Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 331
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L872-L884, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34523355

RESUMEN

The influenza virus infection poses a serious health threat worldwide. Myeloid cells play pivotal roles in regulating innate and adaptive immune defense. A disintegrin and metalloproteinase (ADAM) family of proteins contributes to various immune responses; however, the role of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in influenza virus infection remains largely unknown. Herein, we investigated its role, focusing on myeloid cells, during influenza virus infection in mice. ADAM10 gene (Adam10)flox/flox/Lyz2-Cre (Adam10ΔLyz2) and control Adam10flox/flox mice were intranasally infected with 200 plaque-forming units of influenza virus A/H1N1/PR8/34. Adam10ΔLyz2 mice exhibited a significantly higher mortality rate, stronger lung inflammation, and a higher virus titer in the lungs than control mice. Macrophages and inflammatory cytokines, such as TNF-α, IL-1ß, and CCL2, were increased in bronchoalveolar lavage fluid from Adam10ΔLyz2 mice following infection. CD11b+Ly6G-F4/80+ myeloid cells, which had an inflammatory monocyte/macrophage-like phenotype, were significantly increased in the lungs of Adam10ΔLyz2 mice. Adoptive transfer experiments suggested that these cells likely contributed to the poorer prognosis in Adam10ΔLyz2 mice. Seven days after infection, CD11b+Ly6G-F4/80+ lung cells exhibited significantly higher arginase-1 expression levels in Adam10ΔLyz2 mice than in control mice, whereas an arginase-1 inhibitor improved the prognosis of Adam10ΔLyz2 mice. Enhanced granulocyte-macrophage colony-stimulating factor (GM-CSF)/GM-CSF receptor signaling likely contributed to this process. Collectively, these results indicate that myeloid ADAM10 protects against influenza virus pneumonia and may be a promising therapeutic target.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Arginasa/biosíntesis , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Células Mieloides/inmunología , Infecciones por Orthomyxoviridae/patología , Proteína ADAM10/genética , Traslado Adoptivo/métodos , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Arginasa/antagonistas & inhibidores , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/análisis , Inmunidad Innata/inmunología , Macrófagos/trasplante , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/trasplante , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/prevención & control , Pronóstico , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
2.
Pediatr Res ; 89(4): 830-837, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32464635

RESUMEN

BACKGROUND: Arginases (ARG isoforms, ARG-1/ARG-2) are key regulatory enzymes of inflammation and tissue repair; however, their role after neonatal brain hypoxia (H) and hypoxia-ischemia (HI) remains unknown. METHODS: C57BL/6 mice subjected to the Vannucci procedure on postnatal day (P9) were sacrificed at different timepoints. The degree of brain damage was assessed histologically. ARG spatiotemporal localization was determined via immunohistochemistry. ARG expression was measured by Western blot and activity spectrophotometrically. RESULTS: ARG isoform expression increased during neurodevelopment (P9-P17) in the cortex and hippocampus. This was suppressed with H and HI only in the hippocampus. In the cortex, both isoforms increased with H alone and only ARG-2 increased with HI at 3 days. ARG activity during neurodevelopment remained unchanged, but increased at 1 day with H and not HI. ARG-1 localized with microglia at the injury site as early as 4 h after injury, while ARG-2 localized with neurons. CONCLUSIONS: ARG isoform expression increases with age from P9 to P17, but is suppressed by injury specifically in the hippocampus and not in the cortex. Both levels and activity of ARG isoforms increase with H, while ARG-1 immunolabelling is upregulated in the HI cortex. Evidently, ARG isoforms in the brain differ in spatiotemporal localization, expression, and activity during neurodevelopment and after injury. IMPACT: Arginase isoforms change during neurodevelopment and after neonatal brain HI. This is the first study investigating the key enzymes of inflammation and tissue repair called arginases following murine neonatal brain HI. The highly region- and cell-specific expression suggests the possibility of specific functions of arginases. ARG-1 in microglia at the injury site may regulate neuroinflammation, while ARG-2 in neurons of developmental structures may impact neurodevelopment. While further studies are needed to describe the exact role of ARGs after neonatal brain HI, our study adds valuable data on anatomical localization and expression of ARGs in brain during development and after stroke.


Asunto(s)
Arginasa/biosíntesis , Arginasa/química , Hipoxia-Isquemia Encefálica/patología , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/patología , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Hipoxia/patología , Inmunohistoquímica , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Enfermedades Neuroinflamatorias , Neuronas/metabolismo , Isoformas de Proteínas
3.
World J Microbiol Biotechnol ; 37(7): 123, 2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34160683

RESUMEN

Lipopeptides are important secondary metabolites produced by microbes. They find applications in environmental decontamination and in the chemical, pharmaceutical and food industries. However, their production is expensive. In the present work we propose three strategies to lower the production costs of surfactin. First, the coproduction of surfactin and arginase in a single growth. Second, extract the fraction of surfactin that adsorbs to the biomass and is removed from the growth medium through centrifugation. Third, use microbial biomass for the remediation of organic and inorganic contaminants. The coproduction of surfactin and arginase was evaluated by factorial design experiments using the LB medium supplemented with arginine. The best conditions for surfactin production were 22 h of growth at 37 °C using LB supplemented with arginine 7.3 g/L. Almost similar conditions were found to produce highest levels of arginase, 24 h and 6.45 g/L arginine. Decontamination of phenol and copper from artificial samples was attained by treatment with residues from lipopeptide production. Thus, cell suspensions and wash-waters used to extract surfactin from the biomass. Cell suspensions were used to successfully remove hydroquinone. Cell suspensions and wash-waters containing surfactin were successfully used to recover copper from solution. Specific monitoring methods were used for phenol and metal solutions, respectively a biosensor based on tyrosinase and either atomic absorption flame ionization spectrometry or absorbance coupled to the Arduino™ platform. Therefore, we report three alternative strategies to lower the production costs in lipopeptide production, which include the effective recovery of copper and phenol from contaminated waters using residues from surfactin production. Sustainable and profitable production of surfactin can be achieved by a coproduction strategy of lipopeptides and enzymes. Lipopeptides are collected in the supernatant and enzymes in the biomass. In addition, lipopeptides that coprecipitate with biomass can be recovered by washing. Lipopeptide wash-waters find applications in remediation and cells can also be used for environmental decontamination.


Asunto(s)
Arginasa/biosíntesis , Bacillus/enzimología , Bacillus/crecimiento & desarrollo , Bacillus/metabolismo , Lipopéptidos/biosíntesis , Péptidos Cíclicos/biosíntesis , Bacillus/genética , Proteínas Bacterianas/biosíntesis , Biomasa , Reactores Biológicos , Cobre/metabolismo , Medios de Cultivo , ADN Bacteriano , Microbiología Ambiental , Restauración y Remediación Ambiental , Hidroquinonas/metabolismo , Fenol/metabolismo
4.
Cell Immunol ; 349: 104047, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32019673

RESUMEN

The polarization of macrophages is critical to inflammation and tissue repair, with unbalanced macrophage polarization associated with critical dysfunctions of the immune system. Cytochrome P450 1A1 (CYP1A1) is a hydroxylase mainly controlled by the inflammation-limiting aryl hydrocarbon receptor (AhR), which plays a critical role in mycoplasma infection, oxidative stress injury, and cancer. Arginase-1 (Arg-1) is a surrogate for polarized alternative macrophages and is important to the production of nitric oxide (NO) by the modulation of arginine. In the present study, we found CYP1A1 to be upregulated in IL-4-stimulated mouse peritoneal macrophages (PMs) and human peripheral blood monocytes. Using CYP1A1-overexpressing RAW264.7 cells (CYP1A1/RAW) we found that CYP1A1 augmented Arg-1 expression by strengthening the activation of the JAK1/STAT6 signaling pathway in macrophages treated with IL-4. 15(S)-HETE, a metabolite of CYP1A1 hydroxylase, was elevated in IL-4-induced CYP1A1/RAW cells. Further, in macrophages, the loss-of-CYP1A1-hydroxylase activity was associated with reduced IL-4-induced Arg-1 expression due to impaired 15(S)-HETE generation. Of importance, CYP1A1 overexpressing macrophages reduced the inflammation associated with LPS-induced peritonitis. Taken together, these findings identified a novel signaling axis, CYP1A1-15(S)-HETE-JAK1-STAT6, that may be a promising target for the proper maintenance of macrophage polarization and may also be a means by which to treat immune-related disease due to macrophage dysfunction.


Asunto(s)
Arginasa/biosíntesis , Citocromo P-450 CYP1A1/fisiología , Janus Quinasa 1/antagonistas & inhibidores , Macrófagos Peritoneales/efectos de los fármacos , Peritonitis/prevención & control , Factor de Transcripción STAT6/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacología , Traslado Adoptivo , Animales , Araquidonato 15-Lipooxigenasa/fisiología , Arginasa/genética , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1A1/genética , Endotoxinas/toxicidad , Humanos , Ácidos Hidroxieicosatetraenoicos/biosíntesis , Ácidos Hidroxieicosatetraenoicos/genética , Ácidos Hidroxieicosatetraenoicos/farmacología , Interleucina-4/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/trasplante , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Peritonitis/inducido químicamente , Células RAW 264.7 , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores Citoplasmáticos y Nucleares/genética , Células THP-1 , Regulación hacia Arriba/efectos de los fármacos
5.
Acta Biochim Biophys Sin (Shanghai) ; 52(11): 1257-1264, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33128544

RESUMEN

Arginase I (ARG1) is a cytosolic enzyme that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. The association of ARG1 with cancer has mostly been focused on the ARG1 released by tumor-associated myeloid cells in tumor microenvironment. However, the role of ARG1 expressed in cancer cells is unclear. Here, we showed that the expression of ARG1 in human breast cancer (BC) is related to a good prognosis in BC patients. Overexpression of ARG1 suppresses BC cell proliferation and migration in vitro and xenograft tumor growth and development in mouse models. Furthermore, ARG1 expression down-regulates the expression of p-AKT, leading to the de-activation of AKT signal pathway in BC cells. Thus, our results established that in contrast to the role of ARG1 released from tumor-associated myeloid cells in tumor microenvironment that promotes tumor immune escape, ARG1 expressed in BC cells suppresses AKT signaling pathway and functions as a tumor suppressor.


Asunto(s)
Arginasa/biosíntesis , Arginasa/genética , Neoplasias de la Mama/metabolismo , Animales , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Genes Supresores de Tumor , Xenoinjertos/patología , Xenoinjertos/trasplante , Humanos , Ratones Desnudos , Persona de Mediana Edad , Pronóstico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética
6.
Respir Res ; 19(1): 98, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29792217

RESUMEN

BACKGROUND: Asthma is a chronic respiratory condition, with airway hyperresponsiveness (AHR) and inflammation as hallmarks. The hypothesis that the substantially increased expression of arginase 1 in activated macrophages limits the availability of L-arginine for nitric oxide synthesis, and thus increases AHR in lungs of mice with experimentally induced allergic asthma was recently refuted by several studies. In the present study, we tested the hypothesis that, instead, a low circulating concentration of arginine aggravates AHR in the same murine asthma model. Female FVB F/A2 tg/tg transgenic mice, which overexpress rat arginase 1 in their enterocytes, exhibit a ~ 50% decrease of their plasma L-arginine concentration. METHODS: Adult female F/A2 tg/tg mice and their wild-type littermates (F/A2 wt/wt ) were sensitized and challenged with ovalbumin (OVA/OVA). Lung function was assessed with the flexiVent™ system. Adaptive changes in the expression of arginine-metabolizing or -transporting enzymes, chemokines and cytokines, and lung histology were quantified with qPCR, ELISA, and immunohistochemistry, respectively. RESULTS: Reduction of circulating L-arginine concentration significantly increased AHR in OVA/OVA-treated mice and, to a lesser extent, even in PBS/OVA-treated mice. The pulmonary inflammatory response in OVA/OVA-treated F/A2 tg/tg and F/A2 wt/wt mice was comparable. OVA/OVA-treated F/A2 tg/tg mice differed from similarly treated female mice, in which arginase 1 expression in lung macrophages was eliminated, by a complete absence of an adaptive increase in the expression of arginine-metabolizing or -transporting enzymes. CONCLUSION: A reduction of the circulating L-arginine concentration rather than the macrophage-mediated increase of arginine catabolism worsens AHR.


Asunto(s)
Arginina/sangre , Asma/sangre , Pulmón/metabolismo , Hipersensibilidad Respiratoria/sangre , Animales , Arginasa/biosíntesis , Arginina/deficiencia , Asma/patología , Hiperreactividad Bronquial/sangre , Hiperreactividad Bronquial/patología , Femenino , Pulmón/patología , Ratones , Ratones Transgénicos , Hipersensibilidad Respiratoria/patología
7.
RNA Biol ; 15(6): 756-762, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29578372

RESUMEN

mRNA based therapies hold great promise for the treatment of genetic diseases. However, this therapeutic approach suffers from multiple challenges including the short half-life of exogenously administered mRNA and subsequent protein production. Modulation of untranslated regions (UTR) represents one approach to enhance both mRNA stability and translation efficiency. The current studies describe and validate screening methods using a diverse set of 5'UTR and 3'UTR combinations for improved expression of the Arginase 1 (ARG1) protein, a potential therapeutic mRNA target. Data revealed a number of critical aspects which need to be considered when developing a screening approach for engineering mRNA improvements. First, plasmid-based screening methods do not correlate with protein expression driven by exogenously expressed mRNA. Second, improved ARG1 protein production was driven by increased translation and not improved mRNA stability. Finally, the 5' UTR appears to be the key driver in protein expression for exogenously delivered mRNA. From the testing of the combinatorial library, the 5'UTR for complement factor 3 (C3) and cytochrome p4502E1 (CYP2E1) showed the largest and most consistent increase in protein expression relative to a reference UTR. Collectively, these data provide important information for the development and optimization of therapeutic mRNAs.


Asunto(s)
Regiones no Traducidas 3' , Regiones no Traducidas 5' , Arginasa , Complemento C3/genética , Citocromo P-450 CYP2E1/genética , Biosíntesis de Proteínas/genética , Arginasa/biosíntesis , Arginasa/genética
8.
Clin Exp Pharmacol Physiol ; 45(6): 556-562, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29266319

RESUMEN

The L-arginine/NO pathway is an important regulator of pulmonary hypertension, the leading cause of mortality in patients with the chronic lung disease of prematurity, bronchopulmonary dysplasia. L-arginine can be metabolized by NO synthase (NOS) to form L-citrulline and NO, a potent vasodilator. Alternatively, L-arginine can be metabolized by arginase to form urea and L-ornithine, a precursor to collagen and proline formation important in vascular remodelling. In the current study, we hypothesized that C3H/HeN mice exposed to prolonged hyperoxia would have increased arginase expression and pulmonary vascular wall cell proliferation. C3H/HeN mice were exposed to 14 days of 85% O2 or room air and lung homogenates analyzed by western blot for protein levels of arginase I, arginase II, endothelial NOS (eNOS), ornithine decarboxylase (ODC), ornithine aminotransferase (OAT), and α-smooth muscle actin (α-SMA). Hyperoxia did not change arginase I or eNOS protein levels. However, arginase II protein levels were 15-fold greater after hyperoxia exposure than in lungs exposed to room air. Greater protein levels of ODC and OAT were found in lungs following hyperoxic exposure than in room air animals. α-SMA protein levels were found to be 7-fold greater in the hyperoxia exposed lungs than in room air lungs. In the hyperoxia exposed lungs there was evidence of greater pulmonary vascular wall cell proliferation by α-SMA immunohistochemistry than in room air lungs. Taken together, these data are consistent with a more proliferative vascular phenotype, and may explain the propensity of patients with bronchopulmonary dysplasia to develop pulmonary hypertension.


Asunto(s)
Actinas/metabolismo , Arginasa/biosíntesis , Displasia Broncopulmonar/complicaciones , Displasia Broncopulmonar/metabolismo , Hiperoxia/complicaciones , Animales , Displasia Broncopulmonar/patología , Proliferación Celular , Modelos Animales de Enfermedad , Inducción Enzimática , Pulmón/metabolismo , Pulmón/patología , Ratones , Ornitina Descarboxilasa/metabolismo , Ornitina-Oxo-Ácido Transaminasa/metabolismo
9.
Tumour Biol ; 39(7): 1010428317712512, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28718378

RESUMEN

Glioblastoma is the most common and malignant tumour that occurs primarily in nervous system and has a high morbidity. Research on glioblastoma has recently focused on human cytomegalovirus, belonging to the beta subfamily of Herpesviridae that plays crucial roles in cancer development and progression. This study aimed to investigate the role of human cytomegalovirus-associated microRNA-613 in glioblastoma. In this study, we demonstrate that microRNA-613 expression was frequently reduced in human cytomegalovirus-positive glioblastoma specimens/cells compared with human cytomegalovirus-negative glioblastoma tissue/cells, and a significant correlation was observed between the reduction in microRNA-613 expression and the presence of unfavourable variables, including tumour size (p = 0.0118), World Health Organization stage (p = 0.0169), the overall survival (p = 0.0107) and disease-free (p = 0.0159) survival of patients. Overexpression of microRNA-613 in the glioblastoma cell lines U87 and U251 retarded cell growth and induced cell apoptosis. Upregulation of microRNA-613 inhibited glioblastoma cell clone formation, invasion and migration. Furthermore, we demonstrated that arginase-2 was directly regulated by microRNA-613 and played an essential role in mediating the biological effects of microRNA-613 in glioblastoma. Re-expression of arginase-2 markedly reversed the inhibitory properties of microRNA-613 in glioblastoma cells. Taken together, our data provide compelling evidence that human cytomegalovirus reduced the level of microRNA-613 which functions as an anti-onco-miRNA in glioblastoma, primarily by downregulating the expression of arginase-2.


Asunto(s)
Arginasa/biosíntesis , Citomegalovirus/genética , Glioblastoma/genética , MicroARNs/genética , Adulto , Anciano , Apoptosis/genética , Arginasa/genética , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Citomegalovirus/patogenicidad , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Glioblastoma/virología , Humanos , Estimación de Kaplan-Meier , Masculino , MicroARNs/biosíntesis , Persona de Mediana Edad , Invasividad Neoplásica/genética
10.
Circ Res ; 117(11): 967-77, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26438688

RESUMEN

RATIONALE: Inflammation in post-myocardial infarction (MI) is necessary for myocyte repair and wound healing. Unfortunately, it is also a key component of subsequent heart failure pathology. Transcription factor forkhead box O4 (FoxO4) regulates a variety of biological processes, including inflammation. However, its role in MI remains unknown. OBJECTIVE: To test the hypothesis that FoxO4 promotes early post-MI inflammation via endothelial arginase 1 (Arg1). METHODS AND RESULTS: We induced MI in wild-type and FoxO4(-/-) mice. FoxO4(-/-) mice had a significantly higher post-MI survival, better cardiac function, and reduced infarct size. FoxO4(-/-) hearts had significantly fewer neutrophils, reduced expression of cytokines, and competitive nitric oxide synthase inhibitor Arg1. We generated conditional FoxO4 knockout mice with FoxO4 deleted in cardiac mycoytes or endothelial cells. FoxO4 endothelial cell-specific knockout mice showed significant post-MI improvement of cardiac function and reduction of neutrophil accumulation and cytokine expression, whereas FoxO4 cardiac mycoyte-specific knockout mice had no significant difference in cardiac function and post-MI inflammation from those of control littermates. FoxO4 binds the Foxo-binding site in the Arg1 promoter and activates Arg1 transcription. FoxO4 knockdown in human aortic endothelial cells upregulated nitric oxide on ischemia and suppressed monocyte adhesion that can be reversed by ectopic-expression of Arg1. Furthermore, chemical inhibition of Arg1 in wild-type mice had similar cardioprotection and reduced inflammation after MI as FoxO4 inactivation and administration of nitric oxide synthase inhibitor to FoxO4 KO mice reversed the beneficial effects of FoxO4 deletion on post-MI cardiac function. CONCLUSIONS: FoxO4 activates Arg1 transcription in endothelial cells in response to MI, leading to downregulation of nitric oxide and upregulation of neutrophil infiltration to the infarct area.


Asunto(s)
Arginasa/biosíntesis , Células Endoteliales/enzimología , Factores de Transcripción Forkhead/metabolismo , Inflamación/enzimología , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , Animales , Antiinflamatorios/farmacología , Arginasa/antagonistas & inhibidores , Arginasa/genética , Sitios de Unión , Adhesión Celular , Proteínas de Ciclo Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Inducción Enzimática , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/fisiopatología , Inflamación/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Infiltración Neutrófila , Óxido Nítrico/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Células U937
11.
J Immunol ; 195(11): 5237-50, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491198

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature cells that expand during benign and cancer-associated inflammation and are characterized by their ability to inhibit T cell immunity. Increased metabolism of l-Arginine (l-Arg), through the enzymes arginase 1 and NO synthase 2 (NOS2), is well documented as a major MDSC suppressive mechanism. Therefore, we hypothesized that restricting MDSC uptake of l-Arg is a critical control point to modulate their suppressor activity. Using murine models of prostate-specific inflammation and cancer, we have identified the mechanisms by which extracellular l-Arg is transported into MDSCs. We have shown that MDSCs recruited to localized inflammation and tumor sites upregulate cationic amino acid transporter 2 (Cat2), coordinately with Arg1 and Nos2. Cat2 expression is not induced in MDSCs in peripheral organs. CAT2 contributes to the transport of l-Arg in MDSCs and is an important regulator of MDSC suppressive function. MDSCs that lack CAT2 have significantly reduced suppressive ability ex vivo and display impaired capacity for regulating T cell responses in vivo as evidenced by increased T cell expansion and decreased tumor growth in Cat2(-/-) mice. The abrogation of suppressive function is due to low intracellular l-Arg levels, which leads to the impaired ability of NOS2 to catalyze l-Arg-dependent metabolic processes. Together, these findings demonstrate that CAT2 modulates MDSC function. In the absence of CAT2, MDSCs display diminished capacity for controlling T cell immunity in prostate inflammation and cancer models, where the loss of CAT2 results in enhanced antitumor activity.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Transportador de Aminoácidos Catiônicos 2/biosíntesis , Células Mieloides/inmunología , Neoplasias de la Próstata/inmunología , Linfocitos T/inmunología , Sistemas de Transporte de Aminoácidos Básicos/biosíntesis , Animales , Arginasa/biosíntesis , Arginina/metabolismo , Transporte Biológico , Transportador de Aminoácidos Catiônicos 2/genética , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo
12.
J Immunol ; 194(11): 5520-8, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25926674

RESUMEN

Bacteria colonize cystic fibrosis (CF) airways, and although T cells with appropriate Ag specificity are present in draining lymph nodes, they are conspicuously absent from the lumen. To account for this absence, we hypothesized that polymorphonuclear neutrophils (PMNs), recruited massively into the CF airway lumen and actively exocytosing primary granules, also suppress T cell function therein. Programmed death-ligand 1 (PD-L1), which exerts T cell suppression at a late step, was expressed bimodally on CF airway PMNs, delineating PD-L1(hi) and PD-L1(lo) subsets, whereas healthy control (HC) airway PMNs were uniformly PD-L1(hi). Blood PMNs incubated in CF airway fluid lost PD-L1 over time; in coculture, Ab blockade of PD-L1 failed to inhibit the suppression of T cell proliferation by CF airway PMNs. In contrast with PD-L1, arginase 1 (Arg1), which exerts T cell suppression at an early step, was uniformly high on CF and HC airway PMNs. However, arginase activity was high in CF airway fluid and minimal in HC airway fluid, consistent with the fact that Arg1 activation requires primary granule exocytosis, which occurs in CF, but not HC, airway PMNs. In addition, Arg1 expression on CF airway PMNs correlated negatively with lung function and positively with arginase activity in CF airway fluid. Finally, combined treatment with arginase inhibitor and arginine rescued the suppression of T cell proliferation by CF airway fluid. Thus, Arg1 and PD-L1 are dynamically modulated upon PMN migration into human airways, and, Arg1, but not PD-L1, contributes to early PMN-driven T cell suppression in CF, likely hampering resolution of infection and inflammation.


Asunto(s)
Arginasa/inmunología , Antígeno B7-H1/inmunología , Fibrosis Quística/inmunología , Neutrófilos/inmunología , Linfocitos T/inmunología , Adulto , Apoptosis/inmunología , Arginasa/biosíntesis , Antígeno B7-H1/antagonistas & inhibidores , Proliferación Celular , Exocitosis/inmunología , Femenino , Humanos , Inflamación/inmunología , Inflamación/patología , Pulmón/inmunología , Pulmón/patología , Activación de Linfocitos/inmunología , Masculino , Pruebas de Función Respiratoria , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Adulto Joven
13.
J Immunol ; 195(6): 2560-70, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26246144

RESUMEN

The PI3K signaling cascade in APCs has been recognized as an essential pathway to initiate, maintain, and resolve immune responses. In this study, we demonstrate that a cell type-specific loss of the PI3K antagonist phosphatase and tensin homolog (PTEN) in myeloid cells renders APCs toward a regulatory phenotype. APCs deficient for PTEN exhibit reduced activation of p38 MAPK and reduced expression of T cell-polarizing cytokines. Furthermore, PTEN deficiency leads to upregulation of markers for alternative activation, such as Arginase 1, with concomitant downregulation of inducible NO synthase in APCs in vitro and in vivo. As a result, T cell polarization was dysfunctional in PTEN(-/-) APCs, in particular affecting the Th17 cell subset. Intriguingly, mice with cell type-specific deletions of PTEN-targeting APCs were protected from experimental autoimmune encephalomyelitis, which was accompanied by a pronounced reduction of IL-17- and IL-22-producing autoreactive T cells and reduced CNS influx of classically activated monocytes/macrophages. These observations support the notion that activation of the PI3K signaling cascade promotes regulatory APC properties and suppresses pathogenic T cell polarization, thereby reducing the clinical symptoms and pathology of experimental autoimmune encephalomyelitis.


Asunto(s)
Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Fosfohidrolasa PTEN/genética , Células Th17/inmunología , Animales , Arginasa/biosíntesis , Autoinmunidad/inmunología , Antígeno CD11c/biosíntesis , Diferenciación Celular/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Activación Enzimática/genética , Activación Enzimática/inmunología , Interleucina-17/biosíntesis , Interleucinas/biosíntesis , Activación de Linfocitos , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/inmunología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fragmentos de Péptidos/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Interleucina-22
14.
J Immunol ; 195(6): 2666-74, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26268654

RESUMEN

B lymphopoiesis declines with age, and this decline correlates with increased adipose tissue in the bone marrow (BM). Also, adipocyte-derived factors are known to inhibit B lymphopoiesis. Using cocultures of mouse BM cells with OP9 stromal cells, we found that adipocyte-conditioned medium induces the generation of CD11b(+)Gr1(+) myeloid cells, which inhibit B cell development in vitro. Adipocyte-conditioned medium-induced CD11b(+)Gr1(+) cells express Arg1 (arginase) and Nos2 (inducible NO synthase) and suppress CD4(+) T cell proliferation, indicating that these cells are myeloid-derived suppressor cells (MDSCs). Blocking arginase and inducible NO synthase did not restore B lymphopoiesis, indicating that inhibition is not mediated by these molecules. Transwell and conditioned-medium experiments showed that MDSCs inhibit B lymphopoiesis via soluble factors, and by cytokine array we identified IL-1 as an important factor. Addition of anti-IL-1 Abs restored B lymphopoiesis in BM cultures containing MDSCs, showing that MDSC inhibition of B lymphopoiesis is mediated by IL-1. By treating hematopoietic precursors with IL-1, we found that multipotent progenitors are targets of IL-1. This study uncovers a novel function for MDSCs to inhibit B lymphopoiesis through IL-1. We suggest that inflammaging contributes to a decline of B lymphopoiesis in aged individuals, and furthermore, that MDSCs and IL-1 provide therapeutic targets for restoration of B lymphopoiesis in aged and obese individuals.


Asunto(s)
Adipocitos/citología , Linfocitos B/citología , Inmunosenescencia/inmunología , Interleucina-1/inmunología , Linfopoyesis/efectos de los fármacos , Adipocitos/inmunología , Animales , Anticuerpos/farmacología , Arginasa/biosíntesis , Linfocitos B/inmunología , Células de la Médula Ósea/citología , Antígeno CD11b/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Inflamación/inmunología , Interleucina-1/antagonistas & inhibidores , Interleucina-1/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/inmunología , Células Mieloides/inmunología , Óxido Nítrico Sintasa de Tipo II/biosíntesis
15.
J Immunol ; 194(11): 5211-22, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917103

RESUMEN

Cannabidiol (CBD) is a natural nonpsychotropic cannabinoid from marijuana (Cannabis sativa) with anti-epileptic and anti-inflammatory properties. Effect of CBD on naive immune system is not precisely understood. In this study, we observed that administering CBD into naive mice triggers robust induction of CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSC) in the peritoneum, which expressed functional arginase 1, and potently suppressed T cell proliferation ex vivo. Furthermore, CBD-MDSC suppressed LPS-induced acute inflammatory response upon adoptive transfer in vivo. CBD-induced suppressor cells were comprised of CD11b(+)Ly6-G(+)Ly6-C(+) granulocytic and CD11b(+)Ly6-G(-)Ly6-C(+) monocytic subtypes, with monocytic MDSC exhibiting higher T cell-suppressive function. Induction of MDSC by CBD was markedly attenuated in Kit-mutant (Kit(W/W-v)) mast cell-deficient mice. MDSC response was reconstituted upon transfer of wild-type bone marrow-derived mast cells in Kit(W/W-v) mice, suggesting the key role of cKit (CD117) as well as mast cells. Moreover, mast cell activator compound 48/80 induced significant levels of MDSC in vivo. CBD administration in mice induced G-CSF, CXCL1, and M-CSF, but not GM-CSF. G-CSF was found to play a key role in MDSC mobilization inasmuch as neutralizing G-CSF caused a significant decrease in MDSC. Lastly, CBD enhanced the transcriptional activity of peroxisome proliferator-activated receptor γ in luciferase reporter assay, and PPAR-γ selective antagonist completely inhibited MDSC induction in vivo, suggesting its critical role. Together, the results suggest that CBD may induce activation of PPAR-γ in mast cells leading to secretion of G-CSF and consequent MDSC mobilization. CBD being a major component of Cannabis, our study indicates that marijuana may modulate or dysregulate the immune system by mobilizing MDSC.


Asunto(s)
Antiinflamatorios/farmacología , Cannabidiol/farmacología , Cannabis/metabolismo , Células Mieloides/inmunología , PPAR gamma/genética , Animales , Arginasa/biosíntesis , Antígeno CD11b/metabolismo , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL1/biosíntesis , Femenino , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Mastocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , PPAR gamma/antagonistas & inhibidores , Receptores de Quimiocina/metabolismo , Linfocitos T/inmunología , Activación Transcripcional/genética , p-Metoxi-N-metilfenetilamina/farmacología
16.
J Immunol ; 195(7): 3248-3261, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26311902

RESUMEN

Melioidosis, a severe human disease caused by the bacterium Burkholderia pseudomallei, has a wide spectrum of clinical manifestations ranging from acute septicemia to chronic localized illness or latent infection. Murine models have been widely used to study the pathogenesis of infection and to evaluate novel therapies or vaccines, but how faithfully they recapitulate the biology of human melioidosis at a molecular level is not known. In this study, mice were intranasally infected with either high or low doses of B. pseudomallei to generate either acute, chronic, or latent infection and host blood and tissue transcriptional profiles were generated. Acute infection was accompanied by a homogeneous signature associated with induction of multiple innate immune response pathways, such as IL-10, TREM1, and IFN signaling, largely found in both blood and tissue. The transcriptional profile in blood reflected the heterogeneity of chronic infection and quantitatively reflected the severity of disease. Genes associated with fibrosis and tissue remodeling, including matrix metalloproteases and collagen, were upregulated in chronically infected mice with severe disease. Transcriptional signatures of both acute and chronic melioidosis revealed upregulation of iNOS in tissue, consistent with the expression of IFN-γ, but also Arginase-1, a functional antagonist of the iNOS pathway, and was confirmed by immunohistochemistry. Comparison of these mouse blood datasets by pathway and modular analysis with the blood transcriptional signature of patients with melioidosis showed that many genes were similarly perturbed, including Arginase-1, IL-10, TREM1, and IFN signaling, revealing the common immune response occurring in both mice and humans.


Asunto(s)
Burkholderia pseudomallei/inmunología , Inmunidad Innata/inmunología , Melioidosis/inmunología , Animales , Arginasa/biosíntesis , Arginasa/sangre , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Interferón gamma/biosíntesis , Interferón gamma/sangre , Interferón gamma/inmunología , Interleucina-10/sangre , Interleucina-10/genética , Interleucina-10/inmunología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Metaloproteinasa 9 de la Matriz/sangre , Melioidosis/microbiología , Melioidosis/patología , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Receptores Inmunológicos/sangre , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología , Transcriptoma/genética , Receptor Activador Expresado en Células Mieloides 1
17.
J Neurosci ; 35(44): 14842-60, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26538654

RESUMEN

Tau accumulation remains one of the closest correlates of neuronal loss in Alzheimer's disease. In addition, tau associates with several other neurodegenerative diseases, collectively known as tauopathies, in which clinical phenotypes manifest as cognitive impairment, behavioral disturbances, and motor impairment. Polyamines act as bivalent regulators of cellular function and are involved in numerous biological processes. The regulation of the polyamines system can become dysfunctional during disease states. Arginase 1 (Arg1) and nitric oxide synthases compete for l-arginine to produce either polyamines or nitric oxide, respectively. Herein, we show that overexpression of Arg1 using adeno-associated virus (AAV) in the CNS of rTg4510 tau transgenic mice significantly reduced phospho-tau species and tangle pathology. Sustained Arg1 overexpression decreased several kinases capable of phosphorylating tau, decreased inflammation, and modulated changes in the mammalian target of rapamycin and related proteins, suggesting activation of autophagy. Arg1 overexpression also mitigated hippocampal atrophy in tau transgenic mice. Conversely, conditional deletion of Arg1 in myeloid cells resulted in increased tau accumulation relative to Arg1-sufficient mice after transduction with a recombinant AAV-tau construct. These data suggest that Arg1 and the polyamine pathway may offer novel therapeutic targets for tauopathies.


Asunto(s)
Arginasa/biosíntesis , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Tauopatías/enzimología , Tauopatías/patología , Proteínas tau/metabolismo , Animales , Arginasa/genética , Células HeLa , Hipocampo/enzimología , Hipocampo/patología , Humanos , Ratones , Ratones Transgénicos , Tauopatías/genética , Proteínas tau/genética
18.
Eur J Immunol ; 45(4): 1148-58, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25616156

RESUMEN

Immunosuppressive CD11b(+) Gr-1(+) myeloid-derived suppressor cells (MDSCs) accumulate in the livers of tumor-bearing (TB) mice. We studied hepatic MDSCs in two murine models of immune-mediated hepatitis. Unexpectedly, treatment of TB mice with Concanavalin A (Con A) or α-galactosylceramide resulted in increased alanine aminotransferase (ALT) and aspartate aminotransferase (AST) serum levels in comparison to tumor-free mice. Adoptive transfer of hepatic MDSCs into naïve mice exacerbated Con A induced liver damage. Hepatic CD11b(+) Gr-1(+) cells revealed a polarized proinflammatory gene signature after Con A treatment. An IFN-γ-dependent upregulation of CD40 on hepatic CD11b(+) Gr-1(+) cells along with an upregulation of CD80, CD86, and CD1d after Con A treatment was observed. Con A treatment resulted in a loss of suppressor function by tumor-induced CD11b(+) Gr-1(+) MDSCs as well as enhanced reactive oxygen species (ROS)-mediated hepatotoxicity. CD40 knockdown in hepatic MDSCs led to increased arginase activity upon Con A treatment and lower ALT/AST serum levels. Finally, blockade of arginase activity in Cd40(-/-) tumor-induced myeloid cells resulted in exacerbation of hepatitis and increased ROS production in vivo. Our findings indicate that in a setting of acute hepatitis, tumor-induced hepatic MDSCs act as proinflammatory immune effector cells capable of killing hepatocytes in a CD40-dependent manner.


Asunto(s)
Antígenos CD40/metabolismo , Hepatitis/inmunología , Células Mieloides/inmunología , Traslado Adoptivo , Alanina Transaminasa/sangre , Animales , Antígenos CD1d/biosíntesis , Arginasa/antagonistas & inhibidores , Arginasa/biosíntesis , Arginasa/metabolismo , Aspartato Aminotransferasas/sangre , Antígeno B7-1/biosíntesis , Antígeno B7-2/biosíntesis , Antígeno CD11b/metabolismo , Antígenos CD40/biosíntesis , Antígenos CD40/genética , Línea Celular , Concanavalina A/farmacología , Femenino , Galactosilceramidas/farmacología , Hepatitis/genética , Hepatocitos/inmunología , Hepatocitos/patología , Hígado/citología , Hígado/lesiones , Neoplasias Hepáticas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Mitógenos/farmacología , Células Mieloides/trasplante , Especies Reactivas de Oxígeno/metabolismo , Receptores de Quimiocina/metabolismo
19.
J Immunol ; 193(4): 1690-700, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25009204

RESUMEN

Arginine, a semiessential amino acid implicated in diverse cellular processes, is a substrate for two arginases-Arg1 and Arg2-having different expression patterns and functions. Although appropriately regulated Arg1 expression is critical for immune responses, this has not been documented for Arg2. We show that Arg2 is the dominant enzyme in dendritic cells (DCs) and is repressed by microRNA-155 (miR155) during their maturation. miR155 is known to be strongly induced in various mouse and human DC subsets in response to diverse maturation signals, and miR155-deficient DCs exhibit an impaired ability to induce Ag-specific T cell responses. By means of expression profiling studies, we identified Arg2 mRNA as a novel miR155 target in mouse DCs. Abnormally elevated levels of Arg2 expression and activity were observed in activated miR155-deficient DCs. Conversely, overexpression of miR155 inhibited Arg2 expression. Bioinformatic and functional analyses confirmed that Arg2 mRNA is a direct target of miR155. Finally, in vitro and in vivo functional assays using DCs exhibiting deregulated Arg2 expression indicated that Arg2-mediated arginine depletion in the extracellular milieu impairs T cell proliferation. These results indicate that miR155-induced repression of Arg2 expression is critical for the ability of DCs to drive T cell activation by controlling arginine availability in the extracellular environment.


Asunto(s)
Arginasa/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/enzimología , Activación de Linfocitos/inmunología , MicroARNs/genética , Animales , Arginasa/antagonistas & inhibidores , Arginasa/genética , Arginina/metabolismo , Proliferación Celular , Células Dendríticas/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética
20.
J Immunol ; 193(6): 2801-11, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25127857

RESUMEN

The alternative activation of M2 macrophages in the lungs has been implicated as a causative agent in pulmonary fibrosis; however, the mechanisms underlying M2 polarization are poorly characterized. In this study, we investigated the role of the ubiquitously expressed Src homology domain-containing tyrosine phosphatase Shp2 in this process. Shp2 inactivation augmented IL-4-mediated M2 polarization in vitro, suggesting that Shp2 regulates macrophage skewing and prevents a bias toward the M2 phenotype. Conditional removal of Shp2 in monocytes/macrophages with lysozyme M promoter-driven Cre recombinase caused an IL-4-mediated shift toward M2 polarization. Additionally, an increase in arginase activity was detected in Shp2(∆/∆) mice after i.p. injection of chitin, whereas Shp2-deficient macrophages showed enhanced M2 polarization and protection against schistosome egg-induced schistosomiasis. Furthermore, mutants were more sensitive than control mice to bleomycin-induced inflammation and pulmonary fibrosis. Shp2 was associated with IL-4Rα and inhibited JAK1/STAT6 signaling through its phosphatase activity; loss of Shp2 promoted the association of JAK1 with IL-4Rα, which enhanced IL-4-mediated JAK1/STAT6 activation that resulted in M2 skewing. Taken together, these findings define a role for Shp2 in alveolar macrophages and reveal that Shp2 is required to inhibit the progression of M2-associated pulmonary fibrosis.


Asunto(s)
Activación de Macrófagos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/inmunología , Fibrosis Pulmonar/inmunología , Schistosoma japonicum/inmunología , Esquistosomiasis Japónica/inmunología , Animales , Arginasa/biosíntesis , Bleomicina/efectos adversos , Bleomicina/farmacología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Quitina/farmacología , Inflamación/inducido químicamente , Interleucina-13/biosíntesis , Interleucina-4/biosíntesis , Interleucina-4/inmunología , Interleucina-5/biosíntesis , Janus Quinasa 1/biosíntesis , Pulmón/inmunología , Pulmón/patología , Activación de Macrófagos/genética , Macrófagos Alveolares/inmunología , Ratones , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Fibrosis Pulmonar/inducido químicamente , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Superficie Celular/inmunología , Factor de Transcripción STAT6/biosíntesis , Esquistosomiasis Japónica/parasitología , Transducción de Señal/genética , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA