Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Chinese journal of integrative medicine ; (12): 448-458, 2023.
Article in English | WPRIM | ID: wpr-982293

ABSTRACT

OBJECTIVE@#To investigate the molecular mechanisms underlying the beneficial effect of electroacupuncture (EA) in experimental models of Alzheimer's disease (AD) in vivo.@*METHODS@#Senescence-accelerated mouse prone 8 (SAMP8) mice were used as AD models and received EA at Yingxiang (LI 20, bilateral) and Yintang (GV 29) points for 20 days. For certain experiments, SAMP8 mice were injected intravenously with human fibrin (2 mg). The Morris water maze test was used to assess cognitive and memory abilities. The changes of tight junctions of blood-brain barrier (BBB) in mice were observed by transmission electron microscope. The expressions of fibrin, amyloid- β (Aβ), and ionized calcium-binding adapter molecule 1 (IBa-1) in mouse hippocampus (CA1/CA3) were detected by reverse transcription-quantitative polymerase chain reaction (qRT-PCR), Western blot or immunohistochemical staining. The expression of fibrin in mouse plasma was detected by enzyme-linked immunosorbent assay. The expressions of tight junction proteins zonula occludens-1 and claudin-5 in hippocampus were detected by qRT-PCR and immunofluorescence staining. Apoptosis of hippocampal neurons was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining.@*RESULTS@#Fibrin was time-dependently deposited in the hippocampus of SAMP8 mice and this was inhibited by EA treatment (P<0.05 or P<0.01). Furthermore, EA treatment suppressed the accumulation of Aβ in the hippocampus of SAMP8 mice (P<0.01), which was reversed by fibrin injection (P<0.05 or P<0.01). EA improved SAMP8 mice cognitive impairment and BBB permeability (P<0.05 or P<0.01). Moreover, EA decreased reactive oxygen species levels and neuroinflammation in the hippocampus of SAMP8 mice, which was reversed by fibrin injection (P<0.05 or P<0.01). Mechanistically, EA inhibited the promoting effect of fibrin on the high mobility group box protein 1 (HMGB1)/toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE)/nicotinamide adenine dinucleotide phosphate (NADPH) signaling pathways (P<0.01).@*CONCLUSION@#EA may potentially improve cognitive impairment in AD via inhibition of fibrin/A β deposition and deactivation of the HMGB1/TLR4 and RAGE/NADPH signaling pathways.


Subject(s)
Mice , Humans , Animals , NADP/metabolism , Toll-Like Receptor 4 , HMGB1 Protein/metabolism , Receptor for Advanced Glycation End Products/metabolism , Blood-Brain Barrier/metabolism , Neuroinflammatory Diseases , Electroacupuncture , Alzheimer Disease/therapy , Hippocampus/metabolism , Amyloid beta-Peptides/metabolism
2.
Int. j. med. surg. sci. (Print) ; 9(2): 1-10, June 2022. ilus
Article in Spanish | LILACS | ID: biblio-1512565

ABSTRACT

The receptor for advanced glycation end products (RAGE) is implicated in the pathogenesis of several chronic diseases including diabetes. The interaction between RAGE and advanced glycation end products (AGEs) promotes gene expression, enhances the release of proinflammatory molecules and causes the generation of oxidative stress in numerous cell types. The aim of this investigation was to evaluate the effect of enalapril and losartan on RAGE expression in abdominal aortic endothelium of rats with experimentally induced diabetes. Male Sprague-Dawley rats, weighing approximately 150 - 200 g, were used. Diabetes was induced in 30 rats by intravenous administration of a single dose of 55 mg/kg body weight of streptozotocin (ETZ). The following groups were studied: control (n=10), diabetic (n=10), losartan-treated diabetic (n=10) and enalapril-treated diabetic (n=10) rats. RAGE expression in aortic endothelium was determined by indirect immunofluorescence. A significant increase in RAGE expression was observed in diabetic animals versus controls (p<0.001), there was a decrease in RAGE expression, in animals treated with losartan versus controls (p<0.01) and in those treated with enalapril (p<0.05) versus control and versus diabetes + vehicle. In conclusion, in the experimental model of ETZ-induced diabetes, there is an increase in RAGE expression at the level of the abdominal aortic endothelium, which can be reversed by treatment with losartan and/or enalapril, two drugs that block the renin-angiotensin system, suggesting its involvement in the molecular events related to vascular damage during diabetes.


El receptor para productos finales de glicación avanzada (RAGE) está implicado en la patogénesis de varias enfermedades crónicas incluyendo la diabetes. La interacción entre RAGE y los productos finales de glicación avanzada (AGEs), promueve la expresión génica, potencia la liberación de moléculas proinflamatorias y provoca la generación de estrés oxidativo en numerosos tipos de células. El objetivo de esta investigación fue evaluar el efecto del enalapril y el losartán sobre la expresión de RAGE en el endotelio de la aorta abdominal de ratas con diabetes inducida experimentalmente. Se utilizaron ratas Sprague-Dawley machos, con un peso aproximado de entre 150 - 200 g. La diabetes se indujo en 30 ratas mediante la administración intravenosa de una sola dosis de 55 mg/Kg de peso corporal de estreptozotocina (ETZ). Se estudiaron los siguientes grupos: ratas control (n=10), diabéticas (n=10), diabéticas tratadas con losartán (n=10) y diabéticas tratadas con enalapril (n=10). La expresión de RAGE en el endotelio aórtico se determinó por inmunofluorescencia indirecta. Se observó un incremento significativo en la expresión de RAGE en los animales diabéticos versus los controles (p<0.001), hubo una disminución en la expresión de RAGE, en los animales tratados con losartán versus los controles (p<0.01) y en los tratados con enalapril (p<0.05) versus control y versus diabetes + vehículo. En conclusión, en el modelo experimental de diabetes inducida por ETZ, existe un incremento en la expresión de RAGE a nivel del endotelio de la aorta abdominal, la cual puede revertirse mediante el tratamiento con losartán y/o enalapril, dos fármacos bloqueadores del sistema renina-angiotensina, lo cual sugiere la participación del mismo en los acontecimientos moleculares relacionados con el daño vascular durante la diabetes.


Subject(s)
Animals , Male , Rats , Enalapril/pharmacology , Losartan/pharmacology , Diabetes Mellitus, Experimental , Receptor for Advanced Glycation End Products/drug effects , Aorta, Abdominal , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Immunohistochemistry , Rats, Sprague-Dawley , Angiotensin II Type 1 Receptor Blockers/pharmacology , Endothelium , Receptor for Advanced Glycation End Products/metabolism
3.
Journal of Experimental Hematology ; (6): 493-500, 2022.
Article in Chinese | WPRIM | ID: wpr-928742

ABSTRACT

OBJECTIVE@#To evaluate the value of high mobility group protein B1 (HMGB1) and soluble receptor for advanced glycation end products (sRAGE) in the diagnosis, efficacy monitoring and prognosis of newly diagnosed multiple myeloma (MM) patients.@*METHODS@#Fifty newly diagnosed MM patients before and after chemotherapy and 50 hematological outpatients from October 2018 to May 2020 were selected. Enzyme linked immunosorbent assay (ELISA) was used to detect the serum HMGB1 and sRAGE levels of the patients. ROC was used to further analyze the efficacy of serum HMGB1 and sRAGE levels on the diagnosis of MM. At the same time, the serum levels of HMGB1 and sRAGE before and after chemotherapy were compared, and their values in the evaluation of curative effect of MM patients were analyzed. According to the mean values of serum HMGB1 and sRAGE, all the patients were divided into different groups, the clinical characteristics and survival status of the patients were compared.@*RESULTS@#Before treatment the serum HMGB1 level of the patients in MM group was higher than that in control group, while sRAGE level was lower (t=11.363,6.127, P<0.001). The AUC of serum HMGB1 and sRAGE in the MM patients was 0.955 and 0.811, respectively. After 3 courses of chemotherapy, HMGB1 level of the patients in CR group was lower than before chemotherapy, while in PD group was higher, as well as sRAGE level of the patients in PR group (P<0.05). There were significant differences in R-ISS stage, HGB, CRP, ESR, CD56, CD117, D13S319 deletion between HMGB1 high expression group and HMGB1 low expression group (χ2=3.920, 6.522, 6.65, 4.16, 3.945, 6.65, 4.16, P<0.05), while there were significant differences in ISS stage, CRP and CD56 between sRAGE low expression group (28 cases) and sRAGE high expression group (22 cases) (χ2=4.565, 4.711, 5.547, P<0.05). Kaplan-Meier survival analysis showed that the patients in HMGB1 low expression group had better survival condition, for PFS Tlow>Thigh (χ2=9.470, P<0.05), and for OS Tlow>Thigh (χ2=7.808, P<0.05); there was no difference in the survival of sRAGE high expression group and low expression group, for PFS Tlow<Thigh (χ2=1.661, P>0.05), and for OS Tlow<Thigh (χ2=2.048, P>0.05). Cox analysis showed that LDH and HMGB1 were the factors affecting the prognosis of the patients, and both of them affected PFS (HR=2.771, 95% CI: 1.002-7.662, P=0.049; HR=6.022, 95% CI: 1.689-21.470, P=0.006), while HMGB1 also affected OS (HR=4.275, 95% CI: 1.183-15.451, P=0.027).@*CONCLUSION@#The serum HMGB1 and sRAGE have certain auxiliary value for the diagnosis and curative effect monitoring of newly diagnosed MM patients, and serum HMGB1 is expected to be an auxiliary detection index for the prognosis of MM.


Subject(s)
Humans , Enzyme-Linked Immunosorbent Assay , HMGB1 Protein/blood , Multiple Myeloma/therapy , Prognosis , Receptor for Advanced Glycation End Products/blood
4.
Rev. Assoc. Med. Bras. (1992) ; 67(9): 1251-1255, Sept. 2021. tab, graf
Article in English | LILACS | ID: biblio-1351480

ABSTRACT

SUMMARY OBJECTIVE: To investigate the associations of high-mobility group box 1 and its specific receptor, receptor for advanced glycation end products with acute lung injury in patients with acute aortic dissection. METHODS: A total of 96 acute aortic dissection patients were divided into acute aortic dissection with acute lung injury group (38 cases) and acute aortic dissection without acute lung injury group (58 cases), according to partial pressure of oxygen/fraction of inspired oxygen. In addition, 44 healthy individuals were selected for the control group. The blood samples were taken. The serum high-mobility group box 1 and receptor for advanced glycation end products levels were detected by enzyme-linked immunosorbent assay, and the partial pressure of oxygen/fraction of inspired oxygen was measured. RESULTS: 24 h after admission, the high-mobility group box 1 and receptor for advanced glycation end products levels in acute aortic dissection with acute lung injury and acute aortic dissection without acute lung injury groups were significantly higher than those in the control group, respectively (p<0.05), and each index in acute aortic dissection with acute lung injury group was significantly higher than that in acute aortic dissection without acute lung injury group (p<0.05). At each time point within 96 h after admission, compared with acute aortic dissection without acute lung injury group, in acute aortic dissection with acute lung injury group, the high-mobility group box 1 and receptor for advanced glycation end products levels were increased, respectively, and the partial pressure of oxygen/fraction of inspired oxygen was decreased. The correlation analysis showed that, in acute aortic dissection patients, the high-mobility group box 1 and receptor for advanced glycation end products levels were negatively correlated with partial pressure of oxygen/fraction of inspired oxygen, respectively (p<0.05). CONCLUSIONS: The serum high-mobility group box 1 and receptor for advanced glycation end products levels may be associated with the occurrence of acute lung injury in acute aortic dissection patients. Monitoring the high-mobility group box 1 and receptor for advanced glycation end products levels can evaluate the risk of acute aortic dissection with acute lung injury.


Subject(s)
Humans , HMGB1 Protein/metabolism , Acute Lung Injury/etiology , Receptor for Advanced Glycation End Products/metabolism , Aortic Dissection , Glycation End Products, Advanced
5.
Clinics ; 76: e2348, 2021. graf
Article in English | LILACS | ID: biblio-1153978

ABSTRACT

OBJECTIVES: TTP488, an antagonist of the receptor for advanced glycation end-products, was evaluated as a potential treatment for patients with mild-to-moderate Alzheimer's disease (AD). However, the mechanism underlying the protective action of TTP488 against AD has not yet been fully explored. METHODS: Healthy male rats were exposed to aberrant amyloid β (Aβ) 1-42. Lipopolysaccharide (LPS) and the NOD-like receptor family pyrin domain containing 1 (NLRP1) overexpression lentivirus were injected to activate the NLRP1 inflammasome and exacerbate AD. TTP488 was administered to reverse AD injury. Finally, tofacitinib and fludarabine were used to inhibit the activity of Janus tyrosine kinase (JAK) and signal transducer and activator of transcription (STAT) to prove the relationship between the JAK/STAT signaling pathway and TTP488. RESULTS: LPS and NLRP1 overexpression significantly increased the NLRP1 levels, reduced neurological function, and aggravated neuronal damage, as demonstrated by the impact latency time of, time spent by, and length of the platform covered by, the mice in the Morris water maze assay, Nissl staining, and immunofluorescence staining in rats with AD. CONCLUSIONS: TTP488 administration successfully reduced AD injury and reversed the aforementioned processes. Additionally, tofacitinib and fludarabine administration could further reverse AD injury after the TTP488 intervention. These results suggest a new potential mechanism underlying the TTP488-mediated alleviation of AD injury.


Subject(s)
Animals , Male , Mice , Rats , Janus Kinases/metabolism , Alzheimer Disease/drug therapy , Tyrosine , Transducers , Signal Transduction , Amyloid beta-Peptides , Janus Kinase 2 , Receptor for Advanced Glycation End Products , Imidazoles
6.
China Journal of Chinese Materia Medica ; (24): 5693-5700, 2021.
Article in Chinese | WPRIM | ID: wpr-921754

ABSTRACT

To investigate the potential molecular markers and drug-compound-target mechanism of Mahuang Shengma Decoction(MHSM) in the intervention of acute lung injury(ALI) by network pharmacology and experimental verification. Databases such as TCMSP, TCMIO, and STITCH were used to predict the possible targets of MHSM components and OMIM and Gene Cards were employed to obtain ALI targets. The common differentially expressed genes(DEGs) were therefore obtained. The network diagram of DEGs of MHSM intervention in ALI was constructed by Cytoscape 3. 8. 0, followed by Gene Ontology(GO) and Kyoto Encyclopedia of Genes and Genomes(KEGG) enrichment analyses of target genes. The ALI model was induced by abdominal injection of lipopolysaccharide(LPS) in mice. Bronchoalveolar lavage fluid(BALF) was collected for the detection of inflammatory factors. Pathological sectioning and RT-PCR experiments were performed to verify the therapeutic efficacy of MHSM on ALI. A total of 494 common targets of MHSM and ALI were obtained. Among the top 20 key active compounds of MHSM, 14 from Ephedrae Herba were found to be reacted with pivotal genes of ALI [such as tumor necrosis factor(TNF), tumor protein 53(TP53), interleukin 6(IL6), Toll-like receptor 4(TLR4), and nuclear factor-κB(NF-κB)/p65(RELA)], causing an uncontrolled inflammatory response with activated cascade amplification. Pathway analysis revealed that the mechanism of MHSM in the treatment of ALI mainly involved AGE-RAGE, cancer pathways, PI3 K-AKT signaling pathway, and NF-κB signaling pathway. The findings demonstrated that MHSM could dwindle the content of s RAGE, IL-6, and TNF-α in the BALF of ALI mice, relieve the infiltration of inflammatory cells in the lungs, inhibit alveolar wall thickening, reduce the acute inflammation-induced pulmonary congestion and hemorrhage, and counteract transcriptional activities of Ager-RAGE and NF-κB p65. MHSM could also synergically act on the target DEGs of ALI and alleviate pulmonary pathological injury and inflammatory response, which might be achieved by inhibiting the expression of the key gene Ager-RAGE in RAGE/NF-κB signaling pathway and downstream signal NF-κB p65.


Subject(s)
Animals , Mice , Acute Lung Injury/genetics , Drugs, Chinese Herbal/pharmacology , Lipopolysaccharides , Lung/metabolism , NF-kappa B/metabolism , Network Pharmacology , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction
7.
Rio de Janeiro; s.n; 2019. 71 p. ilus.
Thesis in Portuguese | LILACS | ID: biblio-1053042

ABSTRACT

A doença hepática gordurosa não alcoólica (DHGNA) afeta um terço da população adulta e é definida como o acúmulo de gordura no fígado de indivíduos que não consomem excessiva quantidade de álcool. No entanto, os mecanismos moleculares responsáveis pela progressão da doença ainda não foram completamente elucidados. Estudos anteriores do nosso grupo e outros autores demonstraram que o aumento nos níveis de produtos finais de glicação avançada (AGEs) e distúrbios da microcirculação estão presentes na DHGNA, mas nenhuma evidência direta da ligação entre os níveis de AGEs, estresse oxidativo, inflamação e alterações microcirculatórias hepáticas foi demonstrada. No presente trabalho, nós investigamos os efeitos protetores da piridoxamina (PirPM), um inibidor da formação de AGEs, nas complicações metabólicas e microcirculatórias na DHGNA. A DHGNA foi induzida por uma dieta hiperlipídica (HFD) em ratos Wistar durante 28 semanas. O tratamento com Pir (60 mg/Kg/dia, i.p.) foi administrado entre as semanas 20 e 28. Na microcirculação do fígado, o recrutamento de leucócitos e o número de células estreladas hepáticas (HSC´s) ativadas foram examinados por microscopia intravital.


A perfusão tecidual foi acessada por fluxometria utilizando o laser speckle. O estresse oxidativo e o marcador de inflamação foram avaliados pela dosagem de substâncias reativas ao ácido tiobarbitúrico (TBARs) e RT-PCR, respectivamente. Os AGEs no fígado foram avaliados por espectroscopia de fluorescência. Foi observado que a alteração no metabolismo da glicose e aumento do peso corporal, hepático e de tecido adiposo e esteatose no grupo DHGNA foi revertido pelo tratamento com Pir. Em relação aos parâmetros microcirculatórios hepáticos, ratos com DHGNA apresentaram aumento do rolamento e adesão de leucócitos, da ativação de HSCs e diminuição da perfusão tecidual. A Pir mostrou um efeito vasoprotetor nas alterações da microcirculação hepática induzidas pela DHGNA. O fígado de grupo DHGNA apresentou níveis elevados de peroxidação lipídica, que foram parcialmente revertidos pelo tratamento com Pir. Em conclusão, a Pir apresenta importante efeito vasoprotetor e antioxidante, além de modular os distúrbios metabólicos, sendo, portanto, um potencial tratamento para complicações microcirculatórias e metabólicas associadas à DHGNA. (AU)


Subject(s)
Humans , Animals , Pyridoxamine , Metabolic Syndrome , Fatty Liver , Non-alcoholic Fatty Liver Disease , Receptor for Advanced Glycation End Products , Liver
8.
Journal of Southern Medical University ; (12): 1482-1486, 2019.
Article in Chinese | WPRIM | ID: wpr-781259

ABSTRACT

OBJECTIVE@#To investigate the correlation between soluble receptor for advanced glycation end products (sRAGE) level in the follicular fluid and ovarian responsiveness in non-PCOS patients undergoing controlled ovarian hyperstimulation.@*METHODS@#Ninety non-PCOS patients underwent IVF/ICSI using a short-acting long protocol for ovarian stimulation with a GnRH agonist. For each patient, the level of sRAGE in the follicular fluid was measured by enzyme linked immunosorbent assay (ELISA), and the data including the clinical baseline state, hormone level, number of oocytes obtained and the fertilization rate were collected.@*RESULTS@#Follicular fluid sRAGE level showed significant negative correlations with basal FSH level (=0.0036) and Gn dose ( 15) than in cases with oocytes obtained within the range of the target numbers (7-15) and below the target number (< 7) ( < 0.0001 and =0.0012, respectively).@*CONCLUSIONS@#Follicular fluid sRAGE level can reflect ovarian reserve function in non-PCOS patients, the number of oocytes obtained and the fertilization rate, and can thus predict ovarian responsiveness during controlled hyperstimulation in nonPCOS patients.


Subject(s)
Female , Humans , Fertilization in Vitro , Follicular Fluid , Ovarian Hyperstimulation Syndrome , Receptor for Advanced Glycation End Products
9.
Acta Academiae Medicinae Sinicae ; (6): 283-290, 2019.
Article in Chinese | WPRIM | ID: wpr-776036

ABSTRACT

Objective To investigate the effects of simvastatin on diabetic neuropathic pain and systematic inflammation in diabetic rats and explore their molecular mechanisms.Methods Totally 24 rats were equally randomized into the normal+vehicle(NV)group,diabetic+vehicle(DV)group,and diabetic+simvastatin(DS)group using the random number table.Streptozotocin(STZ)was used to establish the rat models of diabetes.Blood glucose,body mass,paw withdrawal mechanical threshold(PWMT),and paw withdrawal thermal latency(PWTL)in each group were observed on days 7,14,21,and 28 after STZ injection.On day 28 after STZ injection,rats were sacrificed,and the lumbar spinal dorsal horn and serum were collected.Western blotting was used to detect the expression of receptor for advanced glycation end products(RAGE)and the phosphorylation levels of protein kinase B(AKT),extracellular signal-regulated kinase(ERK),p38,and c-Jun N-terminal kinase(JNK)in the spinal dorsal horn of rats in each group.Enzyme-linked immunosorbent assay was performed to determine the serum concentrations of oxidized low density lipoprotein(ox-LDL)and interleukin-1β(IL-1β).Results On days 14,21 and 28 after STZ injection,the PWMT in DV group were(8.6 ± 0.8),(7.1 ± 1.6),and(7.8 ± 0.8)g respectively,which were significantly lower than (12.0 ± 0.9)(=8.482, =0.000),(11.6 ± 1.5)(=11.309, =0.000),and(11.7 ± 1.5)g(=9.801, =0.000)in NV group.The PWMT in DS group on days 21 and 28 were(9.4 ± 1.4)(=5.780, =0.000)and(9.7 ± 0.9)g(=4.775, =0.003),respectively,which were significantly improved comparing with those of DV group.On days 7,14,21,and 28,there were no significant differences in PWTL among these three groups (all <0.05).The expression of RAGE in the spinal dorsal horn of DV group was significantly higher than those of NV group(=6.299, =0.000)and DS group(=2.891, =0.025).The phosphorylation level of AKT in the spinal dorsal horn of DV group was significantly higher than those of NV group(=8.915,=0.000)and DS group(=4.103,=0.003).The phosphorylation levels of ERK( =8.313,=0.000),p38( =2.965, =0.022),and JNK(=7.459, =0.000)in the spinal dorsal horn of DV group were significantly higher than those of NV group;the phosphorylation level of JNK in the spinal dorsal horn of DS group was significant lower than that of DV group(=3.866, =0.004);however,there were no significant differences in the phosphorylation levels of ERK(=1.987,=0.122)and p38(=1.260,=0.375)in the spinal dorsal horn between DS group and DV group.The serum concentrations of ox-LDL and IL-1β in DV group were(41.86 ± 13.40)ng/ml and(108.16 ± 25.88)pg/ml,respectively,which were significantly higher than those in NV group [(24.66 ± 7.87)ng/ml(=3.606,=0.003)and(49.32 ± 28.35)pg/ml(=5.079,=0.000)] and DS group [(18.81 ± 5.62)ng/ml (=4.833, =0.000)and(32.73 ± 11.73)pg/ml(=6.510, =0.000)].Conclusions Simvastatin can relieve the mechanical allodynia of diabetic rats possibly by inhibiting the activation of RAGE/AKT and the phosphorylation of JNK in the spinal dorsal horn.Simvastatin can also decrease the serum concentrations of ox-LDL and IL-1β in diabetic rats,which may contribute to the relief of systematic inflammation.


Subject(s)
Animals , Rats , Diabetes Mellitus, Experimental , Hyperalgesia , Inflammation , Drug Therapy , Interleukin-1beta , Blood , Lipoproteins, LDL , Blood , Neuralgia , Drug Therapy , Proto-Oncogene Proteins c-akt , Metabolism , Random Allocation , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Metabolism , Simvastatin , Pharmacology
10.
Rev. Assoc. Med. Bras. (1992) ; 63(11): 971-977, Nov. 2017. tab, graf
Article in English | LILACS | ID: biblio-896309

ABSTRACT

Summary Objective: Interaction between advanced glycation end-products (AGEs) and receptor for AGEs (RAGE) in cells could affect both extracellular and intracellular structure and function, which plays a pivotal role in diabetic microvascular complications. The results from previous epidemiological studies on the association between RAGE gene -374T/A polymorphism and diabetic retinopathy (DR) risk were inconsistent. Thus, we conducted this meta-analysis to summarize the possible association between RAGE -374T/A polymorphism and DR risk. Method: We searched all relevant articles on the association between RAGE -374T/A polymorphism and DR risk from PubMed, Cochrane Library, ScienceDirect, Wanfang, VIP and Chinese National Knowledge Infrastructure (CNKI) web databases up to August 2016. Odds ratio (OR) with 95% confidence interval (CI) were calculated to assess those associations. All analyses were performed using the Review Manager software. Results: Nine case-control studies, including 1,705 DR cases and 2,236 controls were enrolled, and the results showed that the A allele of RAGE -374T/A polymorphism was significantly associated with increased DR risk in dominant model (TA/AA vs. TT: OR=1.22, 95CI 1.05-1.41, p=0.006) and heterozygote model (TA vs. TT: OR=1.26, 95CI 1.07-1.47, p=0.005). The subgroup analysis by ethnicity showed that significantly increased DR risk was found in both Asian and Caucasian populations. Conclusion: This meta-analysis reveals that the A allele of RAGE -374T/A polymorphism probably increase DR risk.


Subject(s)
Humans , Polymorphism, Genetic , Diabetes Mellitus, Type 2/genetics , Diabetic Retinopathy/genetics , Risk Factors , Genetic Predisposition to Disease , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/etiology , Alleles , Receptor for Advanced Glycation End Products , Genotype
11.
Biol. Res ; 50: 23, 2017. graf
Article in English | LILACS | ID: biblio-950874

ABSTRACT

BACKGROUND: Airway remodeling is a key feature of asthma, characterized by increased proliferation of airway smooth muscle cells (ASMCs). S100A8 is a calcium-binding protein with a potential to regulate cell proliferation. Here, the effect of exogenous S100A8 protein on the proliferation of ASMCs induced by platelet-derived growth factor (PDGF) and the underlying molecular mechanism was investigated. METHODS: Rat ASMCs were cultured with or without a neutralizing antibody to the receptor for advanced glycation end-products (RAGE), a potential receptor for S100A8 protein. Purified recombinant rat S100A8 protein was then added into the cultured cells, and the proliferation of ASMCs induced by PDGF was detected by colorimetric-based WST-8 assay and ampedance-based xCELLigence proliferation assay. The expression levels of RAGE in ASMCs were analyzed using western blotting assay. RESULTS: Results showed that exogenous S100A8 inhibited the PDGF-induced proliferation of rat ASMCs in a dose- dependent manner with the maximal effect at 1 µg/ml in vitro. Furthermore, when ASMCs was pre-treated with anti-RAGE neutralizing antibody, the inhibitory effect of S100A8 on PDGF-induced proliferation was significantly suppressed. In addition, neither the treatment with S100A8 or PDGF alone nor the pre-treatment with rS100A8 followed by PDGF stimulation affected the expression levels of RAGE. CONCLUSIONS: Our study demonstrated that S100A8 inhibits PDGF-induced ASMCs proliferation in a manner dependent on membrane receptor RAGE.


Subject(s)
Animals , Rats , Platelet-Derived Growth Factor/agonists , Myocytes, Smooth Muscle/drug effects , Calgranulin A/administration & dosage , Cell Proliferation/drug effects , Receptor for Advanced Glycation End Products/drug effects , Cells, Cultured
12.
Journal of Breast Cancer ; : 27-34, 2017.
Article in English | WPRIM | ID: wpr-148359

ABSTRACT

PURPOSE: The high mobility group box 1 (HMGB1) protein has roles in apoptosis and immune responses by acting as a ligand for receptor for advanced glycation end products (RAGE), Toll-like receptors (TLRs), and triggering receptor expressed on myeloid cells 1. In particular, HMGB1/RAGE is involved in tumor metastasis by inducing matrix metalloproteinase 2 (MMP2) and MMP9 expression. We investigated the associations between genetic variations in HMGB1-related genes and disease-free survival (DFS) and overall survival (OS) in Korean female breast cancer patients. METHODS: A total of 2,027 patients in the Seoul Breast Cancer Study were included in the analysis. One hundred sixteen single nucleotide polymorphisms (SNPs) were extracted from eight genes. A multivariate Cox proportional hazards model was used to estimate the hazard ratio and 95% confidence interval (CI) of each SNP. The effects of the SNPs on breast cancer prognosis were assessed at cumulative levels with polygenic risk scores. RESULTS: The SNPs significantly associated with DFS were rs243867 (hazard ratio, 1.26; 95% CI, 1.05–1.50) and rs243842 (hazard ratio, 1.24; 95% CI, 1.03–1.50); both SNPs were in MMP2. The SNPs significantly associated with OS were rs243842 in MMP2 (hazard ratio, 1.33; 95% CI 1.03–1.71), rs4145277 in HMGB1 (hazard ratio, 1.29; 95% CI, 1.00–1.66), rs7656411 in TLR2 (hazard ratio, 0.76; 95% CI, 0.60–0.98), and rs7045953 in TLR4 (hazard ratio, 0.50; 95% CI, 0.29–0.84). The polygenic risk score results for the DFS and OS patients showed third tertile hazard ratios of 1.72 (95% CI, 1.27–2.34) and 2.75 (95% CI, 1.79–4.23), respectively, over their first tertile references. CONCLUSION: The results of the present study indicate that genetic polymorphisms in HMGB1-related genes are related to breast cancer prognosis in Korean women.


Subject(s)
Female , Humans , Receptor for Advanced Glycation End Products , Apoptosis , Breast Neoplasms , Breast , Disease-Free Survival , Genetic Predisposition to Disease , Genetic Variation , HMGB1 Protein , Matrix Metalloproteinase 2 , Myeloid Cells , Neoplasm Metastasis , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Prognosis , Proportional Hazards Models , Seoul , Toll-Like Receptors
13.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 844-855, 2016.
Article in English | WPRIM | ID: wpr-812568

ABSTRACT

The aim of this study was to investigate the effects of high-advanced glycation end products (AGEs) diet on diabetic vascular complications. The Streptozocin (STZ)-induced diabetic mice were fed with high-AGEs diet. Diabetic characteristics, indicators of renal and cardiovascular functions, and pathohistology of pancreas, heart and renal were evaluated. AGEs/RAGE/ROS pathway parameters were determined. During the experiments, the diabetic mice exhibited typical characteristics including weight loss, polydipsia, polyphagia, polyuria, high-blood glucose, and low-serum insulin levels. However, high-AGEs diet effectively aggravated these diabetic characteristics. It also increased the 24-h urine protein levels, serum levels of urea nitrogen, creatinine, c-reactive protein (CRP), low density lipoprotein (LDL), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in the diabetic mice. High-AGEs diet deteriorated the histology of pancreas, heart, and kidneys, and caused structural alterations of endothelial cells, mesangial cells and podocytes in renal cortex. Eventually, high-AGEs diet contributed to the high-AGE levels in serum and kidneys, high-levels of reactive oxygen species (ROS) and low-levels of superoxide dismutase (SOD) in serum, heart, and kidneys. It also upregulated RAGE mRNA and protein expression in heart and kidneys. Our results showed that high-AGEs diet deteriorated vascular complications in the diabetic mice. The activation of AGEs/RAGE/ROS pathway may be involved in the pathogenesis of vascular complications in diabetes.


Subject(s)
Animals , Humans , Male , Mice , Diabetes Mellitus, Experimental , Metabolism , Diabetic Angiopathies , Genetics , Metabolism , Diet , Glycation End Products, Advanced , Metabolism , Interleukin-6 , Metabolism , Kidney , Metabolism , Mice, Inbred C57BL , Oxidative Stress , Pancreas , Metabolism , Reactive Oxygen Species , Metabolism , Receptor for Advanced Glycation End Products , Genetics , Metabolism , Superoxide Dismutase , Metabolism , Tumor Necrosis Factor-alpha , Metabolism
14.
Braz. j. med. biol. res ; 48(6): 545-552, 06/2015. tab, graf
Article in English | LILACS | ID: lil-748222

ABSTRACT

Abnormal high mobility group protein B1 (HMGB1) activation is involved in the pathogenesis of pulmonary fibrosis. Pulmonary rehabilitation mixture (PRM), which combines extracts from eight traditional Chinese medicines, has very good lung protection in clinical use. However, it is not known if PRM has anti-fibrotic activity. In this study, we investigated the effects of PRM on transforming growth factor-β1 (TGF-β1)-mediated and bleomycin (BLM)-induced pulmonary fibrosis in vitro and in vivo. The effects of PRM on TGF-β1-mediated epithelial-mesenchymal transition (EMT) in A549 cells, on the proliferation of human lung fibroblasts (HLF-1) in vitro, and on BLM-induced pulmonary fibrosis in vivo were investigated. PRM treatment resulted in a reduction of EMT in A549 cells that was associated with attenuating an increase of vimentin and a decrease of E-cadherin. PRM inhibited the proliferation of HLF-1 at an IC50 of 0.51 µg/mL. PRM ameliorated BLM-induced pulmonary fibrosis in rats, with reduction of histopathological scores and collagen deposition, and a decrease in α-smooth muscle actin (α-SMA) and HMGB1 expression. An increase in receptor for advanced glycation end-product (RAGE) expression was found in BLM-instilled lungs. PRM significantly decreased EMT and prevented pulmonary fibrosis through decreasing HMGB1 and regulating RAGE in vitro and in vivo. PRM inhibited TGF-β1-induced EMT via decreased HMGB1 and vimentin and increased RAGE and E-cadherin levels. In summary, PRM prevented experimental pulmonary fibrosis by modulating the HMGB1/RAGE pathway.


Subject(s)
Animals , Humans , Male , Drugs, Chinese Herbal/pharmacology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/prevention & control , Antibiotics, Antineoplastic , Receptor for Advanced Glycation End Products/drug effects , Apoptosis/drug effects , Bleomycin , Blotting, Western , Cells, Cultured , Collagen/drug effects , Complex Mixtures/pharmacology , Drugs, Chinese Herbal/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Fibroblasts/drug effects , HMGB1 Protein/drug effects , Hydroxyproline/analysis , Immunohistochemistry , Lung/drug effects , Lung/pathology , Platelet-Derived Growth Factor/drug effects , Pulmonary Fibrosis/pathology , Random Allocation , Rats, Sprague-Dawley , Reproducibility of Results , Transforming Growth Factor beta1/drug effects
15.
São Paulo; s.n; s.n; 2015. 158 p. graf, tab, ilus.
Thesis in Portuguese | LILACS | ID: biblio-881862

ABSTRACT

Concentrações séricas basais da proteína amiloide sérica A (SAA) estão significativamente aumentadas em pacientes com câncer e alguns autores sugerem uma relação causal. Trabalho anterior do grupo mostrou que a SAA induz a proliferação de duas linhagens de glioblastoma humano e afeta os processos de invasividade in vitro, sustentando um papel pró-tumoral para esta proteína. Com base nesse trabalho, investigamos a abrangência dos efeitos de SAA para outro tipo de célula tumoral e para isso escolhemos um painel de linhagens de melanoma humano e uma linhagem primária obtida a partir de aspirado de linfonodo de paciente com melanoma, por nós isolada. Observamos que apesar da célula precursora de melanomas, isto é, melanócito, não produzir SAA, todas as linhagens de melanoma produziram a proteína e expressaram alguns dos seus receptores. Além disso, quando estas células foram estimuladas com SAA houve uma inibição da proliferação em tempos curtos de exposição (48 horas) e efeitos citotóxicos após um tempo maior (7 dias). A SAA também afetou processos de invasividade e a produção das citocinas IL-6, IL-8 e TNF-α. Aos avaliarmos o efeito da SAA na interação das células de melanoma com células do sistema imune, vimos que a SAA ativou uma resposta imune anti-tumoral aumentando a expressão de moléculas co-estumolatórias, como CD69 e HLA-DR, e sua função citotóxica. Ainda, vimos que a produção de TNF-α, IFN-γ, IL-10, IL-1ß e IL-8 estimuladas por SAA podem contribuir com os efeitos desta. De forma geral estes resultados nos levam a crer que a SAA tem atividade anti-tumoral em melanomas. Finalizando, com base na importância do desenvolvimento da resistência às terapias atuais para o melanoma, observamos que em células resistentes ao PLX4032, um inibidor de BRAF, os efeitos imunomodulatórios induzidos pela SAA estão abolidos, possivelmente identificando um novo componente da resistência


Basal serum concentrations of the protein serum amyloid A are significantly increased in cancer patients and some authors suggest a causal relationship. Previous work of our research group showed that SAA induces proliferation of two cell lines of human glioblastoma and affects invasiveness processes in vitro, supporting a pro-tumor role for this protein. Based on this work, we investigated the extent of SAA effects to another type of tumor cell and we chose a panel of human melanoma cell lines and primary line obtained from a patient with melanoma by lymph node aspirate. Melanoma cells were isolated by us. We observed that while the precursor cells of melanoma, melanocytes, do not produce SAA, all melanoma cell lines expressed the protein and produced some of their receptors. Moreover, when these cells were stimulated with SAA there was an inhibition of proliferation in short exposure times (48 hours) and cytotoxic effects after a longer period (7 days). SAA also affected invasive procedures and the production of the cytokines IL-6, IL-8 and TNF-α. To evaluate the SAA effect in the interaction of melanoma cells with immune system cells, we found that SAA activated an anti-tumor immune response by increasing the expression of co-estimulatory molecules such as CD69 and HLA-DR, and their cytotoxic function. Furthermore, we found that the production of TNF-α, IFN-γ, IL-10, IL-1ß and IL-8 stimulated by SAA can contribute to this effect. In general these results lead us to believe that the SAA has anti-tumor activity in melanomas. Finally, based on the importance of the resistance development to current therapies for melanoma we observed that in cells resistant to PLX4032, a BRAF inhibitor, the immunomodulatory effects induced by SAA are abolished, possibly identifying a new resistance component


Subject(s)
Serum Amyloid A Protein/analysis , Serum Amyloid A Protein/adverse effects , Melanoma/physiopathology , Gene Expression , Proto-Oncogene Proteins B-raf/adverse effects , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Cell Migration Assays/instrumentation , Receptor for Advanced Glycation End Products/genetics
16.
Chinese Journal of Contemporary Pediatrics ; (12): 81-85, 2015.
Article in Chinese | WPRIM | ID: wpr-289464

ABSTRACT

<p><b>OBJECTIVE</b>To explore the change of RAGE-NF-κB signaling pathway during the course of hyperoxia-induced lung injury in newborn rats, and the effect of glucocorticoid on this pathway.</p><p><b>METHODS</b>Twenty-four Sprague-Dawley neonatal rats were randomly divided into three groups (n=8 each) : sham control (control group), hyperoxia-induced acute lung injury (model group) and glucocorticoid-treated acute lung injury (glucocorticoid group). Rats were sacrificed at 13 days after birth. RAGE and NF-κB expression levels in lung tissues were detected by reverse transcription polymerase chain reaction, Western blot and immunohistochemistry analysis. The levels of tumor necrosis factor α (TNF-α) and sRAGE in bronchoalveolar lavage fluid (BALF) and serum were measured using ELISA. Lung damage was evaluated by histological examinations.</p><p><b>RESULTS</b>RAGE and NF-κB mRNA and protein expression levels in lung tissues were significantly increased in the model and glucocorticoid groups compared with the control group (P<0.05). Serum RAGE concentrations were significantly increased but RAGE concentrations in BALF were significantly reduced in the model and glucocorticoid groups compared with the control group (P<0.05). RAGE and NF-κB expression at both mRNA and protein levels in lung tissues was significantly lower in the glucocorticoid group than in the model group (P<0.05). RAGE concentrations were significantly lower in serum (P<0.05), but were higher in BALF (P<0.05) in the glucocorticoid group than in the model group.</p><p><b>CONCLUSIONS</b>RAGE-NF-κB pathway plays an important role in hyperoxia-induced lung injury in neonatal rats, and glucocorticoid administration may play a protective role against the lung injury by down-regulating RAGE-NF-κB signaling pathway.</p>


Subject(s)
Animals , Rats , Animals, Newborn , Glucocorticoids , Pharmacology , Hyperoxia , Lung Injury , NF-kappa B , Genetics , Physiology , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Receptors, Immunologic , Genetics , Physiology , Signal Transduction , Tumor Necrosis Factor-alpha
17.
Journal of Southern Medical University ; (12): 6-11, 2015.
Article in Chinese | WPRIM | ID: wpr-329183

ABSTRACT

<p><b>OBJECTIVE</b>To investigate lipopolysaccharide (LPS)-induced changes of cytoskeletal filamentous actin in primary isolated pulmonary microvascular endothelial cells (PMVECs) from wild-type and RAGE knock-out mouse.</p><p><b>METHODS</b>The lungs of wild-type and RAGE knock-out mice were digested with collagenase type I to obtain endothelial cells purified by anti-CD31-coupled magnetic beads. The PMVEC identified by factor VIII labeling were stimulated with LPS at different concentrations and the changes of filamentous actin were observed by confocal microscopy.</p><p><b>RESULTS</b>The cultured primary cells showed typical endothelial cell phenotype as examined with factor VIII labeling. LPS stimulation caused rearrangement of the cytoskeletal filament F-actin in wild-type mouse PMVECs with stress fiber formation, but such changes were not obvious in RAGE knock-out mouse PMVECs.</p><p><b>CONCLUSION</b>Mouse PMVECs of a high purity can be obtained by immune magnetic beads. RAGE is involved in LPS-induced destruction of mouse PMVEC cytoskeletons.</p>


Subject(s)
Animals , Mice , Actins , Metabolism , Cells, Cultured , Cytoskeleton , Metabolism , Endothelial Cells , Cell Biology , Lipopolysaccharides , Lung , Cell Biology , Mice, Knockout , Microvessels , Cell Biology , Phenotype , Receptor for Advanced Glycation End Products , Receptors, Immunologic , Genetics , Metabolism
18.
Rev. bras. ter. intensiva ; 26(4): 392-396, Oct-Dec/2014. tab, graf
Article in Portuguese | LILACS | ID: lil-732925

ABSTRACT

Objetivo: Avaliar e compreender as implicações clínicas dos níveis plasmáticos de uma isoforma solúvel de um receptor de produtos finais de glicação avançada (do inglês receptor for advanced glycation end products - sRAGE) em diferentes fases da sepse. Métodos: Os valores do sRAGE sérico em pacientes divididos nos grupos controle na unidade de terapia intensiva, sepse grave, choque séptico e recuperação de choque séptico foram analisados do ponto de vista estatístico para avaliar a quantidade (Kruskal-Wallis), variabilidade (teste de Levine) e correlação (teste Spearman rank) em relação a certos mediadores inflamatórios (IL-1 α, IL-6, IL-8, IL-10, IP-10, G-CSF, MCP-1, IFN-γ e TNF-α). Resultados: Não se observaram modificações nos níveis de sRAGE entre os grupos; contudo o grupo com choque séptico demonstrou diferenças na variabilidade do sRAGE em comparação aos demais grupos. Foi relatada, no grupo com choque séptico, uma correlação positiva com todos os mediadores inflamatórios. Conclusão: Os níveis de sRAGE se associaram com desfechos piores nos pacientes com choque séptico. Entretanto, uma análise de correlação estatística com outras citocinas pró-inflamatórias indicou que as vias que levam a esses desfechos são diferentes, dependendo dos níveis de sRAGE. ...


Objective: To evaluate and understand the clinical implications of the plasma levels of a soluble isoform of a receptor for advanced glycation end products (sRAGE) in different stages of sepsis. Methods: Serum sRAGE values in patients who were divided into intensive care unit control, severe sepsis, septic shock and recovery from septic shock groups were statistically analyzed to assess quantity (Kruskal-Wallis), variability (Levine test) and correlation (Spearman rank test) with certain inflammatory mediators (IL-1 α, IL-6, IL-8, IL-10, IP-10, G-CSF, MCP-1, IFN-γ and TNF-α). Results: No changes in sRAGE levels were observed among the groups; however, the septic shock group showed differences in the variability of sRAGE compared to the other groups. A positive correlation with all the inflammatory mediators was reported in the septic shock group. Conclusion: sRAGE levels are associated with worse outcomes in patients with septic shock. However, a statistical correlation analysis with other proinflammatory cytokines indicated that the pathways leading to those outcomes are different depending on the sRAGE levels. Future studies to elucidate the pathophysiological mechanisms involving sRAGE in models of sepsis are of great clinical importance for the safe handling of this biomarker. .


Subject(s)
Humans , Receptor for Advanced Glycation End Products/blood , Inflammation Mediators/metabolism , Shock, Septic/blood , Biomarkers/blood , Cohort Studies , Prospective Studies , Shock, Septic/mortality , Shock, Septic/physiopathology
19.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 920-928, 2014.
Article in English | WPRIM | ID: wpr-812184

ABSTRACT

Qifu-Yin (QFY), a widely used formula of traditional Chinese medicine (TCM) derived from "Jingyue Quanshu", is one of the most commonly used TCM prescriptions for the clinical treatment of Alzheimer disease. The role of advanced glycation end products (AGEs) and its receptor RAGE have attracted increasing attention as the pivotal role of Aβ has been questioned. The present study was designed to test the neuroprotective effects of QFY, and the possible mechanism in AGE-induced Alzheimer model rats. After injection of AGE in the CA3 area of the hippocampus, QFY (8.6, 4.3, and 2.15 g·kg(-1)), and a positive control drug donepezil (2 mg·kg(-1)) were administrated through gastric intubation to rats once daily for thirty consecutive days. Another positive control group was the AGE + anti-RAGE group, which was simultaneously injected with anti-RAGE antibody before AGE treatment. The control group, sham-operated group, as well as the AGE + anti-RAGE group received saline at the same dosage. The Morris water maze test and the step-down passive avoidance test were conducted to evaluate the cognitive function of the rats. The expression of RAGE and NF-κB were assayed by immunohistochemical staining. The levels of Aβ, TNF-α, and IL-1β in the hippocampus were measured by enzyme-linked immunosorbent assay (ELISA). The results showed that QFY could significantly attenuate the memory impairment induced by AGE, decrease the expressions of RAGE and NF-κB, and reduce the levels of Aβ, TNF-α, and IL-1β in the hippocampus in a dose-dependent manner. Also, the blockage of RAGE could significantly reduce the impairments caused by AGEs. In conclusion, QFY could attenuate AGEs-induced, Alzheimer-like pathophysiological changes. These neuroprotective effects might be related to the RAGE/NF-κB pathway and its anti-inflammatory activity.


Subject(s)
Animals , Male , Alzheimer Disease , Drug Therapy , Metabolism , Amyloid beta-Peptides , Metabolism , Anti-Inflammatory Agents , Pharmacology , Therapeutic Uses , Brain , Metabolism , Dose-Response Relationship, Drug , Drugs, Chinese Herbal , Pharmacology , Therapeutic Uses , Glycation End Products, Advanced , Interleukin-1beta , Metabolism , Learning , Magnoliopsida , Memory Disorders , Drug Therapy , Metabolism , NF-kappa B , Metabolism , Phytotherapy , Plants, Medicinal , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Receptors, Immunologic , Metabolism , Signal Transduction , Tumor Necrosis Factor-alpha , Metabolism
20.
Chinese Journal of Applied Physiology ; (6): 306-313, 2014.
Article in Chinese | WPRIM | ID: wpr-236320

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effects of advanced glycosylation end products (AGEs) modified bovine serum albumin (AGE-BSA) on the expression of reactive oxygen species (ROS) and monocyte chemoattractant protein-1 (MCP-1) in human renal mesangial cells (HRMCs).</p><p><b>METHODS</b>HRMCs were cultured in vitro with medium containing different doses of AGE-BSA or BSA (50,100, 200, 400 mg/L) for 48 hours, or with AGE-BSA (200 mg/L) for different times (12, 24, 48, 72 h). Immunocytochemistry assay was used to estimate the protein level of RAGE. The ROS in cells were measured by flow cytometry and the mRNA expression of MCP-1 were analyzed by semi-quantiative reverse transcription-polymerase chain reaction (RT-PCR) after treatment with AGE-BSA or BSA.</p><p><b>RESULTS</b>The protein level of RAGE was upregulated in the HRMCs with AGE-BSA. The expression of ROS and MCP-1 significantly enhanced by incubation of AGE-BSA in a time- and dose-dependent manner. The effects of AGE-BSA-induced up-regulation of ROS and MCP-1 level was significantly blocked by neutralizing antibodies to RAGE, while the expression of ROS and MCP-1 stood nearly unchanged after cultured with huamn IgG.</p><p><b>CONCLUSION</b>The expression of ROS and MCP-1 in HRMCs is induced by AGE-BSA through RAGE, which may have potential effects in the pathgenic mechanism of diabetic nephropathy.</p>


Subject(s)
Humans , Cells, Cultured , Chemokine CCL2 , Metabolism , Glycation End Products, Advanced , Pharmacology , Mesangial Cells , Metabolism , Oxidative Stress , Reactive Oxygen Species , Metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic , Metabolism , Serum Albumin, Bovine , Pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL