Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chin Med ; 17(1): 27, 2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35193642

RESUMEN

BACKGROUND: Tea trees originated in southwest China 60 million or 70 million years ago. Written records show that Chinese ancestors had begun drinking tea over 3000 years ago. Nowadays, with the aging of populations worldwide and more people suffering from non-communicable diseases or poor health, tea beverages have become an inexpensive and fine complementary and alternative medicine (CAM) therapy. At present, there are 3 billion people who like to drink tea in the world, but few of them actually understand tea, especially on its development process and the spiritual and cultural connotations. METHODS: We searched PubMed, Google Scholar, Web of Science, CNKI, and other relevant platforms with the key word "tea", and reviewed and analyzed tea-related literatures and pictures in the past 40 years about tea's history, culture, customs, experimental studies, and markets. RESULTS: China is the hometown of tea, tea trees, tea drinking, and tea culture. China has the oldest wild and planted tea trees in the world, fossil of a tea leaf from 35,400,000 years ago, and abundant tea-related literatures and art works. Moreover, tea may be the first Chinese herbal medicine (CHM) used by Chinese people in ancient times. Tea drinking has many benefits to our physical health via its antioxidant, anti-inflammatory, immuno-regulatory, anticancer, cardiovascular-protective, anti-diabetic, and anti-obesity activities. At the moment, COVID-19 is wreaking havoc across the globe and causing severe damages to people's health and lives. Tea has anti-COVID-19 functions via the enhancement of the innate immune response and inhibition of viral growth. Besides, drinking tea can allow people to acquire a peaceful, relaxed, refreshed and cheerful enjoyment, and even longevity. According to the meridian theory of traditional Chinese medicine, different kinds of tea can activate different meridian systems in the human body. At present, black tea (fermented tea) and green tea (non-fermented tea) are the most popular in the world. Black tea accounts for over 90% of all teas sold in western countries. The world's top-grade black teas include Qi Men black in China, Darjeeling and Assam black tea in India, and Uva black tea in Sri Lanka. However, all top ten famous green teas in the world are produced in China, and Xi Hu Long Jing tea is the most famous among all green teas. More than 700 different kinds of components and 27 mineral elements can be found in tea. Tea polyphenols and theaflavin/thearubigins are considered to be the major bioactive components of black tea and green tea, respectively. Overly strong or overheated tea liquid should be avoided when drinking tea. CONCLUSIONS: Today, CAM provides an array of treatment modalities for the health promotion in both developed and developing countries all over the world. Tea drinking, a simple herb-based CAM therapy, has become a popular man-made non-alcoholic beverage widely consumed worldwide, and it can improve the growth of economy as well. Tea can improve our physical and mental health and promote the harmonious development of society through its chemical and cultural elements.

2.
Front Pharmacol ; 11: 1131, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848749

RESUMEN

The pathogenesis of preeclampsia (PE) involves several pathophysiological processes that may be affected by glucocorticoid (GC). We confirmed previously that GC exposure could result in PE, while PE is linked to a deficiency of lipoxin A4 (LXA4), an endogenous dual anti-inflammatory and proresolving mediator. The present study was to investigate whether GC exposure induces PE via dampening LXA4. In the study, cortisol levels of PE women were higher than those of normal pregnancies, LXA4 levels were downregulated in both PE patients and GC-mediated PE rats, and leukotriene B4 (LTB4) levels were upregulated in both PE patients and GC- mediated PE rats. Moreover, cortisol levels were negatively correlated to LXA4 levels, while positively correlated to LTB4 levels in PE patients. Mechanically, GC downregulated LXA4 via disturbing its biosynthetic enzymes, including ALOX15, ALOX5B and ALOX5, especially activating ALOX5, the key enzyme for class switching between LXA4 and LTB4. Importantly, replenishing LXA4 could ameliorate PE-related symptoms and placental oxidative stress in PE rat model induced by GC. Moreover, LXA4 could inhibit GC-mediated ALOX5 activation and LTB4 increase, and also suppress 11ß-HSD2 expression and corticosterone upregulation. The protective actions of LXA4 might be explained by its roles in antagonizing the adverse effects of GC on trophoblast development. Together, our findings indicate that GC exposure could contribute to PE through dampening LXA4, and GC/LXA4 axis may represent a common pathway through which PE occurs.

3.
Int Immunopharmacol ; 83: 106435, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32222641

RESUMEN

Acetaminophen (APAP) is a common antipyretic and analgesic drug, but its overdose can induce acute liver failure with lack of effective therapies. Hesperetin, a dihydrogen flavonoid compound, has been revealed to exert multiple pharmacological activities. Here, we explored the protective effects and mechanism of hesperetin on APAP-induced hepatotoxicity. The results showed that pretreatment with hesperetin dose-dependently attenuated APAP-induced acute liver injury in mice, as measured by alleviated serum enzymes activities, hepatic pathological damage and apoptosis. Moreover, hesperetin mitigated APAP-induced oxidative stress and inflammatory response in mice by inhibiting oxidative molecules but increasing antioxidative molecules production, reducing inflammatory cells infiltration and proinflammatory cytokines production, blocking Toll-like receptor (TLR)-4 signal activation. In vitro experiment indicated that hesperetin dose-dependently inhibited APAP-primed cytotoxicity, apoptosis, and reactive oxygen species (ROS) in murine AML12 hepatocytes. Notably, hesperetin up-regulated expression of heme oxygenase-1 (HO-1) mRNA and protein in the liver of mice and AML12 cells exposed to APAP. Furthermore, knockdown of HO-1 by adenovirus-mediated HO-1 siRNA reverted these beneficial effects of hesperetin on APAP-induced hepatocytotoxicity as well as ROS and inflammatory response in vivo and in vitro. These findings demonstrated that hesperetin exerted a protective prophylaxis on APAP-induced acute liver injury by inhibiting hepatocyte necrosis and apoptosis, oxidative stress and inflammatory response via up-regulating HO-1 expression.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Hemo-Oxigenasa 1/metabolismo , Hepatocitos/efectos de los fármacos , Hesperidina/farmacología , Hesperidina/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Hemo-Oxigenasa 1/genética , Hepatocitos/citología , Inflamación/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis/prevención & control , Estrés Oxidativo/efectos de los fármacos , Transaminasas/sangre , Regulación hacia Arriba
4.
Cell Death Dis ; 11(1): 78, 2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-32001671

RESUMEN

Preeclampsia (PE) remains a leading cause of maternal and neonatal morbidity and mortality. Numerous studies have shown that women with PE develop autoantibody, termed angiotensin II type 1 receptor autoantibody (AT1-AA), and key features of the disease result from it. Emerging evidence has indicated that inflammatory cell necrosis, such as pyroptosis, could lead to autoantigen exposure and stimulate autoantibody production. Caspase-1, the central enzyme of inflammasome and key target of pyroptosis, may play roles in AT1R exposure and AT1-AA production. Exploring endogenous regulator that could inhibit AT1-AA production by targeting pyroptosis will be essential for treating PE. Lipoxin A4 (LXA4), endogenous dual anti-inflammatory and proresolving lipid mediator, may inhibit AT1-AA production via modulating caspase-1. Thus, we explore whether caspase-1 is essential for AT1-AA production and LXA4 inhibits AT1-AA via modulating caspase-1. PE patients and mice developed AT1-AA associated with caspase-1 activation. Caspase-1 deletion leaded to AT1-AA decrease in PE mice. Consistent with these findings, we confirmed caspase-1 activation, trophoblast pyroptosis and AT1R exposure in PE mice and trophoblast model, while caspase-1 deficiency showed decreased trophoblast pyroptosis and AT1R exposure in vitro and in vivo. Interestingly, LXA4 could suppress AT1-AA production via regulating caspase-1 as well as enhancing phagocytosis of dead trophoblasts by macrophages. These results suggest that caspase-1 promotes AT1-AA production via inducing trophoblast pyroptosis and AT1R exposure, while LXA4 suppresses AT1-AA production via modulating caspase-1, supporting caspase-1 serving as a therapeutic target for attenuating AT1-AA and LXA4 protecting patients from AT1-AA and PE.


Asunto(s)
Autoanticuerpos/metabolismo , Caspasa 1/metabolismo , Lipoxinas/farmacología , Preeclampsia/inmunología , Piroptosis/efectos de los fármacos , Receptor de Angiotensina Tipo 1/inmunología , Trofoblastos/efectos de los fármacos , Antagonistas de Receptores de Angiotensina , Animales , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Caspasa 1/sangre , Caspasa 1/deficiencia , Caspasa 1/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Lipopolisacáridos/farmacología , Lipoxinas/sangre , Lipoxinas/deficiencia , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Preeclampsia/sangre , Preeclampsia/metabolismo , Embarazo , Piroptosis/genética , Piroptosis/inmunología , ARN Interferente Pequeño , Receptor de Angiotensina Tipo 1/sangre , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Bazo/crecimiento & desarrollo , Bazo/inmunología , Bazo/patología , Trofoblastos/metabolismo
5.
Sci Adv ; 5(11): eaax0629, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31763448

RESUMEN

The mechanisms that drive programmed resolution of inflammation remain elusive. Here, we report the temporal regulation of soluble (s) and transmembrane (m) fibrinogen-like protein 2 (Fgl2) during inflammation and show that both sFgl2 and mFgl2 correlate with the outcome. The expression and ectodomain shedding of Fgl2 are respectively promoted by miR-466l and metalloproteinases (ADAM10 and ADAM17) during inflammation resolution. Deficiency of Fgl2 enhances polymorphonuclear neutrophil (PMN) infiltration but impairs macrophage (MΦ) maturation and phagocytosis and inhibits the production of n-3 docosapentaenoic acid-derived resolvin 5 (RvDp5). In contrast, administration of sFgl2 blunts PMN infiltration as well as promotes PMN apoptosis and RvDp5 biosynthesis. By activating ALX/FPR2, RvDp5 enhances sFgl2 secretion via ADAM17 and synergistically accelerates resolution of inflammation. These results uncover a previously unknown endogenous programmed mechanism by which Fgl2 regulates resolution of inflammation and shed new light on clinical sepsis treatments.


Asunto(s)
Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Fibrinógeno/metabolismo , Sepsis/metabolismo , Proteína ADAM10/genética , Proteína ADAM17/genética , Adulto , Animales , Ácidos Docosahexaenoicos/genética , Fibrinógeno/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Persona de Mediana Edad , Neutrófilos/metabolismo , Sepsis/sangre , Sepsis/genética , Adulto Joven
6.
J Cancer ; 10(19): 4719-4730, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31528237

RESUMEN

Unresolved inflammation, due to insufficient production of proresolving anti-inflammatory lipid mediators, can lead to tumorigenesis. Among these mediators, lipoxin A4 (LXA4) has potent anti-carcinogenic properties, and may serve as key target for modulating inflammation-associated cancer like colorectal cancer. The purpose of present study was to clarify the roles of LXA4 in colorectal cancer. We investigated the effects and underlying mechanisms of LXA4 in colorectal cancer and its relationship with tumor-associated inflammation and immune microenvironment by employing clinical samples and mouse colorectal cancer cell line CT26-bearing tumor model as well as colorectal cancer cells. It was found that colorectal cancer is associated with dysregulation of immune microenvironment and deficiency of LXA4 that could play different roles at different stages of tumor growth: inhibiting early but promoting late tumor growth. Analysis of peripheral immune cells in subcutaneous xenograft mice model disclosed that early LXA4 treatment induced lymphocytes and inhibited neutrophils and monocytes, while late LXA4 treatment induced neutrophils but inhibited lymphocytes. Detailed analysis of tumor microenvironment revealed that early LXA4 treatment could inhibit inflammatory mediators expressions and leukocytes infiltration into tumor. Furthermore, LXA4 could suppress the expressions of p-ERK, p-P38 and NF-κB in subcutaneous xenograft. Additionally, LXA4 could inhibit the proliferation and migration of colorectal cancer cells, and, meanwhile, inhibit the proliferation and migration of colorectal cancer cells stimulated by activated macrophage-conditioned media. These findings suggest that colorectal cancer is associated with a deficiency of LXA4 that could suppress colorectal cancer via modulating tumor-associated inflammation and immune microenvironment as well as inhibiting colorectal cancer cell development.

7.
Am J Reprod Immunol ; 81(5): e13107, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30811719

RESUMEN

PROBLEM: To test whether lipoxin A4 (LXA4) interferes with embryo implantation via suppression of epithelial-mesenchymal transition (EMT). METHOD OF STUDY: We developed a mouse model of LXA4 blocking embryo implantation and detected the indicators of EMT to confirm that LXA4 inhibits EMT might be a mechanism of interfering with the embryo implantation. We detected integrin-linked kinase (ILK), N-formylpeptide receptor 2 (FPR2), vascular endothelial growth factor, matrix metalloproteinases (MMPs), Akt, GSK3ß, NF-ĸB, twist, vimentin, fibronectin, and ß-catenin mRNA expression using reverse transcriptase-polymerase chain reaction (RT-PCR) and real-time RT-PCR; localized protein expression using immunohistochemistry and Western blotting assay; MMPs activity assay by gelatin zymography; and the status of implantation in pregnant animals assessed by pontamine blue reaction test. RESULTS: Preimplantation administration of LXA4 resulted in implantation failure. LXA4 has a time- and dose-dependent effect on embryo implantation. Day 0.5 after fertilization is the most effective time to use LXA4 to block embryo implantation. (a) LXA4 reduced endometrial stroma edema; (b) LXA4 inhibited the activity of MMP9 and significantly upregulated the expression of ß-catenin, and downregulated the expression of vimentin, fibronectin, twist, NF-κB, Akt, and Gsk-3ß in the endometrium and TEV-1 cells; (c) LXA4 upregulated the expression of FPR2, and downregulated the expression of ILK; FPR2-overexpressing had an inhibitory effect on ILK in TEV-1 cells. CONCLUSION: LXA4 inhibits EMT which attenuates ILK action by enhancing FPR2; therefore, this might be a mechanism of interfering with embryo implantation.


Asunto(s)
Infertilidad Femenina/inmunología , Lipoxinas/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Animales , Células Cultivadas , Implantación del Embrión , Transición Epitelial-Mesenquimal , Femenino , Humanos , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Embarazo , Receptores de Formil Péptido/genética , Receptores de Lipoxina/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
J Cancer ; 9(7): 1182-1187, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29675099

RESUMEN

Hepatitis B virus (HBV) infection is a leading cause for hepatocellular carcinoma (HCC). Dysregulation of DNA double-strand break (DSB) repair may explain the pathogenesis of HBV-related HCC. Tumor suppressor CtIP plays a critical role in DSB repair. The purpose of present study was to clarify whether HBV affects CtIP expression in DSB repair of hepatoma cell. HepG2.2.15 was selected as the HBV positive hepatoma cell line, while HepG2 as the HBV negative hepatoma cell line. The two cell lines were treated with bleomycin to induce DSB. Bleomycin treatment could result in DSB by γ-H2AX detection. CtIP gene expression was significantly upregulated after DSB in both HepG2 and HepG2.2.15, while CtIP expression of HepG2.2.15 was higher than that observed in HepG2 before and after DSB. CtIP protein expression was the same pattern as its gene expression. Phosphorylated CtIP (p-CtIP, serine site) was even lower than detectable limit in both HepG2 and HepG2.2.15 before DSB. However, p-CtIP of HepG2.2.15 was significantly lower than that of HepG2 after DSB. These results suggest that HBV could interfere CtIP via enhancing its expression while dampening its phosphorylation, which may disrupt DSB repair pathways and implicate CtIP dysfunction in HBV-related HCC.

9.
Placenta ; 62: 16-24, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29405962

RESUMEN

INTRODUCTION: Preeclampsia is associated with the presence of pathogenic angiotensin-receptor-activating autoantibodies. Cadmium is an increasingly prevalent environmental pollutant that can mimic oestrogens, which may enhance immunoglobulin production. Progesterone exerts opposite effects to oestrogen. METHODS: We measured the levels of cadmium and progesterone in preeclamptic patients and controls. Pregnant rats exposed to cadmium (0.125 mg/kg body weight) from gestational day 9-12 were treated with/without progesterone (3 mg/kg) beginning from gestational day 9 to delivery. We analysed the main features of preeclampsia and circulating level of the angiotensin II type 1 receptor agonistic autoantibody. We also measured the expression of activation-induced cytosine deaminase in B cells. RESULTS: There were higher cadmium levels and lower progesterone levels in the blood of preeclamptic women than in the blood of those with a healthy pregnancy. Based on this finding, a rat model of preeclampsia was established by intraperitoneally administrating low-dose cadmium on gestational days 9-12. Rats were then treated with/without progesterone. Key features of preeclampsia, including hypertension, proteinuria and placental abnormalities, appeared in pregnant rats after cadmium injection and improved after treatment with progesterone. Cadmium increased immunoglobulin production, mainly angiotensin II type 1-receptor-agonistic autoantibodies, by increasing the expression of activation-induced cytosine deaminase in B cells; progesterone exerted an opposite effect. CONCLUSION: Cadmium induced immune abnormalities that may be a key pathogenic contributor to preeclampsia. Progesterone supplementation to correct hormonal imbalance may be a viable strategy for preeclampsia management.


Asunto(s)
Autoanticuerpos/sangre , Cadmio/farmacología , Hipertensión/tratamiento farmacológico , Preeclampsia/sangre , Progesterona/uso terapéutico , Receptor de Angiotensina Tipo 1/inmunología , Adulto , Animales , Cadmio/sangre , Femenino , Humanos , Hipertensión/sangre , Inmunoglobulinas/sangre , Embarazo , Progesterona/sangre , Progesterona/farmacología , Ratas
10.
Cell Physiol Biochem ; 45(4): 1377-1389, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29462800

RESUMEN

BACKGROUND/AIMS: This study aimed to explore the metabololipidome in mice upon cupping treatment. METHODS: A nude mouse model mimicking the cupping treatment in humans was established by administrating four cupping sets on the back skin for 15 minutes. UPLC-MS/ MS was performed to determine the PUFA metabolome in mice skin and blood before and after cupping treatment. The significantly changed lipids were administered in macrophages to assess the production of pro-inflammatory cytokines IL-6 and TNF-α by ELISA. RESULTS: The anti-inflammatory lipids, e.g. PGE1, 5,6-EET, 14,15-EET, 10S,17S-DiHDoHE, 17R-RvD1, RvD5 and 14S-HDoHE were significantly increased while pro-inflammatory lipids, e.g. 12-HETE and TXB2 were deceased in the skin or plasma post cupping treatment. Cupping treatment reversed the LPS-stimulated IL-6 and TNF-α expression in mouse peritoneal exudates. Moreover, 5,6-EET, PGE1 decreased the level of TNF-α, while 5,6-EET, 5,6-DHET downregulated IL-6 production in macrophages. Importantly, 14,15-EET and 14S-HDoHE inhibited both IL-6 and TNF-α induced by lipopolysaccharide (LPS). 17-RvD1, RvD5 and PGE1 significantly reduced the LPS-initiated TNF-α, while TXB2 and 12-HETE further upregulated the LPS-enhanced IL-6 and TNF-α expression in macrophages. CONCLUSION: Our results reveal the identities of anti-inflammatory versus pro-inflammatory metabolipidome and suggest the potential therapeutic mechanism of cupping treatment.


Asunto(s)
Ácidos Grasos Insaturados/análisis , Hematoma/patología , Lípidos/análisis , Metaboloma , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/análisis , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/análisis , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Ácidos Grasos Insaturados/metabolismo , Hematoma/metabolismo , Interleucina-6/análisis , Lípidos/sangre , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaboloma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Células RAW 264.7 , Piel/metabolismo , Tromboxano B2/análisis , Factor de Necrosis Tumoral alfa/análisis , Regulación hacia Arriba/efectos de los fármacos
11.
Hypertens Res ; 41(2): 104-111, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29093562

RESUMEN

The pathogenesis of preeclampsia (PE) involves a number of biological processes that may be directly or indirectly affected by glucocorticoid (GC) and vitamin D. GC exposure increases the risk of PE, and 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) deficiency may result in PE. The purpose of the present study was to confirm the involvement of GC/1,25-(OH)2D3 axis in the pathogenesis of PE. In the study, cortisol levels of PE patients were found to be higher than that of non-complicated pregnancies, while 1,25-(OH)2D3 were decreased in both PE women and GC-induced PE rats. Mechanically, GC reduced 1,25-(OH)2D3 levels via disturbing its biosynthetic and catabolic enzymes, including Cyp3a1,Cyp24a1 and Cyp27b1, especially enhancing the expressions of Cyp3a1, the dominant enzyme for vitamin D degeneration. Moreover, replenishing 1,25-(OH)2D3 ameliorated the symptoms and placental oxidative stress of GC-induced rat PE. The protective actions of 1,25-(OH)2D3 might be explained by its roles in antagonizing the effects of GC on trophoblast proliferation and apoptosis. Together, these findings suggest that GC exposure could lead to PE via dampening 1,25-(OH)2D3 biosynthesis, and GC/1,25-(OH)2D3 axis might represent a common pathway through which PE occurs.


Asunto(s)
Calcitriol/antagonistas & inhibidores , Glucocorticoides/toxicidad , Preeclampsia/inducido químicamente , Preeclampsia/metabolismo , Albuminuria/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Calcitriol/sangre , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP3A/biosíntesis , Citocromo P-450 CYP3A/genética , Femenino , Glucocorticoides/sangre , Humanos , Hidrocortisona/metabolismo , Estrés Oxidativo/efectos de los fármacos , Placenta/efectos de los fármacos , Placenta/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Trofoblastos/efectos de los fármacos
12.
Cell Death Differ ; 24(2): 263-275, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27768123

RESUMEN

Abnormal features of the systemic lupus erythematosus (SLE)-derived neutrophils, promoted aberrant immune response, have inspired new studies of the induction of autoimmunity and the development of organ damage in SLE. In this study, we explore the effect of milk fat globule-EGF factor 8 (MFG-E8) on the aberrant nitrification features in pristane-induced lupus. SLE patients and mice with pristane-induced lupus develop autoantibodies associated with MFG-E8 overproduction. However, the deletion of MFG-E8 leads to uncontrolled early pulmonary and peritoneal inflammation and tissue damage in mice with pristane-induced lupus. Consistent with these findings, MFG-E8-deficient mice that are exposed to pristane show enhanced neutrophil accumulation and increased neutrophil death, including apoptosis, necrosis and NETosis, as well as impaired phagocytosis of macrophages. The consequences are the expansion of diffuse pulmonary hemorrhage, increased anti-nuclear antibody, anti-dsDNA antibody and anti-neutrophil cytoplasmic antibody levels, and enhanced immune complexes deposition and neutrophil extracellular traps (NETs) formation in the lung and kidney tissues of MFG-E8-deficient mice exposed to pristane. In patients with SLE and mice with pristane-induced lupus, neutrophil accumulation is elevated, which depends on higher expression of the surface receptor CXCR2. After pretreatment with recombinant MFG-E8, the surface expression of CXCR2 on neutrophil is downregulated, and the MFG-E8 deletion increase CXCR2 expression by ~40%. These studies indicate that MFG-E8 reduces neutrophil migration and NETosis via downregulating surface CXCR2 expression in parallel with its role in the phagocytosis of apoptotic neutrophils, suggesting that MFG-E8 may serve as a therapeutic agent for attenuating the early inflammatory responses of SLE and protect patients from lupus-related damage.


Asunto(s)
Antígenos de Superficie/metabolismo , Lupus Eritematoso Sistémico/patología , Proteínas de la Leche/metabolismo , Neutrófilos/inmunología , Adulto , Animales , Antígenos de Superficie/genética , Apoptosis/efectos de los fármacos , Autoanticuerpos/sangre , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/análisis , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Femenino , Fulerenos , Glomerulonefritis/patología , Humanos , Riñón/metabolismo , Pulmón/metabolismo , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteínas de la Leche/genética , Neutrófilos/citología , Neutrófilos/metabolismo , Fagocitosis/efectos de los fármacos , Receptores de Interleucina-8B/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología , Adulto Joven
13.
Int Immunopharmacol ; 39: 229-235, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27494686

RESUMEN

Sepsis is a serious disease that leads to severe inflammation, dysregulation of immune system, multi-organ failure and death. Innate response activator (IRA) B cells, which produce granulocyte-macrophage colony-stimulating factor (GM-CSF), protect against microbial sepsis. Lipid mediator lipoxin A4 (LXA4) exerts anti-inflammatory and immunoregulatory effects, and it has been reported that LXA4 receptor ALX/FPR2 is expressed on B cells. Here, we investigated the potential role of LXA4 on IRA B cells in lipopolysaccharide (LPS)-induced sepsis. We found that LXA4 significantly promoted the expansion of splenic IRA B cells and increased GM-CSF expression in splenic B cells with LPS stimulation. After splenectomy, LXA4 treatment did not change the serum or peritoneal IL-1ß, IL-6 and TNF-α levels in LPS-induced sepsis. LXA4 accelerated the migration of peritoneal B cells to spleen for their differentiation into IRA B cells, whereas this effect was independent of peritoneal macrophage. Furthermore, LXA4 enhanced the phosphorylation level of signal transducer and activator of transcription 5 (STAT5) in splenic B cells. These results suggest that LXA4 protects against LPS-induced sepsis by promoting the generation and migration of splenic IRA B cells, and the underlying molecular mechanism may be related to STAT5 activation. It might provide new insights and therapeutic approaches for treating sepsis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Linfocitos B/efectos de los fármacos , Inmunidad Innata , Lipoxinas/uso terapéutico , Factor de Transcripción STAT5/metabolismo , Sepsis/tratamiento farmacológico , Animales , Linfocitos B/inmunología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , Sepsis/inducido químicamente
14.
Environ Pollut ; 218: 770-782, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27511439

RESUMEN

With increased industrial development, cadmium is an increasingly important environmental pollutant. Studies have identified various adverse effects of cadmium on human beings. However, the relationships between cadmium pollution and the pathogenesis of preeclampsia remain elusive. The objective of this study is to explore the effects of cadmium on immune system among preeclamptic patients and rats. The results showed that the cadmium levels in the peripheral blood of preeclamptic patients were significantly higher than those observed in normal pregnancy. Based on it, a novel rat model of preeclampsia was established by the intraperitoneal administration of cadmium chloride (CdCl2) (0.125 mg of Cd/kg body weight) on gestational days 9-14. Key features of preeclampsia, including hypertension, proteinuria, placental abnormalities and small foetal size, appeared in pregnant rats after the administration of low-dose of CdCl2. Cadmium increased immunoglobulin production, mainly angiotensin II type 1-receptor-agonistic autoantibodies (AT1-AA), by increasing the expression of activation-induced cytosine deaminase (AID) in B cells. AID is critical for the maturation of antibody and autoantibody responses. In addition, angiotensin II type 1-receptor-agonistic autoantibody, which emerged recently as a potential pathogenic contributor to PE, was responsible for the deposition of complement component 5 (C5) in kidneys of pregnant rats via angiotensin II type 1 receptor (AT1R) activation. C5a is a fragment of C5 that is released during C5 activation. Selectively interfering with C5a signalling by a complement C5a receptor-specific antagonist significantly attenuated hypertension and proteinuria in Cd-injected pregnant rats. Our results suggest that cadmium induces immune abnormalities that may be a key pathogenic contributor to preeclampsia and provide new insights into treatment strategies of preeclampsia.


Asunto(s)
Autoanticuerpos/sangre , Cadmio/efectos adversos , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/efectos adversos , Preeclampsia/inducido químicamente , Receptor de Angiotensina Tipo 1/sangre , Animales , Linfocitos B/metabolismo , Cadmio/sangre , Complemento C5/metabolismo , Citosina Desaminasa/metabolismo , Femenino , Humanos , Hipertensión/inducido químicamente , Riñón/efectos de los fármacos , Riñón/metabolismo , Placenta/efectos de los fármacos , Placenta/metabolismo , Preeclampsia/sangre , Preeclampsia/inmunología , Embarazo , Proteinuria/inducido químicamente , Ratas , Ratas Wistar
15.
Gen Comp Endocrinol ; 225: 61-70, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26407501

RESUMEN

In pregnancy, placenta can be exposed to glucocorticoids (GCs) via several ways, which may disturb placentation and adversely affect pregnancy. Preeclampsia (PE) is thought to be attributed, in part, to impaired trophoblast development. The purpose of the present study was to confirm that GC exposure in early placentation could lead to PE in rats, with the mechanisms involving dysregulated trophoblast development. In the study, pregnant rats were administered with 2.5mg/kg Dex subcutaneously once per day from gestational day 7 to 13. Maternal systolic blood pressure and urinary albumin were increased, while both fetus and placenta were restricted after GC exposure relative to the control group. GC exposure also contributed to placental abnormalities and renal impairment. Moreover, placental oxidative damage was increased along with placental hypoxia-inducible factor 1-alpha (HIF1A) overexpression after GC treatment. Mechanically, GC induced PE in rat partially through inhibiting trophoblast proliferation, migration, invasion and epithelial-mesenchymal transition (EMT), which involved phospho-extracellular signal regulated kinase (p-ERK) downregulation. Furthermore, GC receptor was required for the inhibition of GC on trophoblast proliferation, migration, invasion and EMT in vitro. These findings suggest that GC exposure in early placentation could contribute to PE in pregnant rats, with the mechanisms involving inhibition of trophoblast proliferation, migration, invasion and EMT by GC.


Asunto(s)
Dexametasona/farmacología , Glucocorticoides/farmacología , Placenta/efectos de los fármacos , Placentación/efectos de los fármacos , Preeclampsia/inducido químicamente , Trofoblastos/efectos de los fármacos , Animales , Regulación hacia Abajo/efectos de los fármacos , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Placenta/metabolismo , Placenta/patología , Preeclampsia/metabolismo , Preeclampsia/patología , Embarazo , Ratas , Trofoblastos/metabolismo , Trofoblastos/patología
16.
Biol Trace Elem Res ; 170(1): 119-27, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26194818

RESUMEN

To explore the possible mechanisms of cadmium (Cd)-induced preeclamptic conditions in rats. In the present study, we introduced the in vivo model of preeclampsia by giving intraperitoneal injections of cadmium chloride (CdCl2) to pregnant rats from gestational day (GD) 4 to 19. Maternal body weights were recorded on GD 0, 14, and 20, while their systolic blood pressures (SBPs) monitored on GD 3, 11, and 18. On GD 20, rats were sacrificed and the specimens were collected. The morphological changes of placenta and kidney tissues of pregnant rats were examined by hematoxylin and eosin staining assay. Blood Cd level was detected by inductively coupled plasma mass spectrometry. Total antioxidant capacity (TAC) was evaluated using FRAP method and total nitrite (NOx) was detected with Griess reagent. Antioxidative factors and DNA damage/repair biomarkers were measured by real-time qPCR, western blot or immunohistochemistry study. The current results showed that CdCl2-treated pregnant rats developed preeclampsia (PE)-like manifestations, such as hypertension, albuminuria, with decreased TAC and increased blood Cd level, and pro-oxidative/antioxidative or DNA damage/repair biomarkers. Our study demonstrated that increased oxidative DNA damage in placenta could contribute to Cd-induced preeclamptic conditions in rat.


Asunto(s)
Cloruro de Cadmio/toxicidad , Daño del ADN , Estrés Oxidativo , Placenta/metabolismo , Preeclampsia/inducido químicamente , Animales , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley
17.
Cell Rep ; 12(3): 388-95, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26166565

RESUMEN

Hypoxia and intracellular Ca(2+) transients are fundamental traits of cancer, whereas the route and regulation of Ca(2+) mobilization in hypoxic tumorigenesis are unknown. Here, we show that stromal-interaction molecule 1 (STIM1), an ER Ca(2+) sensor, correlates with elevated hypoxia-inducible factor-1 alpha (HIF-1α) in hypoxic hepatocarcinoma cells (HCCs) and is upregulated during hepatocarcinoma growth. HIF-1 directly controls STIM1 transcription and contributes to store-operated Ca(2+) entry (SOCE). STIM1-mediated SOCE is also required for HIF-1 accumulation in hypoxic HCCs via activation of Ca(2+)/calmodulin-dependent protein kinase II and p300. Administration of YC-1, a HIF-1 inhibitor, or knockdown of HIF1A significantly diminishes hypoxia-enhanced STIM1 and suppresses tumorigenesis. Moreover, ectopic expression of STIM1 or HIF-1α partially reverses impaired growth of tumors treated with YC-1. These results suggest a mutual dependency and regulation of STIM1 and HIF-1 in controlling Ca(2+) mobilization and hypoxic tumor growth and highlight a potential target for early hypoxia-related intervention.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Calcio/metabolismo , Carcinogénesis , Hipoxia de la Célula/fisiología , Células Hep G2 , Xenoinjertos , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Molécula de Interacción Estromal 1
18.
Cancer Lett ; 364(2): 118-24, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-25979229

RESUMEN

Lipoxin A4 (LXA4), an arachidonic acid-derived anti-inflammatory lipid mediator, shows anti-tumor potential by regulating tumor immune microenvironments. However, the underlying molecular and cellular basis of this function remains unclear. IL-10-producing B (Breg) cells display tumor-promoting effects by negatively regulating anti-tumor immunity. Here we show that LXA4 inhibits tumor growth by suppressing the generation of Breg cells in tumor-bearing mice. The administration of LXA4 inhibited the induction of Breg cells. Breg cell deficiency, in turn, resulted in LXA4 losing its anti-tumor properties. Intriguingly, regulatory T (Treg) cells also had a role in this process. Targeting Breg cells by LXA4 decreased the number of Treg cells in draining lymph nodes and tumor tissues as well as enhanced cytotoxic T cell activities. In addition, we further demonstrated that LXA4 inhibited Breg cells through its dephosphorylating STAT3 and ERK. These findings unveil a new anti-tumor mechanism underlying LXA4 targeting Breg cells with potential clinical applications.


Asunto(s)
Linfocitos B Reguladores/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Interleucina-10/antagonistas & inhibidores , Lipoxinas/farmacología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Animales , Linfocitos B Reguladores/inmunología , Neoplasias Colorrectales/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Neoplasias Hepáticas Experimentales/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Factor de Transcripción STAT3/metabolismo
19.
J Mol Neurosci ; 56(4): 848-857, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25702137

RESUMEN

Lipoxin A4 (LXA4), a potent antioxidant and anti-inflammation mediator, protects brains against cerebral ischemia/reperfusion (I/R) injury in vivo. However, few reports concern its function on astrocytes during cerebral I/R injury. The pathogenesis of cerebral I/R injury involves oxidative stress caused by reactive oxygen species (ROS). Upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) is generally considered to reduce oxidative stress. Nrf2 can induce heme oxygenase-1 (HO-1) expression and glutathione (GSH) release to combat increased oxidative stress. We investigated the effects of LXA4 on astrocytic cell damage, the production of ROS, and Nrf2 pathway, especially on HO-1 expression and GSH release in cultured cortical astrocytes exposed to oxygen-glucose deprivation (OGD)/recovery (OGDR) insults. Primary astrocytes were subjected to a 4-h OGD, followed by 8-h recovery. Cell viability, the production of ROS, and GSH release were measured. Furthermore, Nrf2, HO-1, and p62 expression levels were determined by Western blot. Moreover, Nrf2 location was studied by immunofluorescence staining. Treatment of LXA4 attenuates OGDR-induced cell damage and the production of ROS in a concentration-related manner. LXA4 induced Nrf2 expression and its nuclear translocation, as well as HO-1 expression and GSH release. Moreover, LXA4 induced the excess p62 accumulation. These results indicate that LXA4 can effectively protect against OGDR-induced cell damage in astrocytes, and activation of Nrf2 pathway to reduce oxidative stress may be involved in its protective effects. p62 accumulation induced by LXA4 may be closely related to Nrf2 activation.


Asunto(s)
Antioxidantes/farmacología , Astrocitos/efectos de los fármacos , Isquemia Encefálica/metabolismo , Lipoxinas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/farmacología , Animales , Astrocitos/metabolismo , Hipoxia de la Célula , Células Cultivadas , Glucosa/deficiencia , Glutatión/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Factor 2 Relacionado con NF-E2/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
20.
Med Hypotheses ; 84(3): 223-6, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25613564

RESUMEN

The gut microbiota is intimately involved in numerous aspects of normal human physiology, including nutrition and metabolism, immunomodulation and behavior and stress response. Intestinal dysbiosis can be a contributing cause of many diseases, altering the function of both near and far organ systems. During pregnancy, the maternal body undergoes dramatic physiological changes to support the growth of fetus-placenta, while intestinal dysbiosis may directly or indirectly disturb the remodeling of physiological balance, leading to maternal maladaptation. Thus, intestinal dysbiosis, i.e., altered composition or metabolism of microbiota may adversely affect pregnancy outcome and lead to pregnancy complications via disrupting maternal adaptation. Indeed, pregnant women with potential maladaptations are at high risk of developing pregnancy disorders, which is increasingly observed in clinical cases. Here we discuss the hypothesis that intestinal dysbiosis may induce pregnancy complications via affecting maternal adaptation and the possible mechanistic pathways.


Asunto(s)
Adaptación Fisiológica/fisiología , Disbiosis/complicaciones , Intestinos/microbiología , Modelos Biológicos , Complicaciones del Embarazo/etiología , Femenino , Humanos , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...