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30663703

RESUMEN

Leukocyte egress from peripheral tissues to draining lymph nodes is not only critical for immune surveillance and initiation but also contributes to the resolution of peripheral tissue responses. While a variety of methods are used to quantify leukocyte egress from non-lymphoid, peripheral tissues, the cellular and molecular mechanisms that govern context-dependent egress remain poorly understood. Here, we describe the use of in situ photoconversion for quantitative analysis of leukocyte egress from murine skin and tumors. Photoconversion allows for the direct labeling of leukocytes resident within cutaneous tissue. Though skin exposure to violet light induces local inflammatory responses characterized by leukocyte infiltrates and vascular leakiness, in a head-to-head comparison with transdermal application of fluorescent tracers, photoconversion specifically labeled migratory dendritic cell populations and simultaneously enabled the quantification of myeloid and lymphoid egress from cutaneous microenvironments and tumors. The mechanisms of leukocyte egress remain a missing component in our understanding of intratumoral leukocyte complexity, and thus the application of the tools described herein will provide unique insight into the dynamics of tumor immune microenvironments both at steady state and in response to therapy.


Asunto(s)
Leucocitos/metabolismo , Vasos Linfáticos/metabolismo , Neoplasias/patología , Piel/inmunología , Animales , Ganglios Linfáticos/inmunología , Ratones
10.
Pharmacol Ther ; 179: 158-170, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28549596

RESUMEN

Chemokine networks regulate a variety of cellular, physiological, and immune processes. These normal functions can become appropriated by cancer cells to facilitate a more hospitable niche for aberrant cells by enhancing growth, proliferation, and metastasis. This is especially true in pancreatic cancer, where chemokine signaling is a vital component in the development of the supportive tumor microenvironment and the signaling between the cancer cells and surrounding stromal cells. Although expression patterns vary among cancer types, the chemokine receptor CXCR4 has been implicated in nearly every major malignancy and plays a prominent role in pancreatic cancer development and progression. This receptor, in conjunction with its primary chemokine ligand CXCL12, promotes pancreatic cancer development, invasion, and metastasis through the management of the tumor microenvironment via complex crosstalk with other pathways. Thus, CXCR4 likely contributes to the poor prognoses observed in patients afflicted with this malignancy. Recent exploration of combination therapies with CXCR4 antagonists have demonstrated improved outcomes, and abolishing the contribution of this pathway may prove crucial to effectively treat pancreatic cancer at both the primary tumor and metastases.


Asunto(s)
Quimiocina CXCL12/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores CXCR4/metabolismo , Animales , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico
11.
Cancer Lett ; 381(1): 217-36, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-26742462

RESUMEN

This review summarizes current knowledge of the biology, pathology and clinical understanding of lymphatic invasion and metastasis in pancreatic cancer. We discuss the clinical and biological consequences of lymphatic invasion and metastasis, including paraneoplastic effects on immune responses and consider the possible benefit of therapies to treat tumors that are localized to lymphatics. A review of current techniques and methods to study interactions between tumors and lymphatics is presented.


Asunto(s)
Carcinoma Ductal Pancreático/secundario , Sistema Linfático/patología , Neoplasias Pancreáticas/patología , Animales , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/cirugía , Comunicación Celular , Humanos , Escisión del Ganglio Linfático , Linfangiogénesis , Metástasis Linfática , Sistema Linfático/inmunología , Sistema Linfático/metabolismo , Sistema Linfático/cirugía , Invasividad Neoplásica , Pancreatectomía , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/cirugía , Resultado del Tratamiento , Microambiente Tumoral
12.
Oncotarget ; 7(26): 39996-40011, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27220889

RESUMEN

The MUC1 glycoprotein is overexpressed and aberrantly glycosylated in >90% of pancreatic ductal adenocarcinoma cases and impacts tumor progression by initiating downstream signaling through phosphorylation of its cytoplasmic tail. Previous studies have demonstrated that MUC1 alters expression of known targets of activator protein 1 (AP-1); however, no studies have evaluated the precise impact of MUC1 signaling on the activity and formation of AP-1. Given the known role of these proteins in modulating migration, invasion, and tumor progression, we explored the effects of MUC1 on AP-1 dimer formation and function. We determined that MUC1 increased the protein levels of c-Jun, the major component of AP-1, and promoted dimerization of c-Jun with the Fos-protein FRA-1. We demonstrate that FRA-1 acts as a potent mediator of migration and invasion in a manner that is modulated by signals through MUC1, which acts as a dominant regulator of specific AP-1 and FRA-1 target genes. Our results provide the first in vivo evidence of a FRA-1 mediated expression profile that impacts pancreatic tumor growth properties. In summary, we show that MUC1 enhancement of ERK activation influences FRA-1 activity to modulate tumor migration, invasion and metastasis in a subset of pancreatic cancer cases.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Mucina-1/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Glicosilación , Humanos , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Regulación hacia Arriba
13.
PLoS One ; 9(11): e112737, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25383879

RESUMEN

The cellular and physiologic mechanisms that regulate the resolution of inflammation remain poorly defined despite their widespread importance in improving inflammatory disease outcomes. We studied the resolution of two cardinal signs of inflammation-pain and swelling-by investigating molecular mechanisms that regulate neural and lymphatic vessel remodeling during the resolution of corneal inflammation. A mouse model of corneal inflammation and wound recovery was developed to study this process in vivo. Administration of nerve growth factor (NGF) increased pain sensation and inhibited neural remodeling and lymphatic vessel regression processes during wound recovery. A complementary in vivo approach, the corneal micropocket assay, revealed that NGF-laden pellets stimulated lymphangiogenesis and increased protein levels of VEGF-C. Adult human dermal lymphatic endothelial cells did not express canonical NGF receptors TrkA and p75NTR or activate downstream MAPK- or Akt-pathway effectors in the presence of NGF, although NGF treatment increased their migratory and tubulogenesis capacities in vitro. Blockade of the VEGF-R2/R3 signaling pathway ablated NGF-mediated lymphangiogenesis in vivo. These findings suggest a hierarchical relationship with NGF functioning upstream of the VEGF family members, particularly VEGF-C, to stimulate lymphangiogenesis. Taken together, these studies show that NGF stimulates lymphangiogenesis and that NGF may act as a pathogenic factor that negatively regulates the normal neural and lymphatic vascular remodeling events that accompany wound recovery.


Asunto(s)
Córnea/patología , Células Endoteliales/metabolismo , Inflamación/patología , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Cicatrización de Heridas , Adulto , Animales , Células Cultivadas , Córnea/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Factor de Crecimiento Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
14.
PLoS One ; 8(10): e73356, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204560

RESUMEN

Transmembrane mucins, MUC4 and MUC16 are associated with tumor progression and metastatic potential in human pancreatic adenocarcinoma. We discovered that miR-200c interacts with specific sequences within the coding sequence of MUC4 and MUC16 mRNAs, and evaluated the regulatory nature of this association. Pancreatic cancer cell lines S2.028 and T3M-4 transfected with miR-200c showed a 4.18 and 8.50 fold down regulation of MUC4 mRNA, and 4.68 and 4.82 fold down regulation of MUC16 mRNA compared to mock-transfected cells, respectively. A significant reduction of glycoprotein expression was also observed. These results indicate that miR-200c overexpression regulates MUC4 and MUC16 mucins in pancreatic cancer cells by directly targeting the mRNA coding sequence of each, resulting in reduced levels of MUC4 and MUC16 mRNA and protein. These data suggest that, in addition to regulating proteins that modulate EMT, miR-200c influences expression of cell surface mucins in pancreatic cancer.


Asunto(s)
Antígeno Ca-125/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , MicroARNs/genética , Mucina 4/genética , Sistemas de Lectura Abierta , Neoplasias Pancreáticas/genética , Emparejamiento Base , Secuencia de Bases , Sitios de Unión , Antígeno Ca-125/metabolismo , Línea Celular Tumoral , Perfilación de la Expresión Génica , Orden Génico , Humanos , Proteínas de la Membrana/metabolismo , Mucina 4/metabolismo , Interferencia de ARN
15.
Matrix Biol ; 30(3): 218-24, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21406228

RESUMEN

Lymphatic vessel growth requires extensive remodeling of the extracellular matrix, a process hypothesized to be related to the expression and function of the matrix metalloproteinases. We used a protein based screening strategy to demonstrate increased matrix matalloproteinase-10 expression in human lymphatic endothelial cells undergoing collagen I induced tubulogenesis. Knock-down experiments showed that matrix metalloproteinase-10 regulated lymphatic endothelial cell tubulogenesis. ß1 integrin signaling via the ERK/MAPK pathway increased matrix metalloproteinase-10 mRNA and protein expression in human lymphatic endothelial cells. These findings demonstrate a novel mechanism by which ß1 integrin regulates matrix metalloproteinase-10 expression during lymphatic vessel remodeling.


Asunto(s)
Endotelio Linfático/fisiología , Integrina beta1/metabolismo , Metaloproteinasa 10 de la Matriz/metabolismo , Células Cultivadas , Colágeno Tipo I/farmacología , Medios de Cultivo Condicionados , Endotelio Linfático/efectos de los fármacos , Endotelio Linfático/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Análisis por Matrices de Proteínas , Transducción de Señal
16.
Cornea ; 30(12): 1442-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21955630

RESUMEN

PURPOSE: To determine whether glucocorticoids suppress corneal lymphatic vessel growth (lymphangiogenesis) or induce lymphatic vessel regression. METHODS: We measured human lymphatic endothelial cell proliferation and collagen-induced tubulogenesis in culture conditions with and without dexamethasone, a potent glucocorticoid. We developed a modification of the mouse corneal suture model that allowed us to visualize lymphatic vessel growth (with suture) or regression (suture removed) using immunofluorescence and microscopic techniques. We administered dexamethasone or vehicle control to mice with sutured corneas. We visualized and quantified the corneal lymphatic vessels. We measured vascular endothelial growth factor-C and tumor necrosis factor-α messenger RNA expression in unsutured or sutured corneas using quantitative reverse transcriptase-polymerase chain reaction. RESULTS: High-dose dexamethasone did not change the proliferation or tubulogenesis properties of human lymphatic endothelial cells in vitro. We demonstrated suppressed corneal lymphatic vessel growth rather than lymphatic vessel regression in mice treated with dexamethasone. Expressions of corneal vascular endothelial growth factor-C and tumor necrosis factor-α messenger RNA were similar in mice treated with or without dexamethasone. CONCLUSIONS: Dexamethasone suppressed new lymphatic vessel growth and did not induce lymphatic vessel regression. These findings identify a novel mechanism of glucocorticoid action: suppression of corneal lymphangiogenesis.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Endotelio Corneal/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Glucocorticoides/farmacología , Linfangiogénesis/efectos de los fármacos , Animales , Células Cultivadas , Dexametasona/farmacología , Endotelio Corneal/citología , Endotelio Corneal/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Ratones , Modelos Animales , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA