Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
Más filtros













Intervalo de año de publicación
1.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34058750

RESUMEN

BACKGROUND: The clinical consequences of SARS-CoV-2 and DENGUE virus co-infection are not promising. However, their treatment options are currently unavailable. Current studies have shown that quercetin is both resistant to COVID-19 and DENGUE; this study aimed to evaluate the possible functional roles and underlying mechanisms of action of quercetin as a potential molecular candidate against COVID-19 and DENGUE co-infection. METHODS: We used a series of bioinformatics analyses to understand and characterize the biological functions, pharmacological targets and therapeutic mechanisms of quercetin in COVID-19 and DENGUE co-infection. RESULTS: We revealed the clinical characteristics of COVID-19 and DENGUE, including pathological mechanisms, key inflammatory pathways and possible methods of intervention, 60 overlapping targets related to the co-infection and the drug were identified, the protein-protein interaction (PPI) was constructed and TNFα, CCL-2 and CXCL8 could become potential drug targets. Furthermore, we disclosed the signaling pathways, biological functions and upstream pathway activity of quercetin in COVID-19 and DENGUE. The analysis indicated that quercetin could inhibit cytokines release, alleviate excessive immune responses and eliminate inflammation, through NF-κB, IL-17 and Toll-like receptor signaling pathway. CONCLUSIONS: This study is the first to reveal quercetin as a pharmacological drug for COVID-19 and DENGUE co-infection. COVID-19 and DENGUE co-infection remain a potential threat to the world's public health system. Therefore, we need innovative thinking to provide admissible evidence for quercetin as a potential molecule drug for the treatment of COVID-19 and DENGUE, but the findings have not been verified in actual patients, so further clinical drug trials are needed.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Virus del Dengue/química , Dengue/tratamiento farmacológico , Quercetina/química , SARS-CoV-2/química , COVID-19/complicaciones , COVID-19/genética , COVID-19/virología , Quimiocina CCL2/química , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/genética , Coinfección/tratamiento farmacológico , Coinfección/genética , Coinfección/virología , Dengue/complicaciones , Dengue/genética , Dengue/virología , Virus del Dengue/efectos de los fármacos , Humanos , Interleucina-17/genética , Interleucina-8/química , Interleucina-8/efectos de los fármacos , Interleucina-8/genética , FN-kappa B/efectos de los fármacos , FN-kappa B/genética , Mapas de Interacción de Proteínas/efectos de los fármacos , Quercetina/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
2.
Acta Neurol Scand ; 143(6): 602-607, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33626181

RESUMEN

OBJECTIVE: To identify serum proteins associated with MS and affected by interferon beta treatment. METHODS: Plasma samples from 29 untreated relapsing-remitting MS patients and 15 healthy controls were investigated with a multiplexed panel containing 92 proteins related to inflammation. Follow-up samples were available from 13 patients at 1 and 3 months after initiation of treatment with interferon beta-1a. RESULTS: Ten proteins were differentially expressed in MS patients. Five of these were altered by treatment with IFN-ß 1a: uPA, CX3CL1, CCL2, TRAIL and IL18. CONCLUSION: CCL2 and TRAIL were confirmed to be modulated with interferon beta treatment in MS. As novel findings, we now report that uPA and CX3CL1 were differentially expressed in MS and increased after IFN-beta-1a treatment. Conflicting results have been reported on how interferon beta affects IL-18.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Citocinas/sangre , Citocinas/efectos de los fármacos , Interferón beta-1a/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/sangre , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Adulto , Quimiocina CCL2/sangre , Quimiocina CCL2/efectos de los fármacos , Quimiocina CX3CL1/sangre , Quimiocina CX3CL1/efectos de los fármacos , Femenino , Humanos , Interleucina-18/sangre , Masculino , Proteínas de la Membrana/sangre , Proteínas de la Membrana/efectos de los fármacos , Persona de Mediana Edad , Ligando Inductor de Apoptosis Relacionado con TNF/sangre , Ligando Inductor de Apoptosis Relacionado con TNF/efectos de los fármacos
3.
Biomed Pharmacother ; 137: 111365, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33588264

RESUMEN

BACKGROUND: Defective absorption of acute allergic airway inflammation is involved in the initiation and development of chronic asthma. After allergen exposure, there is a rapid recruitment of macrophages around the airways, which promote acute inflammatory responses. The Ang-(1-7)/Mas receptor axis reportedly plays protective roles in various tissue inflammation and remodeling processes in vivo. However, the exact role of Mas receptor and their underlying mechanisms during the pathology of acute allergic airway inflammation remains unclear. OBJECTIVE: We investigated the role of Mas receptor in acute allergic asthma and explored its underlying mechanisms in vitro, aiming to find critical molecules and signal pathways. METHODS: Mas receptor expression was assessed in ovalbumin (OVA)-induced acute asthmatic murine model. Then we estimated the anti-inflammatory role of Mas receptor in vivo and explored expressions of several known inflammatory cytokines as well as phosphorylation levels of MAPK pathways. Mas receptor functions and underlying mechanisms were studied further in the human bronchial epithelial cell line (16HBE). RESULTS: Mas receptor expression decreased in acute allergic airway inflammation. Multiplex immunofluorescence co-localized Mas receptor and EpCAM, indicated that Mas receptor may function in the bronchial epithelium. Activating Mas receptor through AVE0991 significantly alleviated macrophage infiltration in airway inflammation, accompanied with down-regulation of CCL2 and phosphorylation levels of MAPK pathways. Further studies in 16HBE showed that AVE0991 pre-treatment inhibited LPS-induced or anisomycin-induced CCL2 increase and THP-1 macrophages migration via JNK pathways. CONCLUSION: Our findings suggested that Mas receptor activation significantly attenuated CCL2 dependent macrophage recruitments in acute allergic airway inflammation through JNK pathways, which indicated that Mas receptor, CCL2 and phospho-JNK could be potential targets against allergic airway inflammation.


Asunto(s)
Asma/tratamiento farmacológico , Quimiocina CCL2/efectos de los fármacos , Imidazoles/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteínas Proto-Oncogénicas/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Sistema Respiratorio/patología , Enfermedad Aguda , Angiotensina I , Animales , Asma/inducido químicamente , Asma/patología , Citocinas/metabolismo , Imidazoles/farmacología , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ovalbúmina , Fragmentos de Péptidos , Fosforilación/efectos de los fármacos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/agonistas , Receptores Acoplados a Proteínas G/agonistas
4.
Clin Exp Allergy ; 51(5): 685-695, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33275825

RESUMEN

BACKGROUND: Neuromuscular-blocking agents (NMBAs) can cause both IgE-dependent and IgE-independent anaphylactic reactions, with activation of the mast cell receptor MRGPRX2 being important to the latter. Sugammadex, a reversal agent for certain aminosteroid NMBAs, has been proposed as an antidote for these anaphylactic events with conflicting outcomes. OBJECTIVE: We further characterize the involvement of MRGPRX2 in NMBA-induced mast cell activation and determine how this is influenced by sugammadex. We then apply these in vitro results to infer the possible utility of sugammadex in the acute management of non-IgE-dependent anaphylaxis. METHODS: The LAD2 human mast cell line and a MRGPRX2 knock-down derivative were used to validate the involvement of MRGPRX2 and to test the effect of sugammadex on mast cell activation by NMBAs and other MRGPRX2 agonists. RESULTS: All MRGPRX2 agonists tested were shown to induce MRGPRX2-dependent LAD2 mast cell calcium mobilization and cytokine release and all, apart from rocuronium, induced degranulation. Co-treatment of mast cells with sugammadex and some MRGPRX2 agonists significantly reduced cell activation, but if sugammadex was administered a few minutes following stimulation, degranulation was not attenuated. However, addition of sugammadex up to 180 min following LAD2 MRGPRX2 stimulation, significantly reduced CCL2 mRNA and protein induction. CONCLUSIONS AND CLINICAL RELEVANCE: We show that sugammadex, known to reverse muscle blockade by certain NMBAs, is also able to reduce MRGPRX2 activation by NMBAs and other, but not all, MRGPRX2 agonists. As sugammadex was ineffective in attenuating mast cell degranulation when added rapidly post MRGPRX2 activation, this suggests against the agent having efficacy in controlling acute symptoms of anaphylaxis to NMBAs caused by MRGPRX2 activation. Interestingly, however, sugammadex did impair MRGPRX2-induced CCL2 release, suggesting that it may have some benefit in perhaps dampening less well-defined adverse effects of MRGPRX2-dependent anaphylaxis associated with the more slowly elaborated mast cell mediators.


Asunto(s)
Anafilaxia/tratamiento farmacológico , Quimiocina CCL2/efectos de los fármacos , Mastocitos/efectos de los fármacos , Proteínas del Tejido Nervioso/efectos de los fármacos , Bloqueantes Neuromusculares/farmacología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Neuropéptido/efectos de los fármacos , Sugammadex/farmacología , Anafilaxia/inducido químicamente , Antídotos/farmacología , Atracurio/efectos adversos , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Técnicas In Vitro , Mastocitos/inmunología , Mastocitos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Bloqueantes Neuromusculares/efectos adversos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/metabolismo , Rocuronio/efectos adversos
5.
Osteoarthritis Cartilage ; 29(3): 402-412, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33227437

RESUMEN

OBJECTIVE: Cellular senescence is a phenotypic state characterized by stable cell-cycle arrest, enhanced lysosomal activity, and the secretion of inflammatory molecules and matrix degrading enzymes. Senescence has been implicated in osteoarthritis (OA) pathophysiology; however, the mechanisms that drive senescence induction in cartilage and other joint tissues are unknown. While numerous physiological signals are capable of initiating senescence, one emerging theme is that damaged cells convert to senescence in response to sustained mitogenic stimulation. The goal of this study was to develop an in vitro articular cartilage explant model to investigate the mechanisms of senescence induction. DESIGN: This study utilized healthy cartilage derived from cadaveric equine stifles and human ankles. Explants were irradiated to initiate DNA damage, and mitogenic stimulation was provided through serum-containing medium and treatment with transforming growth factor ß1 and basic fibroblastic growth factor. Readouts of senescence were a quantitative flow cytometry assay to detect senescence-associated ß galactosidase activity (SA-ß-gal), immunofluorescence for p16 and γH2AX, and qPCR for the expression of inflammatory genes. RESULTS: Human cartilage explants required both irradiation and mitogenic stimulation to induce senescence as compared to baseline control conditions (7.16% vs 2.34% SA-ß-gal high, p = 0.0007). These conditions also resulted in chondrocyte clusters within explants, a persistent DNA damage response, increased p16, and gene expression changes. CONCLUSIONS: Treatment of cartilage explants with mitogenic stimuli in the context of cellular damage reliably induces high levels of SA-ß-gal activity and other senescence markers, which provides a physiologically relevant model system to investigate the mechanisms of senescence induction.


Asunto(s)
Cartílago Articular/metabolismo , Senescencia Celular/genética , Condrocitos/metabolismo , Animales , Articulación del Tobillo , Cartílago Articular/citología , Cartílago Articular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/genética , Condrocitos/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica/efectos de los fármacos , Histonas/efectos de los fármacos , Histonas/metabolismo , Caballos , Humanos , Técnicas In Vitro , Inflamación/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/efectos de los fármacos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Interleucina-6/genética , Metaloproteinasa 13 de la Matriz/efectos de los fármacos , Metaloproteinasa 13 de la Matriz/genética , Mitógenos/farmacología , Rodilla de Cuadrúpedos , Factor de Crecimiento Transformador beta1/farmacología , beta-Galactosidasa/efectos de los fármacos , beta-Galactosidasa/metabolismo
6.
Aging (Albany NY) ; 12(24): 26121-26139, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-33373319

RESUMEN

Isoflurane (ISO) elicits protective effects on ischemia-induced brain injury. We investigated whether sub-anesthetic (0.7%) ISO post-conditioning attenuates the inflammation and apoptosis in oxygen-glucose deprivation (OGD)-insulted co-cultures (microglia and neurons) in vitro and the brain injury of the middle cerebral arterial occlusion (MCAO) rat. We demonstrated that ISO augmented the viability of OGD-treated microglia and neurons. ISO reduced the expression and activation of COX2 and iNOS in OGD-challenged microglia. ISO repressed the production of tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, IL-8, and monocyte chemoattractant protein-1 in OGD-exposed microglia. ISO also decreased nucleosomal fragmentation and caspase-3 activity but increased mitochondrial membrane potential in OGD-stimulated microglia and neurons. Mechanistically, ISO suppressed OGD-induced microglial inflammation by blocking ROS-regulated p38 MAPK/NF-κB signaling pathway and hampered OGD-triggered microglial apoptosis in a ROS- or NO-dependent fashion. In vivo results with MCAO rats were partly consistent with the in vitro observation. These findings indicate that sub-anesthetic ISO post-conditioning abates the inflammation and apoptosis in OGD-stimulated rat microglia and the apoptosis of OGD-exposed neurons and the brain injuries of MCAO rats, suggesting it as a potentially effective therapeutic approach for ischemic brain damages.


Asunto(s)
Anestésicos por Inhalación/farmacología , Apoptosis/efectos de los fármacos , Isquemia Encefálica/metabolismo , Isoflurano/farmacología , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/metabolismo , Técnicas de Cocultivo , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Inflamación/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucina-8/efectos de los fármacos , Interleucina-8/metabolismo , Microglía/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo II , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
J Am Heart Assoc ; 9(24): e018418, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33283594

RESUMEN

Background Venous neointimal hyperplasia and venous stenosis (VS) formation can result in a decrease in arteriovenous fistula (AVF) patency in patients with end-stage renal disease. There are limited therapies that prevent VNH/VS. Systemic delivery of simvastatin has been shown to reduce VNH/VS but local delivery may help decrease the side effects associated with statin use. We determined if microparticles (MP) composed of cyclodextrins loaded with simvastatin (MP-SV) could reduce VS/VNH using a murine arteriovenous fistula model with chronic kidney disease. Methods and Results Male C57BL/6J mice underwent nephrectomy to induce chronic kidney disease. Four weeks later, an arteriovenous fistula was placed and animals were randomized to 3 groups: 20 µL of PBS or 20 µL of PBS with 16.6 mg/mL of either MP or MP-SV. Animals were euthanized 3 days later and the outflow veins were harvested for quantitative reverse transcriptase-polymerase chain reaction analysis and 28 days later for immunohistochemistical staining with morphometric analysis. Doppler ultrasound was performed weekly. Gene expression of vascular endothelial growth factor-A (Vegf-A), matrix metalloproteinase-9 (Mmp-9), transforming growth factor beta 1 (Tgf-ß1), and monocyte chemoattractant protein-1 (Mcp-1) were significantly decreased in MP-SV treated vessels compared with controls. There was a significant decrease in the neointimal area, cell proliferation, inflammation, and fibrosis, with an increase in apoptosis and peak velocity in MP-SV treated outflow veins. MP-SV treated fibroblasts when exposed to hypoxic injury had decreased gene expression of Vegf-A and Mmp-9. Conclusions In experimental arteriovenous fistulas, periadventitial delivery of MP-SV decreased gene expression of Vegf-A, Mmp-9, Tgf-ß1 and Mcp-1, VNH/VS, inflammation, and fibrosis.


Asunto(s)
Fístula Arteriovenosa/patología , Hiperplasia/prevención & control , Neointima/patología , Simvastatina/uso terapéutico , Animales , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/efectos adversos , Anticolesterolemiantes/uso terapéutico , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/metabolismo , Ciclodextrinas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Fibrosis/metabolismo , Oclusión de Injerto Vascular/prevención & control , Hiperplasia/etiología , Inflamación/metabolismo , Fallo Renal Crónico/terapia , Masculino , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Simvastatina/administración & dosificación , Simvastatina/efectos adversos , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Remodelación Vascular/efectos de los fármacos , Venas/metabolismo
8.
Eur J Pharmacol ; 885: 173505, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32861662

RESUMEN

Pharmacological modulation of the Janus kinase (JAK) family has achieved clinically meaningful therapeutic outcomes for the treatment of inflammatory and hematopoietic diseases. Several JAK1 selective compounds are being investigated clinically to determine their anti-inflammatory potential. We used recombinant enzymes and primary human lymphocytes to assess the JAK1 specificity of itacitinib (INCB039110) and study inhibition of signal transducers and activators of transcription (STAT) signaling. Rodent models of arthritis and inflammatory bowel disease were subsequently explored to elucidate the efficacy of orally administered itacitinib on inflammatory pathogenesis. Itacitinib is a potent and selective JAK1 inhibitor when profiled against the other JAK family members. Upon oral administration in rodents, itacitinib achieved dose-dependent pharmacokinetic exposures that highly correlated with STAT3 pharmacodynamic pathway inhibition. Itacitinib ameliorated symptoms and pathology of established experimentally-induced arthritis in a dose-dependent manner. Furthermore, itacitinib effectively delayed disease onset, reduced symptom severity, and accelerated recovery in three distinct mouse models of inflammatory bowel disease. Low dose itacitinib administered via cannula directly into the colon was highly efficacious in TNBS-induced colitis but with minimal systemic drug exposure, suggesting localized JAK1 inhibition is sufficient for disease amelioration. Itacitinib treatment in an acute graft-versus-host disease (GvHD) model rapidly reduced inflammatory markers within lymphocytes and target tissue, resulting in a marked improvement in disease symptoms. This is the first manuscript describing itacitinib as a potent and selective JAK1 inhibitor with anti-inflammatory activity across multiple preclinical disease models. These data support the scientific rationale for ongoing clinical trials studying itacitinib in select GvHD patient populations.


Asunto(s)
Azetidinas/farmacología , Inflamación/tratamiento farmacológico , Ácidos Isonicotínicos/farmacología , Janus Quinasa 1/antagonistas & inhibidores , Animales , Artritis Experimental/tratamiento farmacológico , Azetidinas/farmacocinética , Azetidinas/uso terapéutico , Quimiocina CCL2/efectos de los fármacos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Ácidos Isonicotínicos/farmacocinética , Ácidos Isonicotínicos/uso terapéutico , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Cultivo Primario de Células , Ratas , Ratas Endogámicas Lew , Factores de Transcripción STAT/efectos de los fármacos , Factor de Transcripción STAT3/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos
9.
Eur J Pharmacol ; 886: 173445, 2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-32758571

RESUMEN

The incidence of colon cancer increased worldwide in 2019 and its treatment is urgent from a quality of life perspective. A relationship has been reported between elevated numbers of tumor-associated macrophages (TAMs) in the tumor microenvironment and a poor prognosis in cancer patients, and M2 TAMs have been shown to promote tumor growth by immunosuppression through the stimulation of programmed death-1 (PD-1, an immune check point receptor), interleukin (IL)-1ß, and monocyte chemoattractant protein (MCP)-1. We herein examined the effects of three synthetic dihydroxystilbenes (2,3-, 3,4-, and 4,4'-dihydroxystilbenes) on colon carcinogenesis, colon tumor growth, and colon cytokines (IL-1ß, IL-6, and tumor necrosis factor (TNF)-α), a chemokine (MCP-1), vascular endothelial growth factor (VEGF), and PD-1 levels in azoxymethane (AOM) plus dextran sulfate sodium (DSS)-treated C57BL/6J mice. The three dihydroxystilbenes inhibited colon carcinogenesis and tumor growth as well as increases in colon IL-1ß, IL-6, MCP-1, and PD-1 levels in AOM/DDS-treated mice (in vivo). The three dihydroxystilbenes also suppressed COX-2 expression in colon tumors (in vivo). The results obtained also revealed that the three dihydroxystilbenes inhibited PD-1 elevations in M2-THP-1 macrophages (in vitro). Therefore, the inhibition of AOM/DSS-induced colon carcinogenesis and colon tumor growth by 2,3-, 3,4-, and 4,4'-dihydroxystilbenes appears to be due to the suppression of M2 TAM differentiation and activation and PD-1 expression (immunosuppression) via reductions in COX-2 expression levels in the colon tumor microenvironment.


Asunto(s)
Apoptosis/efectos de los fármacos , Quimiocinas/antagonistas & inhibidores , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/prevención & control , Citocinas/antagonistas & inhibidores , Dihidrostilbenoides/uso terapéutico , Animales , Azoximetano , Carcinógenos/antagonistas & inhibidores , Quimiocina CCL2/efectos de los fármacos , Neoplasias del Colon/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Sulfato de Dextran , Dihidrostilbenoides/síntesis química , Dihidrostilbenoides/química , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral/efectos de los fármacos
10.
Basic Res Cardiol ; 115(3): 34, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32323032

RESUMEN

Endocannabinoids are important lipid-signaling mediators. Both protective and deleterious effects of endocannabinoids in the cardiovascular system have been reported but the mechanistic basis for these contradicting observations is unclear. We set out to identify anti-inflammatory mechanisms of endocannabinoids in the murine aorta and in human vascular smooth muscle cells (hVSMC). In response to combined stimulation with cytokines, IL-1ß and TNFα, the murine aorta released several endocannabinoids, with anandamide (AEA) levels being the most significantly increased. AEA pretreatment had profound effects on cytokine-induced gene expression in hVSMC and murine aorta. As revealed by RNA-Seq analysis, the induction of a subset of 21 inflammatory target genes, including the important cytokine CCL2 was blocked by AEA. This effect was not mediated through AEA-dependent interference of the AP-1 or NF-κB pathways but rather through an epigenetic mechanism. In the presence of AEA, ATAC-Seq analysis and chromatin-immunoprecipitations revealed that CCL2 induction was blocked due to increased levels of H3K27me3 and a decrease of H3K27ac leading to compacted chromatin structure in the CCL2 promoter. These effects were mediated by recruitment of HDAC4 and the nuclear corepressor NCoR1 to the CCL2 promoter. This study therefore establishes a novel anti-inflammatory mechanism for the endogenous endocannabinoid AEA in vascular smooth muscle cells. Furthermore, this work provides a link between endogenous endocannabinoid signaling and epigenetic regulation.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Quimiocina CCL2/biosíntesis , Endocannabinoides/metabolismo , Músculo Liso Vascular/metabolismo , Alcamidas Poliinsaturadas/metabolismo , Animales , Antiinflamatorios/farmacología , Ácidos Araquidónicos/farmacología , Quimiocina CCL2/efectos de los fármacos , Endocannabinoides/farmacología , Epigénesis Genética/efectos de los fármacos , Humanos , Ratones , Músculo Liso Vascular/efectos de los fármacos , Alcamidas Poliinsaturadas/farmacología , Transducción de Señal/efectos de los fármacos
11.
J Am Soc Nephrol ; 31(3): 560-577, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31996409

RESUMEN

BACKGROUND: Prolyl hydroxylase domain (PHD) inhibitors, which stimulate erythropoietin production through the activation of hypoxia-inducible factor (HIF), are novel therapeutic agents used for treating renal anemia. Several PHD inhibitors, including enarodustat, are currently undergoing phase 2 or phase 3 clinical trials. Because HIF regulates a broad spectrum of genes, PHD inhibitors are expected to have other effects in addition to erythropoiesis, such as protection against metabolic disorders. However, whether such beneficial effects would extend to metabolic disorder-related kidney disease is largely unknown. METHODS: We administered enarodustat or vehicle without enarodustat in feed to diabetic black and tan brachyury (BTBR) ob/ob mice from 4 to 22 weeks of age. To elucidate molecular changes induced by enarodustat, we performed transcriptome analysis of isolated glomeruli and in vitro experiments using murine mesangial cells. RESULTS: Compared with BTBR ob/ob mice that received only vehicle, BTBR ob/ob mice treated with enarodustat displayed lower body weight, reduced blood glucose levels with improved insulin sensitivity, lower total cholesterol levels, higher adiponectin levels, and less adipose tissue, as well as a tendency for lower macrophage infiltration. Enarodustat-treated mice also exhibited reduced albuminuria and amelioration of glomerular epithelial and endothelial damage. Transcriptome analysis of isolated glomeruli revealed reduced expression of C-C motif chemokine ligand 2/monocyte chemoattractant protein-1 (CCL2/MCP-1) in enarodustat-treated mice compared with the vehicle-only group, accompanied by reduced glomerular macrophage infiltration. In vitro experiments demonstrated that both local HIF-1 activation and restoration of adiponectin by enarodustat contributed to CCL2/MCP-1 reduction in mesangial cells. CONCLUSIONS: These results indicate that the PHD inhibitor enarodustat has potential renoprotective effects in addition to its potential to protect against metabolic disorders.


Asunto(s)
Quimiocina CCL2/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Nefropatías Diabéticas/prevención & control , Inhibidores de Prolil-Hidroxilasa/farmacología , Animales , Quimiocina CCL2/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Resistencia a la Insulina , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/prevención & control , Ratones , Ratones Obesos , Glicinas N-Sustituídas/farmacología , Prolil Hidroxilasas/metabolismo , Piridinas/farmacología , Distribución Aleatoria , Valores de Referencia , Resultado del Tratamiento , Triazoles/farmacología
12.
Cartilage ; 11(1): 88-97, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-29808705

RESUMEN

OBJECTIVE: Cetylated fatty acids are a group of naturally occurring fats of plant and/or animal origin. Cetyl myristoleate, in particular, was initially involved in osteoarthritis related research as its therapeutic administration prevented experimentally induced arthritis in Swiss Albino mice. In this context, the aim of our study was to investigate the possible mechanisms of Celadrin cetylated fatty acids action at the cellular level inflammation related pain relief and chondrogenesis. DESIGN: For this, we tested the effects of the cetylated fatty acids mixture from Celadrin on an in vitro scaffold-free 3-dimensional mesenchymal stem cells culture model of chondrogenesis. Furthermore, we treated stimulated mouse macrophage cells with the cetylated fatty acids mixture to investigate the expression profile of secreted inflammatory cytokines. RESULTS: The cetylated fatty acids mixture from Celadrin significantly decreased the production of IL-6, MCP-1, and TNF, key regulators of the inflammatory process, in stimulated RAW264.7 mouse macrophage cells. The treatment with cetylated fatty acids mixture initiated and propagated the process of chondrogenesis as demonstrated by the increased expression and deposition of chondrogenic markers by the differentiating mesenchymal cells. CONCLUSION: The cetylated fatty acids mixture from Celadrin reduces inflammation in vitro by significantly decreasing the expression of IL-6, MCP-1, and TNF in stimulated RAW264.7 mouse macrophage cells. These compounds facilitate the chondrogenic differentiation process of human adipose-derived stem cells by stimulating the expression of chondrogenic markers under chondrogenic induction conditions.


Asunto(s)
Acetilación , Condrogénesis/efectos de los fármacos , Ácidos Grasos/farmacología , Osteoartritis/tratamiento farmacológico , Ceras/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL2/efectos de los fármacos , Ácidos Grasos/química , Humanos , Inflamación , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/efectos de los fármacos
13.
Planta Med ; 85(5): 406-411, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30609436

RESUMEN

Catalpol, an iridoid glycoside, is an isolated natural product of Rehmannia glutinosa, which has been reported to have antidiabetic properties. This study investigated the vascular protective effects of catalpol in hyperglycemic rats with balloon-injured carotid arteries. Balloon injury stress led to the upregulation of monocyte chemoattractant protein-1 expression in rats with streptozotocin-induced diabetes. Western blotting and real-time PCR were performed. In situ hybridization, immunohistochemistry, and confocal analyses were employed. Monocyte chemoattractant protein-1 levels were increased through streptozotocin induction or balloon injury. After treatment with catalpol, the neointimal hyperplasia area was reduced 2 weeks after balloon injury in hyperglycemic rats. Real-time PCR and immunohistochemical analysis demonstrated reduced levels of monocyte chemoattractant protein-1 2 weeks after the balloon injury. Monocyte chemoattractant protein-1 expression was significantly increased in balloon-injured rats compared with the control groups. Thus, treatment with catalpol affected monocyte chemoattractant protein-1 expression. This study demonstrated that catalpol downregulated monocyte chemoattractant protein-1 expression in carotid arteries and ameliorated neointimal hyperplasia in hyperglycemic rats. The suppressive effect of monocyte chemoattractant protein-1 suggests that it plays a key role in neointimal hyperplasia. The results imply that catalpol is potentially effective for preventing hyperglycemia-related ischemic cardiac diseases.


Asunto(s)
Quimiocina CCL2/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Hipoglucemiantes/farmacología , Glucósidos Iridoides/farmacología , Neointima/patología , Rehmannia/química , Animales , Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/patología , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/genética , Modelos Animales de Enfermedad , Hiperglucemia/complicaciones , Hiperplasia/tratamiento farmacológico , Masculino , Isquemia Miocárdica/etiología , Isquemia Miocárdica/prevención & control , Ratas , Ratas Wistar , Estreptozocina
14.
BMC Res Notes ; 12(1): 59, 2019 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683148

RESUMEN

OBJECTIVES: The purpose of this study was to evaluate the effect of gabapentin on Ehrlich tumor growth in Swiss mice, a highly aggressive and inflammatory tumor model. Mice were grouped into sets of 5 animals and treated from days 2 to 8 with gabapentin 30 mg/kg body weight (G30) or 100 mg/kg body weight (G100), or normal sterile saline (control). RESULTS: The mice were euthanized on day 10. Tumor growth, tumoricidal agents and inflammatory cytokines levels were assessed. At day 10, G30 and G100 mice gained weight, but there were no differences in tumor cell count or in ascites volume. In G100, there was a reduction in arginase and an increase in SOD activities. There was an increase in IL-6 and MCP-1 levels, especially in G100, but no alterations in TNF-α. There was no direct evidence of tumor induction by gabapentin. However, the findings suggest that its use modulates immune response to a more effector and less deleterious profile, with increase in activity of anti-oxidant enzymes and in cytokines that favor activation of macrophages, which could improve the general status of the tumor host.


Asunto(s)
Analgésicos/farmacología , Arginasa/efectos de los fármacos , Neoplasias de la Mama , Carcinoma de Ehrlich , Quimiocina CCL2/efectos de los fármacos , Gabapentina/farmacología , Interleucina-6 , Superóxido Dismutasa/efectos de los fármacos , Aumento de Peso/efectos de los fármacos , Analgésicos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Femenino , Gabapentina/administración & dosificación , Ratones
15.
Clin Exp Hypertens ; 41(3): 220-230, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29672166

RESUMEN

OBJECTIVE: This study was focused on screening leech extracts to identify those with little or no anti-coagulation effect or with significant anti-endothelial dysfunction activity. METHODS: Different leech extracts were prepared by enzymolysis and microbial transformation and their cytotoxicity were measured by MTT assay. The effect of different leech extracts on mRNA expression of coagulation-related factors (PAI, vWF, tPA, PS, TFPI, TM) was quantified by RT-PCR. After identifying a leech extract with little anti-coagulatory effect, RT-PCR was then used to assess the effect of this extract on the mRNA expression of endothelial dysfunction-related molecules (ET-1, iNOS, MCP-1, IL-6). RESULTS: 8 leech extracts were obtained, including 4 enzymatic extracts (LP, PHL, PTHL, CEHL) and 4 Lactobacillus metabolites (MRS, MRS-1, MRS-2, and MRS-3). Following optimization of conditions using MTT assays, we treated EA.hy926 cells with 0, 12.5, 25, 50 µg/mL of LP, PTHL, CEHL, MRS, MRS-1 or MRS-3 extract for 24 h. We found that PHL and MRS-1 had no significant effect on coagulation-related factors. Furthermore, treatment with 50 µg/mL PHL resulted in significant decreases in ET-1, iNOS, MCP-1, and IL-6 mRNA expression by 28.06%, 33.30%, 19.80%, and 52.34%, respectively. CONCLUSIONS: In the present study, we found that PHL, a pepsin hydrolysate of leech with little anti-coagulatory effect, could significantly suppress TNF-α induced mRNA overexpression of endothelial dysfunction-related molecules (ET-1, iNOS, MCP-1, and IL-6). These results provide a reliable experimental basis for identifying new anti-atherosclerosis therapeutics for long term use and with minimal bleeding side effects.


Asunto(s)
Aterosclerosis/sangre , Coagulación Sanguínea/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Enzimas/farmacología , Sanguijuelas , Extractos de Tejidos/farmacología , Animales , Factores de Coagulación Sanguínea/efectos de los fármacos , Factores de Coagulación Sanguínea/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/metabolismo , Células Endoteliales/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
16.
Eur J Pharmacol ; 845: 91-98, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30287151

RESUMEN

Vitamin D has been suggested to harbor multiple biological activities, among them the potential of vitamin D in the protection of diabetic nephropathy (DN) has attracted special attention. Both animal studies and clinical trials have documented an inverse correlation between low vitamin D levels and DN risk, and supplementation with vitamin D or its active derivatives has been demonstrated to improve endothelial cell injury, reduce proteinuria, attenuate renal fibrosis, and resultantly retard DN progression. Vitamin D exerts its pharmacological effects primarily via vitamin D receptor, whose activation inhibits the renin-angiotensin system, a key culprit for DN under hyperglycemia. The anti-DN benefit of vitamin D can be enhanced when administrated in combination with angiotensin converting enzyme inhibitors or angiotensin II receptor blockers. Mechanistic studies reveal that pathways relevant to inflammation participate in the pathogenesis of DN, however, consumption of vitamin D-related products negatively regulates inflammatory response at multiple levels, indicated by inhibiting macrophage infiltration, nuclear factor-kappa B (NF-κB) activation, and production of such inflammatory mediators as transforming growth factor-ß(TGF-ß), monocyte chemoattractant protein 1(MCP-1), and regulated upon activation normal T cell expressed and secreted protein(RANTES). The robust anti-inflammatory property of vitamin D-related products allows them with a promising renoprotective therapeutic option for DN. This review summarizes new advances in our understanding of vitamin D-related products in the DN management.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Proteinuria/tratamiento farmacológico , Receptores de Calcitriol/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Vitamina D/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL5/efectos de los fármacos , Quimioterapia Combinada , Células Endoteliales/patología , Humanos , FN-kappa B/efectos de los fármacos , Sustancias Protectoras/farmacología , Factor de Crecimiento Transformador beta/efectos de los fármacos , Vitamina D/farmacología
17.
Zhonghua Zhong Liu Za Zhi ; 40(10): 744-749, 2018 Oct 23.
Artículo en Chino | MEDLINE | ID: mdl-30392338

RESUMEN

Objective: To investigate the effect of tumor-associated macrophages on the stemness of esophageal cancer cells and the potential mechanism of antiproliferative effects of aspirin (ASA). Methods: The effects of aspirin on the stemness characteristics of KYSE-450 cells and KYSE-450 cells co-cultured with M2 macrophages (KYSE-450+ M2) were performed using spheroid formation assay. After treatment with aspirin, the expression of different chemokines, the core pluripotency gene Nanog and the stem cell marker CD90 in different cell groups were determined by real-time quantitative PCR, flow cytometry and Western blot. Results: The number of spheres formed in the ASA and KYSE-450+ M2 cell groups were 7.00±1.23 and 34.33±2.33, respectively, showing statistically significant difference compared with that of control group (14.50±2.33, all P<0.05). The number of spheres in KYSE-450+ M2+ ASA cell group were 20.67±2.33, which was significantly lower than that of KYSE-450+ M2 group (P<0.05). The expression levels of Nanog gene in control and ASA groups were 1.00 and 0.50±0.10, respectively, and the difference was statistically significant (P<0.05). Moreover, the expression of Nanog gene in cells of KYSE-450+ M2 group and M2+ KYSE-450+ ASA group was 1.74±0.13 and 1.43±0.05, showing statistically significant difference (P<0.05). When chemokine CCL2 was knocked down, the levels of Nanog gene in M2+ shCCL2-KYSE450+ ASA group and M2+ shCCL2-KYSE450 group were decreased to 1.22±0.11 and 1.17±0.08, respectively, and there was no statistically significant difference between them (P=0.69). Flow cytometry analyses showed that the expression levels of CD90 in control and ASA cells were (2.93±0.52)% and (1.30±0.17)%, respectively, and the difference was statistically significant (P<0.05). Moreover, the expression levels of CD90 in M2+ shCCL2-KYSE450 cells and M2+ shCCL2-KYSE450+ ASA cells were (4.07±0.12)% and (4.73±0.38)%, respectively, showing no statistically significant difference (P=0.17). Conclusions: Tumor-associated macrophages enhances the stemness of esophageal cancer cells, whereas aspirin attenuates the stemness by suppressing the expression of CCL2. Aspirin plays an anti-tumor effect in esophageal cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Aspirina/farmacología , Quimiocina CCL2/efectos de los fármacos , Regulación hacia Abajo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Macrófagos/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Proteína Homeótica Nanog/genética , Células Madre Pluripotentes/citología , Esferoides Celulares , Antígenos Thy-1/genética , Células Tumorales Cultivadas
18.
Braz J Med Biol Res ; 51(11): e7655, 2018 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-30328934

RESUMEN

Previous studies have indicated that propofol has immunomodulatory and antioxidative properties. However, the renoprotection effect and the precise mechanisms of propofol in sepsis-induced renal injury remain unclear. The purpose of the present study was to investigate the role of miR-290-5p/CCL-2 signaling in septic mice treatment with propofol. Mice were treated with propofol (50 mg/kg) twice within 24 h. Survival outcome was monitored within 48 h. The mRNA and protein levels were assayed by qRT-PCR and western blotting, respectively. Mouse podocytes (MPC5) were treated with lipopolysaccharide (LPS) to establish the cell model in vitro. The proliferation of MPC5 was monitored using the MTS assay. Cell apoptosis was analyzed by flow cytometry. Propofol improved survival outcome and alleviated acute kidney injury in cecal ligation and puncture-operated mice. Propofol increased miR-290-5p expression and decreased CCL-2 and inflammatory cytokines levels in the kidney for septic mice. We found that miR-290-5p was a direct regulator of CCL-2 in MPC5. Propofol could abrogate LPS-induced growth inhibition and apoptosis in MPC5. Meanwhile, propofol inhibited CCL-2 expression in LPS-treated MPC5, however, knockdown of miR-290-5p abrogated the inhibitory effect propofol on the mRNA and protein expressions of CCL-2. Propofol could serve as an effective therapeutic medication to suppress sepsis-induced renal injury in vivo and in vitro by regulating the miR-290-5p/CCL-2 signaling pathway.


Asunto(s)
Lesión Renal Aguda/prevención & control , Quimiocina CCL2/efectos de los fármacos , MicroARNs/efectos de los fármacos , Propofol/farmacología , Sepsis/complicaciones , Transducción de Señal/efectos de los fármacos , Lesión Renal Aguda/etiología , Animales , Western Blotting , Quimiocina CCL2/metabolismo , Citometría de Flujo , Masculino , Ratones , MicroARNs/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sepsis/metabolismo
19.
Am J Obstet Gynecol ; 219(1): 113.e1-113.e9, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29709511

RESUMEN

BACKGROUND: Maternal inflammation is a risk factor for neonatal brain injury and future neurological deficits. Pomegranates have been shown to exhibit anti-inflammatory, anti-apoptotic and anti-oxidant activities. OBJECTIVE: We hypothesized that pomegranate juice (POM) may attenuate fetal brain injury in a rat model of maternal inflammation. STUDY DESIGN: Pregnant rats (24 total) were randomized for intraperitoneal lipopolysaccharide (100 µg/kg) or saline at time 0 at 18 days of gestation. From day 11 of gestation, 12 dams were provided ad libitum access to drinking water, and 12 dams were provided ad libitum access to drinking water with pomegranate juice (5 mL per day), resulting in 4 groups of 6 dams (saline/saline, pomegranate juice/saline, saline/lipopolysaccharide, pomegranate juice/lipopolysaccharide). All dams were sacrificed 4 hours following the injection and maternal blood and fetal brains were collected from the 4 treatment groups. Maternal interleukin-6 serum levels and fetal brain caspase 3 active form, nuclear factor-κB p65, neuronal nitric oxide synthase (phosphoneuronal nitric oxide synthase), and proinflammatory cytokine levels were determined by enzyme-linked immunosorbent assay and Western blot. RESULTS: Maternal lipopolysaccharide significantly increased maternal serum interleukin-6 levels (6039 ± 1039 vs 66 ± 46 pg/mL; P < .05) and fetal brain caspase 3 active form, nuclear factor-κB p65, phosphoneuronal nitric oxide synthase, and the proinflammatory cytokines compared to the control group (caspase 3 active form 0.26 ± 0.01 vs 0.20 ± 0.01 U; nuclear factor-κB p65 0.24 ± 0.01 vs 0.1 ± 0.01 U; phosphoneuronal nitric oxide synthase 0.23 ± 0.01 vs 0.11 ± 0.01 U; interleukin-6 0.25 ± 0.01 vs 0.09 ± 0.01 U; tumor necrosis factor-α 0.26 ± 0.01 vs 0.12 ± 0.01 U; chemokine (C-C motif) ligand 2 0.23 ± 0.01 vs 0.1 ± 0.01 U). Maternal supplementation of pomegranate juice to lipopolysaccharide-exposed dams (pomegranate juice/lipopolysaccharide) significantly reduced maternal serum interleukin-6 levels (3059 ± 1121 pg/mL, fetal brain: caspase 3 active form (0.2 ± 0.01 U), nuclear factor-κB p65 (0.22 ± 0.01 U), phosphoneuronal nitric oxide synthase (0.19 ± 0.01 U) as well as the brain proinflammatory cytokines (interleukin-6, tumor necrosis factor-α and chemokine [C-C motif] ligand 2) compared to lipopolysaccharide group. CONCLUSION: Maternal pomegranate juice supplementation may attenuate maternal inflammation-induced fetal brain injury. Pomegranate juice neuroprotective effects might be secondary to the suppression of both the maternal inflammatory response and inhibition of fetal brain apoptosis, neuronal nitric oxide synthase, and nuclear factor-κB activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Feto/efectos de los fármacos , Jugos de Frutas y Vegetales , Lipopolisacáridos/farmacología , Lythraceae , Óxido Nítrico Sintasa de Tipo I/efectos de los fármacos , Factor de Transcripción ReIA/efectos de los fármacos , Animales , Antioxidantes , Apoptosis/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/inmunología , Citocinas/efectos de los fármacos , Citocinas/inmunología , Suplementos Dietéticos , Femenino , Feto/inmunología , Feto/metabolismo , Inflamación , Interleucina-6/inmunología , FN-kappa B/efectos de los fármacos , FN-kappa B/inmunología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo I/inmunología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Embarazo , Ratas , Factor de Transcripción ReIA/inmunología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
20.
Arch Endocrinol Metab ; 62(2): 212-220, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29641741

RESUMEN

OBJECTIVE: To observe the effect of short-term insulin intensive treatment on the monocyte chemoattractant protein-1 (MCP-1) as well as on the nuclear factor-kappa B (NF-κB) expression of peripheral blood monocyte. This is also in addition to observing the serum MCP-1 level in newlydiagnosed type 2 diabetic patients and probing its anti-inflammation effects. SUBJECTS AND METHODS: Twenty newly-diagnosed type 2 diabetic patients were treated with an insulin intensive treatment for 2 weeks. MCP-1 and NF-κB expression on the monocyte surface were measured with flow cytometry, the serum MCP-1 level was measured by enzyme linked immunosorbent assay (ELISA) during pretreatment and post-treatment. RESULTS: After 2 weeks of the treatment, MCP-1 and NF-κB protein expression of peripheral blood monocyte and serum MCP-1 levels decreased significantly compared with those of pre-treatment, which were (0.50 ± 0.18)% vs (0.89 ± 0.26)% (12.22 ± 2.80)% vs (15.53 ± 2.49)% and (44.53 ± 3.97) pg/mL vs (49.53 ± 3.47) pg/mL, respectively (P < 0.01). The MCP-1 expression on monocyte surface had a significant positive relationship with serum MCP-1 levels (r = 0.47, P < 0.01). CONCLUSIONS: Short-term insulin intensive therapy plays a role in alleviating the increased inflammation reaction in type 2 diabetics.


Asunto(s)
Quimiocina CCL2/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inflamación/prevención & control , Insulina/administración & dosificación , Monocitos/química , FN-kappa B/efectos de los fármacos , Estudios de Casos y Controles , Quimiocina CCL2/sangre , Diabetes Mellitus Tipo 2/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , FN-kappa B/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA