Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
2.
Virus Genes ; 52(5): 613-9, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27234312

RESUMEN

Genogroup II, genotype 3 noroviruses (GII.3 NoVs) are secondary to GII.4 NoVs in causing acute non-bacterial gastroenteritis worldwide. In our previous study, we found that virus-like particles (VLPs) derived from a GII.3 NoV strain exhibited no binding activity to any salivary and synthetic histo-blood group antigens (HBGAs) tested. In this study, the nucleotide sequence encoding the major capsid protein of another documented GII.3 NoV strain was codon-optimized and synthesized, and the major capsid protein was expressed using recombinant baculovirus virus expression system. The assembly of VLPs was verified by electron microscopy, and the binding profiles of the assembled VLPs to salivary HBGAs were determined, and in vitro VLP-salivary HBGAs binding blockade assay was used to test the cross-blocking effects of hyperimmune sera produced against different genotypes (GI.2, GII.3, and GII.4). The expression of the major capsid proteins led to the successful assembly of VLPs, and in vitro VLP-salivary HBGAs binding assay indicated that the assembled VLPs bound to salivary HBGAs from blood type A, B, AB, and O individuals, with the highest binding capacity to type A salivary HBGAs. In vitro VLP-salivary HBGAs binding blockade assay demonstrated the absence of blocking activities for hyperimmune sera produced against GI.2and GII.4 VLPs and the presence of blocking activity for that against GII.3 VLPs. Our results suggest the absence of cross-blocking activities among different genotypes and the presence of blocking activities between GII.3 NoVs from different clusters, which might have implications for the design of multivalent NoV vaccines.


Asunto(s)
Proteínas de la Cápside/genética , Norovirus/genética , Baculoviridae/genética , Secuencia de Bases/genética , Antígenos de Grupos Sanguíneos/genética , Genotipo , Humanos , Receptores Virales/genética , Glándulas Salivales/virología , Acoplamiento Viral
3.
Cell Death Discov ; 10(1): 148, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509100

RESUMEN

Diabetic cardiomyopathy (DCM), an important complication of diabetes mellitus (DM), is one of the most serious chronic heart diseases and has become a major cause of heart failure worldwide. At present, the pathogenesis of DCM is unclear, and there is still a lack of effective therapeutics. Previous studies have shown that the homeostasis of mitochondria and the endoplasmic reticulum (ER) play a core role in maintaining cardiovascular function, and structural and functional abnormalities in these organelles seriously impact the occurrence and development of various cardiovascular diseases, including DCM. The interplay between mitochondria and the ER is mediated by the mitochondria-associated ER membrane (MAM), which participates in regulating energy metabolism, calcium homeostasis, mitochondrial dynamics, autophagy, ER stress, inflammation, and other cellular processes. Recent studies have proven that MAM is closely related to the initiation and progression of DCM. In this study, we aim to summarize the recent research progress on MAM, elaborate on the key role of MAM in DCM, and discuss the potential of MAM as an important therapeutic target for DCM, thereby providing a theoretical reference for basic and clinical studies of DCM treatment.

4.
Front Pharmacol ; 15: 1349069, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38384297

RESUMEN

The global prevalence of diabetes mellitus (DM) has led to widespread multi-system damage, especially in cardiovascular and renal functions, heightening morbidity and mortality. Emerging antidiabetic drugs sodium-glucose cotransporter 2 inhibitors (SGLT2i), glucagon-like peptide-1 receptor agonists (GLP-1RAs), and dipeptidyl peptidase-4 inhibitors (DPP-4i) have demonstrated efficacy in preserving cardiac and renal function, both in type 2 diabetic and non-diabetic individuals. To understand the exact impact of these drugs on cardiorenal protection and underlying mechanisms, we conducted a comprehensive review of recent large-scale clinical trials and basic research focusing on SGLT2i, GLP-1RAs, and DPP-4i. Accumulating evidence highlights the diverse mechanisms including glucose-dependent and independent pathways, and revealing their potential cardiorenal protection in diabetic and non-diabetic cardiorenal disease. This review provides critical insights into the cardiorenal protective effects of SGLT2i, GLP-1RAs, and DPP-4i and underscores the importance of these medications in mitigating the progression of cardiovascular and renal complications, and their broader clinical implications beyond glycemic management.

5.
Adv Sci (Weinh) ; 11(10): e2305563, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38145959

RESUMEN

Diabetic nephropathy (DN) is a serious microvascular complication of diabetes. Ferroptosis, a new form of cell death, plays a crucial role in the pathogenesis of DN. Renal tubular injury triggered by ferroptosis might be essential in this process. Numerous studies demonstrate that the vitamin D receptor (VDR) exerts beneficial effects by suppressing ferroptosis. However, the underlying mechanism has not been fully elucidated. Thus, they verified the nephroprotective effect of VDR activation and explored the mechanism by which VDR activation suppressed ferroptosis in db/db mice and high glucose-cultured proximal tubular epithelial cells (PTECs). Paricalcitol (PAR) is a VDR agonist that can mitigate kidney injury and prevent renal dysfunction. PAR treatment could inhibit ferroptosis of PTECs through decreasing iron content, increasing glutathione (GSH) levels, reducing malondialdehyde (MDA) generation, decreasing the expression of positive ferroptosis mediator transferrin receptor 1 (TFR-1), and enhancing the expression of negative ferroptosis mediators including ferritin heavy chain (FTH-1), glutathione peroxidase 4 (GPX4), and cystine/glutamate antiporter solute carrier family 7 member 11 (SLC7A11). Mechanistically, VDR activation upregulated the NFE2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) signaling pathway to suppress ferroptosis in PTECs. These findings suggested that VDR activation inhibited ferroptosis of PTECs in DN via modulating the Nrf2/HO-1 signaling pathway.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Ferroptosis , Animales , Ratones , Células Epiteliales , Glutatión , Hemo-Oxigenasa 1 , Factor 2 Relacionado con NF-E2 , Receptores de Calcitriol , Transducción de Señal
6.
Cell Death Discov ; 9(1): 256, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37479697

RESUMEN

Diabetic cardiomyopathy (DCM) mainly refers to myocardial metabolic dysfunction caused by high glucose, and hyperglycemia is an independent risk factor for cardiac function in the absence of coronary atherosclerosis and hypertension. DCM, which is a severe complication of diabetes, has become the leading cause of heart failure in diabetic patients. The initial symptoms are inconspicuous, and patients gradually exhibit left ventricular dysfunction and eventually develop total heart failure, which brings a great challenge to the early diagnosis of DCM. To date, the underlying pathological mechanisms of DCM are complicated and have not been fully elucidated. Although there are therapeutic strategies available for DCM, the treatment is mainly focused on controlling blood glucose and blood lipids, and there is a lack of effective drugs targeting myocardial injury. Thus, a large percentage of patients with DCM inevitably develop heart failure. Given the neglected initial symptoms, the intricate cellular and molecular mechanisms, and the lack of available drugs, it is necessary to explore early diagnostic biomarkers, further understand the signaling pathways involved in the pathogenesis of DCM, summarize the current therapeutic strategies, and develop new targeted interventions.

7.
Int J Biol Sci ; 19(12): 3726-3743, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37564215

RESUMEN

Ferroptosis is an iron-dependent programmed cell death pattern that is characterized by iron overload, reactive oxygen species (ROS) accumulation and lipid peroxidation. Growing viewpoints support that the imbalance of iron homeostasis and the disturbance of lipid metabolism contribute to tissue or organ injury in various kidney diseases by triggering ferroptosis. At present, the key regulators and complicated network mechanisms associated with ferroptosis have been deeply studied; however, its role in the initiation and progression of kidney diseases has not been fully revealed. Herein, we aim to discuss the features, key regulators and complicated network mechanisms associated with ferroptosis, explore the emerging roles of organelles in ferroptosis, gather its pharmacological progress, and systematically summarize the most recent discoveries about the crosstalk between ferroptosis and kidney diseases, including renal cell carcinoma (RCC), acute kidney injury (AKI), diabetic kidney disease (DKD), autosomal dominant polycystic kidney disease (ADPKD), renal fibrosis, lupus nephritis (LN) and IgA nephropathy. We further conclude the potential therapeutic strategies by targeting ferroptosis for the prevention and treatment of kidney diseases and hope that this work will provide insight for the further study of ferroptosis in the pathogenesis of kidney-related diseases.


Asunto(s)
Lesión Renal Aguda , Nefropatías Diabéticas , Ferroptosis , Neoplasias Renales , Humanos , Hierro , Peroxidación de Lípido
8.
Front Immunol ; 13: 956090, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35958563

RESUMEN

LAG-3, a type of immune checkpoint receptor protein belonging to the immunoglobulin superfamily, is confirmed to be expressed on activated immune cells, mainly including activated T cells. LAG-3 can negatively regulate the function of T cells, exerting important effects on maintaining the homeostasis of the immune system under normal physiological conditions and promoting tumor cells immune escape in the tumor microenvironment. Given its important biological roles, LAG-3 has been regarded as a promising target for cancer immunotherapy. To date, many LAG-3 inhibitors have been reported, which can be divided into monoclonal antibody, double antibody, and small molecule drug, some of which have entered the clinical research stage. LAG-3 inhibitors can negatively regulate and suppress T cell proliferation and activation through combination with MHC II ligand. Besides, LAG-3 inhibitors can also affect T cell function via binding to Galectin-3 and LSECtin. In addition, LAG-3 inhibitors can prevent the FGL1-LAG-3 interaction, thereby enhancing the human body's antitumor immune effect. In this review, we will describe the function of LAG-3 and summarize the latest LAG-3 inhibitors in the clinic for cancer therapy.


Asunto(s)
Neoplasias , Anticuerpos Monoclonales/uso terapéutico , Fibrinógeno , Humanos , Inmunoterapia , Escape del Tumor , Microambiente Tumoral
9.
Front Immunol ; 13: 972345, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36159787

RESUMEN

Cancer immunotherapy has emerged as a novel anti-tumor treatment. Despite significant breakthroughs, cancer immunotherapy remains focused on several types of tumors that are sensitive to the immune system. Therefore, effective strategies to expand its indications and improve its efficacy become key factors for the further development of cancer immunotherapy. In recent decades, the anticancer activities of natural products are reported to have this effect on cancer immunotherapy. And the mechanism is largely attributed to the remodeling of the tumor immunosuppressive microenvironment. The compelling data highlight that natural products offer an alternative method option to improve immune function in the tumor microenvironment (TME). Currently, more attention is being paid to the discovery of new potential modulators of tumor immunotherapy from natural products. In this review, we describe current advances in employing natural products and natural small-molecule drugs targeting immune cells to avoid tumor immune escape, which may bring some insight for guiding tumor treatment.


Asunto(s)
Productos Biológicos , Neoplasias , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Humanos , Inmunoterapia/métodos , Microambiente Tumoral
10.
J Med Chem ; 65(1): 163-190, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34939411

RESUMEN

DCN1, a co-E3 ligase, interacts with UBC12 and activates cullin-RING ligases (CRLs) by catalyzing cullin neddylation. Although DCN1 has been recognized as an important therapeutic target for human diseases, its role in the cardiovascular area remains unknown. Here, we first found that DCN1 was upregulated in isolated cardiac fibroblasts (CFs) treated by angiotensin (Ang) II and in mouse hearts after pressure overload. Then, structure-based optimizations for DCN1-UBC12 inhibitors were performed based on our previous work, yielding compound DN-2. DN-2 specifically targeted DCN1 at molecular and cellular levels as shown by molecular modeling studies, HTRF, cellular thermal shift and co-immunoprecipitation assays. Importantly, DN-2 effectively reversed Ang II-induced cardiac fibroblast activation, which was associated with the inhibition of cullin 3 neddylation. Our findings indicate a potentially unrecognized role of DCN1 inhibition for anticardiac fibrotic effects. DN-2 may be used as a lead compound for further development.


Asunto(s)
Antifibróticos , Descubrimiento de Drogas , Inhibidores Enzimáticos , Fibrosis , Cardiopatías , Péptidos y Proteínas de Señalización Intracelular , Pirimidinas , Enzimas Ubiquitina-Conjugadoras , Animales , Masculino , Ratones , Ratas , Antifibróticos/química , Antifibróticos/farmacología , Proteínas Cullin/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Fibrosis/patología , Cardiopatías/tratamiento farmacológico , Cardiopatías/metabolismo , Cardiopatías/patología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Ratones Endogámicos C57BL , Proteína NEDD8/metabolismo , Pirimidinas/química , Ratas Sprague-Dawley , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Ubiquitinas
11.
Eur J Med Chem ; 211: 113108, 2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33385852

RESUMEN

In this work, we reported the discovery of compound 6i with potent antiproliferative activity against MGC-803. Among these compounds, the most potent compound 6i could effectively inhibit MGC-803 (IC50 = 0.96 µM), being around 38-fold selectivity over GES-1. Further underlying mechanism studies indicated that 6i inhibited the colony formation, migration of MGC-803, and exerted anti-proliferative effect by inducing G0/G1 phase arrest in MGC-803 cells. Cell apoptosis was induced by 6i through activating mitochondria-mediated intrinsic pathway and the death receptor-mediated extrinsic pathway. 6i induced cell apoptosis by elevating the level of ROS. Also, 6i up-regulated pro-apoptotic Bax and p53 level, while down-regulating anti-apoptotic Bcl-2 protein expression. Furthermore, acute toxicity experiment indicated 6i exhibited good safety in vivo. Therefore, 6i may be a template for future development of [1,2,4]triazolo [1,5-a]pyrimidine-based anti-cancer agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Pirimidinas/uso terapéutico , Antineoplásicos/farmacología , Diseño de Fármacos , Humanos , Pirimidinas/farmacología , Relación Estructura-Actividad
12.
Eur J Med Chem ; 210: 112970, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33153765

RESUMEN

To discover novel anticancer agents with potent and low toxicity, we designed and synthesized a range of new thiosemicarbazone-indole analogues based on lead compound 4 we reported previously. Most compounds displayed moderate to high anticancer activities against five tested tumor cells (PC3, EC109, DU-145, MGC803, MCF-7). Specifically, the represented compound 16f possessed strong antiproliferative potency and high selectivity toward PC3 cells with the IC50 value of 0.054 µM, compared with normal WPMY-1 cells with the IC50 value of 19.470 µM. Preliminary mechanism research indicated that compound 16f could significantly suppress prostate cancer cells (PC3, DU-145) growth and colony formation in a dose-dependent manner. Besides, derivative 16f induced G1/S cycle arrest and apoptosis, which may be related to ROS accumulation due to the activation of MAPK signaling pathway. Furthermore, molecule 16f could effectively inhibit tumor growth through a xenograft model bearing PC3 cells and had no evident toxicity in vivo. Overall, based on the biological activity evaluation, analogue 16f can be viewed as a potential lead compound for further development of novel anti-prostate cancer drug.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Indoles/farmacología , Tiosemicarbazonas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indoles/química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad , Tiosemicarbazonas/química , Células Tumorales Cultivadas
13.
Eur J Med Chem ; 202: 112471, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32619887

RESUMEN

Drug repurposing plays a vital role in the discovery of undescribed bioactivities in clinical drugs. Based on drug repurposing strategy, we for the first time reported a novel series of troxipide analogs and then evaluated their antiproliferative activity against MCF-7, PC3, MGC-803, and PC9 cancer cell lines and WPMY-1, most of which showed obvious selectivity toward PC-3 over the other three cancer cell lines and WPMY-1. Compound 5q, especially, could effectively inhibit PC3 with an IC50 value of 0.91 µM, which exhibited around 53-fold selectivity toward WPMY-1. Data indicated that 5q effectively inhibited the colony formation, suppressed the cell migration, and induced G1/S phase arrest in PC3 cells. Also, compound 5q induced cell apoptosis by activating the two apoptotic signaling pathways in PC3 cells: death receptor-mediated extrinsic pathway and mitochondria-mediated intrinsic pathway. Compound 5q up-regulated the expression of both pro-apoptotic Bax and P53, while down-regulated anti-apoptotic Bcl-2 expression. Besides, compound 5q significantly increased the expression of cleaved caspase 3/9 and cleaved PARP. Therefore, the successful discovery of compound 5q may further validate the feasibility of this theory, which will encourage researchers to reveal undescribed bioactivities in traditional drugs.


Asunto(s)
Antineoplásicos/farmacología , Reposicionamiento de Medicamentos , Piperidinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Relación Estructura-Actividad
14.
Int J Biol Macromol ; 101: 427-437, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28315766

RESUMEN

Drug resistance and metastasis significantly hinder chemotherapy and worsen prognoses in cancer. Bone morphogenetic protein 4 (BMP4) belongs to the TGF-ß superfamily, has broad biological activities in cell proliferation and cartilage differentiation and is also able to induce migration and invasion. Herein, we investigated the role of BMP4 in the regulation of metastasis in paclitaxel-resistant human esophageal carcinoma EC109 cells (EC109/Taxol) and docetaxel-resistant human gastric cancer MGC803 cells (MGC/Doc). In these drug-resistant cell lines, we found the cell motility was enhanced and BMP4 was up-regulated relative to their respective parental cell lines. Consistent with in vitro assays, migration potential and BMP4 expression were increased in EC109/Taxol nude mice. Furthermore, to address whether BMP4 was required to enhance the metastatic in EC109/Taxol cells, the pharmacological inhibitor of BMP signaling dorsomorphin was used; meanwhile, we found that the migration and invasion abilities were inhibited. Moreover, the canonical Smad signaling pathway was investigated. Overall, our studies demonstrated that BMP4 participates in the regulation of invasion and migration by EC109/Taxol cells, and inhibition of BMP4 may be a novel strategy to interfere with metastasis in cancer therapy.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Movimiento Celular , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas Smad/metabolismo
15.
Biomed Pharmacother ; 90: 897-905, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28441715

RESUMEN

Paclitaxel (PTX) is one of the most effective drugs used in the treatment of esophageal cancer, however, paclitaxel resistance represents a key limitation during the treatment process. In this study, we investigated the changes of Bcl-2 family members in the moderate paclitaxel-resistance of esophageal carcinoma EC109/PTX cells both in vitro and in vivo. Moreover, we evaluated the reversal effect using siRNAs and the recombinant inhibitor TW37 targeting Bcl-2, Bcl-XL and Mcl-1. Our findings show that downregulation of Bcl-2, Bcl-XL and Mcl-1 can significantly promote EC109/PTX cell apoptosis and reduce the EC109/PTX cell resistance index (RI). Furthermore, TW37 in combination with a P-gp inhibitor can synergistically reverse the paclitaxel resistance in EC109/PTX cells. These results suggest that targeting of the Bcl-2 family and P-gp is capable of reversing the resistance in EC109/PTX cells and the two-inhibitor combination may be a novel treatment strategy for resistant esophageal cancer.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Resistencia a Múltiples Medicamentos/fisiología , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Paclitaxel/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Antineoplásicos Fitogénicos/farmacología , Apoptosis/fisiología , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteína bcl-X/metabolismo
16.
Virus Res ; 224: 1-5, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27521750

RESUMEN

Noroviruses (NoVs) are a leading cause of non-bacterial acute gastroenteritis worldwide. The prevalence of Genogroup II, genotype 3 (GII.3) NoVs is secondary to the epidemic GII.4 strains which show broad spectrum binding activities against multiple types of histo-blood group antigens (HBGAs). In our previous study it was found that GII.3 NoV VLPs exhibited no binding activity to all synthetic and salivary HBGAs tested. To determine the compatibility of P2 domains between different genotypes and its effect over the binding specificity to HBGAs, we swapped the P2 domain of a GII.4 strain (Sydney 2012-like variant) with that of a GII.3 strain (GII.4-VP1/GII.3-P2). In vitro VLP-HBGA binding and binding blockade assays were used to characterize the binding patterns of GII.4-VP1/GII.3-P2 chimeric capsid protein. Expression of GII.4-VP1/GII.3-P2 chimeric capsid protein using recombinant bacuolovirus expression system led to assembly of virus like particles (VLPs). In vitro VLP-HBGA binding assay using synthetic and salivary HBGAs indicated binding activities to blood type A (trimer), Le(x) and blood type A, B and O salivary HBGAs. In vitro VLP-HBGA binding blockade assay indicated that the binding could be blocked by rabbit hyperimmune serum against GII.3 VLPs, but not hyperimmune sera against GI.2 and GII.4 VLPs. These results indicate that the observed binding activities may be caused by conformational changes of inserted P2 domain and possibly reflect the actual binding profile of GII.3 VLPs. The currently observed absence of binding of GII.3 NoV VLPs to salivary or synthetic HBGAs might be due to absence of other unknown factors.


Asunto(s)
Antígenos de Grupos Sanguíneos/metabolismo , Proteínas de la Cápside/química , Gastroenteritis/virología , Norovirus/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Virión/metabolismo , Proteínas de la Cápside/genética , Genotipo , Humanos , Sueros Inmunes/metabolismo , Norovirus/química , Norovirus/genética , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Virión/química , Virión/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA