Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Postgrad Med ; 134(8): 763-775, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36094155

RESUMO

Smoking is a well-established risk factor for chronic obstructive pulmonary disease (COPD). Chronic lung inflammation continues even after smoking cessation and leads to COPD progression. To date, anti-inflammatory therapies are ineffective in improving pulmonary function and COPD symptoms, and new molecular targets are urgently needed to deal with this challenge. The receptor for advanced glycation end-products (RAGE) was shown to be relevant in COPD pathogenesis, since it is both a genetic determinant of low lung function and a determinant of COPD susceptibility. Moreover, RAGE is involved in the physiological response to cigarette smoke exposure. Since innate and acquired immunity plays an essential role in the development of chronic inflammation and emphysema in COPD, here we summarized the roles of RAGE and its ligand HMGB1 in COPD immunity.


Assuntos
Proteína HMGB1 , Doença Pulmonar Obstrutiva Crônica , Humanos , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Ligantes , Pulmão
2.
Dis Markers ; 2022: 4487435, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35280439

RESUMO

Background: Receptor for advanced glycation end products (RAGE) is implicated in tumor biology. Released high mobility group box protein 1 (HMGB1) ligand binding to RAGE receptor in tumor cells promotes tumor progression. The mechanisms of HMGB1-RAGE signaling in M2 macrophages involved in lymphangiogenesis in laryngeal carcinoma remain poorly understood. Here, we assessed the effect of HMGB1-RAGE signaling on M2 macrophages in lymphangiogenesis. Methods: HMGB1, CD163, and D2-40 in laryngeal squamous cell carcinoma (LSCC, n = 123), laryngeal precursor lesions (LPLs, n = 102), and vocal polyp (VP, n = 55) were analyzed by immunohistochemistry. THP-1 cell-expressed RAGE gene was knocked down and then polarized to M0 macrophages and M2 macrophages. IL-23, TNF-α, TGF-ß, and IL-10 were measured by ELISA; IL-1ß, IL-12, IL-10, and CCL-13 were evaluated by RT-qPCR, and CD206, CD163, and RAGE were evaluated by western blot to evaluate whether classical M2 macrophages were obtained. Conditioned media from RAGE+/- M0 macrophages and RAGE+/- M2 macrophages incubated in the presence or absence of HMGB1, anti-Toll-like receptor (TLR)2, anti-TLR4 antibodies, and anti-VEGF-C antibodies were collected separately for human dermal lymphatic endothelial cells (HDLEC) for proliferation, migration, lymphangiogenesis assay, and VEGF-C concentration analysis. Results: HMGB1 and M2 macrophage densities were increased in LSCC (P < 0.01). HMGB1 and M2 macrophage densities were significantly correlated with lymphatic vessel density (LVD) in LSCC (P < 0.01). The HMGB1 overexpression and higher M2 macrophage density were involved in lymph node metastasis (P < 0.01) and poor prognosis (P < 0.05). In vitro, conditioned medium from HMGB1-stimulated RAGE+ M2 macrophages activated lymphangiogenesis by upregulating the VEGF compared to controls (P < 0.05). On the contrary, RAGE knockdown obviously decreased the corresponding effects of HMGB1-preconditioned M2 macrophages upon HDLEC (P < 0.05). HMGB1-TLR pathway does not significantly increase HDLEC proliferation, migration, and lymphangiogenesis on M2 macrophages. Conclusions: HMGB1 promotes lymphangiogenesis by activation of RAGE on M2 macrophages. Targeting RAGE may provide an effective therapeutic strategy against M2 macrophages in LSCC patients with lymph node metastasis.


Assuntos
Carcinoma de Células Escamosas/etiologia , Proteína HMGB1/fisiologia , Neoplasias Laríngeas/etiologia , Linfangiogênese , Macrófagos/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
3.
Front Immunol ; 12: 697071, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745088

RESUMO

Background: High mobility group box 1 (HMGB1) causes microvascular endothelial cell barrier dysfunction during acute lung injury (ALI) in sepsis, but the mechanisms have not been well understood. We studied the roles of RAGE and Rho kinase 1 (ROCK1) in HMGB1-induced human pulmonary endothelial barrier disruption. Methods: In the present study, the recombinant human high mobility group box 1 (rhHMGB1) was used to stimulate human pulmonary microvascular endothelial cells (HPMECs). The endothelial cell (EC) barrier permeability was examined by detecting FITC-dextran flux. CCK-8 assay was used to detect cell viability under rhHMGB1 treatments. The expression of related molecules involved in RhoA/ROCK1 pathway, phosphorylation of myosin light chain (MLC), F-actin, VE-cadherin and ZO-1 of different treated groups were measured by pull-down assay, western blot and immunofluorescence. Furthermore, we studied the effects of Rho kinase inhibitor (Y-27632), ROCK1/2 siRNA, RAGE-specific blocker (FPS-ZM1) and RAGE siRNA on endothelial barrier properties to elucidate the related mechanisms. Results: In the present study, we demonstrated that rhHMGB1 induced EC barrier hyperpermeability in a dose-dependent and time-dependent manner by measuring FITC-dextran flux, a reflection of the loss of EC barrier integrity. Moreover, rhHMGB1 induced a dose-dependent and time-dependent increases in paracellular gap formation accompanied by the development of stress fiber rearrangement and disruption of VE-cadherin and ZO-1, a phenotypic change related to increased endothelial contractility and endothelial barrier permeability. Using inhibitors and siRNAs directed against RAGE and ROCK1/2, we systematically determined that RAGE mediated the rhHMGB1-induced stress fiber reorganization via RhoA/ROCK1 signaling activation and the subsequent MLC phosphorylation in ECs. Conclusion: HMGB1 is capable of disrupting the endothelial barrier integrity. This study demonstrates that HMGB1 activates RhoA/ROCK1 pathway via RAGE, which phosphorylates MLC inducing stress fiber formation at short time, and HMGB1/RAGE reduces AJ/TJ expression at long term independently of RhoA/ROCK1 signaling pathway.


Assuntos
Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Proteína HMGB1/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Quinases Associadas a rho/fisiologia , Células Cultivadas , Humanos , Cadeias Leves de Miosina/fisiologia , Transdução de Sinais/fisiologia
4.
Sci Rep ; 11(1): 17373, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34462492

RESUMO

Advanced glycation endproducts (AGEs) are involved in several diseases, including NAFLD and NASH. RAGE is the main receptor mediating the pro-inflammatory signalling induced by AGEs. Therefore, targeting of RAGE has been proposed for prevention of chronic inflammatory diseases. However, the role of RAGE in the development of NAFLD and NASH remains poorly understood. We thus aimed to analyse the effect of obesity on AGEs accumulation, AGE-receptors and AGE-detoxification, and whether the absence of RAGE might improve hepatosteatosis and inflammation, by comparing the liver of lean control, obese (LeptrDb-/-) and obese RAGE-deficient (RAGE-/- LeptrDb-/-) mice. Obesity induced AGEs accumulation and RAGE expression with hepatosteatosis and inflammation in LeptrDb-/-, compared to lean controls. Despite the genetic deletion of RAGE in the LeptrDb-/- mice, high levels of intrahepatic AGEs were maintained accompanied by decreased expression of the protective AGE-receptor-1, impaired AGE-detoxifying system glyoxalase-1, and increased expression of the alternative AGE-receptor galectin-3. We also found sustained hepatosteatosis and inflammation as determined by persistent activation of the lipogenic SREBP1c and proinflammatory NLRP3 signalling pathways. Thus, RAGE targeting is not effective in the prevention of NAFLD in conditions of obesity, likely due to the direct liver specific crosstalk of RAGE with other AGE-receptors and AGE-detoxifying systems.


Assuntos
Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Tecido Adiposo/metabolismo , Animais , Feminino , Deleção de Genes , Inflamassomos , Inflamação/metabolismo , Lipídeos/química , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Receptores Imunológicos/metabolismo , Transdução de Sinais
5.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34360919

RESUMO

Hypoxia is characterized by an inadequate supply of oxygen to tissues, and hypoxic regions are commonly found in solid tumors. The cellular response to hypoxic conditions is mediated through the activation of hypoxia-inducible factors (HIFs) that control the expression of a large number of target genes. Recent studies have shown that the receptor for advanced glycation end products (RAGE) participates in hypoxia-dependent cellular adaptation. We review recent evidence on the role of RAGE signaling in tumor biology under hypoxic conditions.


Assuntos
Hipóxia Celular , Produtos Finais de Glicação Avançada/metabolismo , Neoplasias/metabolismo , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Animais , Linhagem Celular Tumoral , Humanos
6.
Pharmacol Res ; 169: 105650, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33964468

RESUMO

Stroke is one of the leading causes of death worldwide. Hemorrhagic transformation (HT) is a common serious complication of ischemic stroke (IS) and is related to poor prognosis. Hyperglycemia after stroke is associated with the occurrence of HT and seriously affects the clinical treatment of stroke. Our previous experiments demonstrated that the Xiao-Xu-Ming decoction effective components group (XXMD), which is a Chinese medicine formula reconstituted by active ingredients, has multiple pharmacological effects in the treatment of IS. However, the effects of XXMD on HT after IS remain unclear. Thus, we investigated the preventive effects of XXMD on hyperglycemia-induced HT and further explored the underlying mechanism. Acute hyperglycemia combined with the electrocoagulation cerebral ischemia model was used to establish the HT model. XXMD (37.5, 75, 150 mg/kg/d) was given by gavage for 5 days. Network pharmacology was used to predict potential targets and pathways of XXMD in HT occurrence, and further studies confirmed the related targets. The results showed that hyperglycemia aggravated neurological deficits and blood-brain barrier (BBB) disruption, leading to intracerebral hemorrhage. Pretreatment with XXMD improved neurological function and BBB integrity and inhibited HT occurrence. Network pharmacology revealed that AGE-RAGE-mediated neuroinflammation may be associated with hyperglycemia-induced HT. Further studies confirmed that hyperglycemia activated the AGE-RAGE signaling pathway, increased the expression of HMGB1, TLR4 and p-p65, and induced the release of inflammatory factors and neutrophil infiltration, leading to HT. XXMD could inhibit AGE-RAGE-mediated neuroinflammation. These findings indicated that pretreatment with XXMD alleviated hyperglycemia-induced HT, which may be associated with the inhibition of AGE-RAGE-mediated neuroinflammation. Therefore, XXMD may be a potential therapeutic drug for HT.


Assuntos
Hemorragia Cerebral/prevenção & controle , Medicamentos de Ervas Chinesas/uso terapêutico , Hiperglicemia/tratamento farmacológico , Doenças Neuroinflamatórias/prevenção & controle , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Animais , Isquemia Encefálica/complicações , Hemorragia Cerebral/tratamento farmacológico , Hiperglicemia/complicações , Masculino , Farmacologia em Rede/métodos , Doenças Neuroinflamatórias/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Receptor para Produtos Finais de Glicação Avançada/fisiologia
7.
Sci Rep ; 11(1): 8336, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863932

RESUMO

Thoracic dorsal root ganglia (tDRG) contribute to fluid secretion in the upper airways. Inflammation potentiates DRG responses, but the mechanisms remain under investigation. The receptor for advanced glycation end-products (RAGE) underlies potentiation of DRG responses in pain pathologies; however, its role in other sensory modalities is less understood. We hypothesize that RAGE contributes to electrophysiological and biochemical changes in tDRGs during inflammation. We used tDRGs and tracheas from wild types (WT), RAGE knock-out (RAGE-KO), and with the RAGE antagonist FPS-ZM1, and exposed them to lipopolysaccharides (LPS). We studied: capsaicin (CAP)-evoked currents and action potentials (AP), tracheal submucosal gland secretion, RAGE expression and downstream pathways. In WT neurons, LPS increased CAP-evoked currents and AP generation, and it caused submucosal gland hypersecretion in tracheas from WT mice exposed to LPS. In contrast, LPS had no effect on tDRG excitability or gland secretion in RAGE-KO mice or mice treated with FPS-ZM1. LPS upregulated full-length RAGE (encoded by Tv1-RAGE) and downregulated a soluble (sRAGE) splice variant (encoded by MmusRAGEv4) in tDRG neurons. These data suggest that sensitization of tDRG neurons contributes to hypersecretion in the upper airways during inflammation. And at least two RAGE variants may be involved in these effects of LPS.


Assuntos
Gânglios Espinais/fisiopatologia , Lipopolissacarídeos/efeitos adversos , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Mucosa Respiratória/metabolismo , Traqueia/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Benzamidas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
Int J Cancer ; 148(5): 1233-1244, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33205453

RESUMO

Tumor-associated macrophages (TAMs) are crucial components of the tumor microenvironment. They play vital roles in hepatocellular carcinoma (HCC) progression. However, the interactions between TAMs and HCC cells have not been fully characterized. In this study, TAMs were induced using human monocytic cell line THP-1 cells in vitro to investigate their functions in HCC progression. S100 calcium-binding protein A9 (S100A9), an inflammatory microenvironment-related secreted protein, was identified to be significantly upregulated in TAMs. S100A9 expression in tumor tissues was associated with poor survival of HCC patients. It could enhance the stem cell-like properties of HepG2 and MHCC-97H cells by activating nuclear factor-kappa B signaling pathway through advanced glycosylation end product-specific receptor in a Ca2+ -dependent manner. Furthermore, we found that, after treatment with S100A9, HepG2 and MHCC-97H cells recruited more macrophages via chemokine (CC motif) ligand 2, which suggests a positive feedback between TAMs and HCC cells. Taken together, our findings reveal that TAMs could upregulate secreted protein S100A9 and enhance the stem cell-like properties of HCC cells and provide a potential therapeutic target for combating HCC.


Assuntos
Calgranulina B/fisiologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/fisiologia , Macrófagos Associados a Tumor/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia
9.
Mol Med Rep ; 22(6): 5293-5303, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33174028

RESUMO

S100 calcium binding protein A8 (S100A8) and A9 (S100A9) belong to the S100 family of calcium­binding proteins and have important roles in inflammation. They increase endothelial cell proliferation, thereby affecting inflammation, angiogenesis and tumorigenesis. However, the mechanism of action of S100A8/9 in endothelial cells needs further study. Therefore, the present study sought to investigate the effects of S100A8/9 on the proliferation and angiogenesis of human umbilical vein endothelial cells (HUVECs) and their mechanism of action. The viability of HUVECs was determined through a Cell Counting Kit­8 assay. The effect of S100A8/9 on the proliferation of HUVECs was detected by flow cytometry. Migration was evaluated by a Transwell migration assay. Apoptosis was evaluated by Annexin V­FITC and PI staining via flow cytometry. Western blot analysis and reverse transcription­quantitative polymerase chain reaction assays were performed to evaluate the activation of the phosphatidylinositol 3­phosphate kinase (PI3K)/Akt/mTOR pathway and mTOR complex 2 (mTORC2). We previously confirmed that S100A8/9 were consistently overexpressed at 1 and 7 days post­surgery in a rabbit vein graft model, which is the period when apoptosis changes to proliferation in neointimal hyperplasia. In the present study, proliferation, viability and migration were increased after treating HUVECs with S100A8/9. S100A8/9 stimulated the PI3K/Akt/mTOR pathway and mTORC2, which was significantly suppressed by a receptor for advanced glycation end products (RAGE)­blocking antibody. Furthermore, depleting expression of RAGE or mTORC2 protein components (rapamycin­insensitive companion of mTOR) by small interfering RNA was found to reduce the cell viability, migration and angiogenesis of S100A8/9­treated HUVECs. The development of neointimal hyperplasia is a complex process initiated by damage to endothelial cells. In conclusion, S100A8/9 has an important role in intimal hyperplasia by promoting cell growth and angiogenesis via RAGE signaling and activation of mTORC2.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Células Endoteliais/metabolismo , Apoptose , Calgranulina A/genética , Calgranulina B/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
10.
Gene ; 761: 145039, 2020 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-32777527

RESUMO

The wood frog (Rana sylvatica) can tolerate full body freezing in winter. As a protective response, wood frogs dehydrate their cells and accumulate large quantities of glucose as an intracellular cryoprotectant. Freezing causes ischemia since blood delivery to organs is interrupted. Fascinatingly, wood frogs can tolerate dehydration, extreme hyperglycemia, and anoxia independently of freezing. In response to low oxygen levels, wood frogs strategically reduce their metabolic rates and allocate the finite amount of intracellular fuel available to pro-survival processes while reducing or interrupting all others. In this study, the involvement of advanced glycation end products (AGEs) and the high mobility group box 1 (HMGB1) protein in activating RAGE (AGE receptor) were investigated. The results show that freezing, anoxia and dehydration induced the expression of total HMGB1 and its acetylation in the heart. RAGE levels were induced in response to all stress conditions, which resulted in differential regulation of the ETS1 transcription factor. While the nuclear localization of total ETS1 was not affected, the DNA binding activity of total and its active form increased in response to freezing and dehydration but not in response to anoxia. Current results indicate that ETS1 acts as a transcriptional activator for peroxiredoxin 1 in response to freezing but acts as a transcriptional repressor of several nuclear-encoded mitochondrial genes in response to all stresses. Altogether, current results show that the HMGB1/RAGE axis may activate ETS1 and that this activation could result in both transcriptional activation and/or repression in a stress-dependent manner.


Assuntos
Mitocôndrias/metabolismo , Miocárdio/metabolismo , Ranidae/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Animais , Desidratação/metabolismo , Congelamento , Glucose/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Produtos Finais de Glicação Avançada/fisiologia , Proteína HMGB1/metabolismo , Proteína HMGB1/fisiologia , Coração/fisiologia , Hipóxia/metabolismo , Oxigênio/metabolismo , Ranidae/genética , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Estresse Fisiológico/fisiologia
11.
Exp Parasitol ; 216: 107946, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32622941

RESUMO

This study was aimed at investigating the involvement of Receptor for Advanced Glycation End Products (RAGE) during malaria infection and the effects of modulating RAGE on the inflammatory cytokines release and histopathological conditions of affected organs in malarial animal model. Plasmodium berghei (P. berghei) ANKA-infected ICR mice were treated with mRAGE/pAb and rmRAGE/Fc Chimera drugs from day 1 to day 4 post infection. Survival and parasitaemia levels were monitored daily. On day 5 post infection, mice were sacrificed, blood were drawn for cytokines analysis and major organs including kidney, spleen, liver, brain and lungs were extracted for histopathological analysis. RAGE levels were increased systemically during malaria infection. Positive correlation between RAGE plasma concentration and parasitaemia development was observed. Treatment with RAGE related drugs did not improve survival of malaria-infected mice. However, significant reduction on the parasitaemia levels were recorded. On the other hand, inhibition and neutralization of RAGE production during the infection significantly increased the plasma levels of interleukin (IL-4, IL-17A, IL-10 and IL-2) and reduced interferon (IFN)-γ secretion. Histopathological analysis revealed that all treated malarial mice showed a better outcome in histological assessment of affected organs (brain, liver, spleen, lungs and kidney). RAGE is involved in malaria pathogenesis and targeting RAGE could be beneficial in malaria infected host in which RAGE inhibition or neutralization increased the release of anti-inflammatory cytokines (IL-10 and IL-4) and reduce pro-inflammatory cytokine (IFNγ) which may help alleviate tissue injury and improve histopathological conditions of affected organs during the infection.


Assuntos
Citocinas/metabolismo , Malária/imunologia , Malária/patologia , Plasmodium berghei/imunologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Animais , Encéfalo/parasitologia , Encéfalo/patologia , Citocinas/sangue , Rim/parasitologia , Rim/patologia , Modelos Lineares , Fígado/parasitologia , Fígado/patologia , Pulmão/parasitologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Parasitemia/imunologia , Distribuição Aleatória , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/sangue , Receptor para Produtos Finais de Glicação Avançada/imunologia , Baço/parasitologia , Baço/patologia
13.
Arterioscler Thromb Vasc Biol ; 40(8): 1838-1853, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32460581

RESUMO

OBJECTIVE: Vascular calcification is a cardiovascular risk factor and accelerated in diabetes mellitus. Previous work has established a role for calcification-prone extracellular vesicles in promoting vascular calcification. However, the mechanisms by which diabetes mellitus provokes cardiovascular events remain incompletely understood. Our goal was to identify that increased S100A9 promotes the release of calcification-prone extracellular vesicles from human macrophages in diabetes mellitus. Approach and Results: Human primary macrophages exposed to high glucose (25 mmol/L) increased S100A9 secretion and the expression of receptor for advanced glycation end products (RAGE) protein. Recombinant S100A9 induced the expression of proinflammatory and osteogenic factors, as well as the number of extracellular vesicles with high calcific potential (alkaline phosphatase activity, P<0.001) in macrophages. Treatment with a RAGE antagonist or silencing with S100A9 siRNA in macrophages abolished these responses, suggesting that stimulation of the S100A9-RAGE axis by hyperglycemia favors a procalcific environment. We further showed that an imbalance between Nrf-2 (nuclear factor 2 erythroid related factor 2) and NF-κB (nuclear factor-κB) pathways contributes to macrophage activation and promotes a procalcific environment. In addition, streptozotocin-induced diabetic Apoe-/-S100a9-/- mice and mice treated with S100a9 siRNA encapsulated in macrophage-targeted lipid nanoparticles showed decreased inflammation and microcalcification in atherosclerotic plaques, as gauged by molecular imaging and comprehensive histological analysis. In human carotid plaques, comparative proteomics in patients with diabetes mellitus and histological analysis showed that the S100A9-RAGE axis associates with osteogenic activity and the formation of microcalcification. CONCLUSIONS: Under hyperglycemic conditions, macrophages release calcific extracellular vesicles through mechanisms involving the S100A9-RAGE axis, thus contributing to the formation of microcalcification within atherosclerotic plaques.


Assuntos
Calgranulina B/fisiologia , Complicações do Diabetes/etiologia , Vesículas Extracelulares/fisiologia , Macrófagos/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Calcificação Vascular/etiologia , Animais , Diabetes Mellitus Experimental/complicações , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Placa Aterosclerótica/etiologia
14.
Biomed Pharmacother ; 127: 110131, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32325348

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disease. AD has become an important social health problem but there are few therapeutic drugs. Many researchers devote to the development of drugs for the treatment of AD. GJ-4 is crocin enrichments from Gardenia jasminoides J. Ellis, and our previous studies have shown GJ-4 had potent neuroprotective effects on several AD animal models. However, the underlying mechanisms have not been fully elucidated. The aim of the present study was to explore the mechanism of GJ-4 on a Aß25-35-intoxicated mouse model. The results demonstrated that GJ-4 treatment significantly improved spatial learning and memory abilities of the AD mice challenged by Aß25-35. Mechanistic study indicated that GJ-4 could alleviate endothelial dysfunction, as GJ-4 markedly reduced endothelial cell edema, as well as improved tight junction structures by up-regulating Zonula occludens-1 (ZO-1), Claudin-5 and Occludin expressions. Moreover, GJ-4 markedly reduced receptor for advanced glycation end products (RAGE) expression and increased low-density lipoprotein receptor-related protein-1 (LRP-1) expression, suggesting endothelial transduction and clearance of toxic species capabilities improved by GJ-4 treatment. The results also indicated that GJ-4 significantly decreased IL-6 and IL-1ß mRNA expressions, as well as intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) expressions, implying the inhibition of glial activation and vascular inflammation by GJ-4 treatment. Furthermore, GJ-4 treatment inhibited glial activation mediated neuroinflammation through inhibiting high-mobility group box protein 1(HMGB-1)/RAGE/NF-κB signaling pathway, which might confer to the neuroprotection. In conclusion, our present study proved GJ-4 could protect the neurovascular unit (NVU), through attenuating endothelial cell damage, enhancing tight junction function, inhibiting of glial activation and protecting of neurons. This study provided evidence that the beneficial effects of GJ-4 on AD might be owing to its protection on NVU.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/toxicidade , Células Endoteliais/efeitos dos fármacos , Gardenia/química , Memória/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/toxicidade , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fármacos Neuroprotetores/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Junções Íntimas/química , Junções Íntimas/efeitos dos fármacos
15.
J Cachexia Sarcopenia Muscle ; 11(4): 929-946, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32159297

RESUMO

BACKGROUND: Cachexia, a multifactorial syndrome affecting more than 50% of patients with advanced cancer and responsible for ~20% of cancer-associated deaths, is still a poorly understood process without a standard cure available. Skeletal muscle atrophy caused by systemic inflammation is a major clinical feature of cachexia, leading to weight loss, dampening patients' quality of life, and reducing patients' response to anticancer therapy. RAGE (receptor for advanced glycation end-products) is a multiligand receptor of the immunoglobulin superfamily and a mediator of muscle regeneration, inflammation, and cancer. METHODS: By using murine models consisting in the injection of colon 26 murine adenocarcinoma (C26-ADK) or Lewis lung carcinoma (LLC) cells in BALB/c and C57BL/6 or Ager-/- (RAGE-null) mice, respectively, we investigated the involvement of RAGE signalling in the main features of cancer cachexia, including the inflammatory state. In vitro experiments were performed using myotubes derived from C2C12 myoblasts or primary myoblasts isolated from C57BL/6 wild type and Ager-/- mice treated with the RAGE ligand, S100B (S100 calcium-binding protein B), TNF (tumor necrosis factor)α±IFN (interferon) γ, and tumour cell- or masses-conditioned media to analyse hallmarks of muscle atrophy. Finally, muscles of wild type and Ager-/- mice were injected with TNFα/IFNγ or S100B in a tumour-free environment. RESULTS: We demonstrate that RAGE is determinant to activate signalling pathways leading to muscle protein degradation in the presence of proinflammatory cytokines and/or tumour-derived cachexia-inducing factors. We identify the RAGE ligand, S100B, as a novel factor able to induce muscle atrophy per se via a p38 MAPK (p38 mitogen-activated protein kinase)/myogenin axis and STAT3 (signal transducer and activator of transcription 3)-dependent MyoD (myoblast determination protein 1) degradation. Lastly, we found that in cancer conditions, an increase in serum levels of tumour-derived S100B and HMGB1 (high mobility group box 1) occurs leading to chronic activation/overexpression of RAGE, which induces hallmarks of cancer cachexia (i.e. muscle wasting, systemic inflammation, and release of tumour-derived pro-cachectic factors). Absence of RAGE in mice translates into reduced serum levels of cachexia-inducing factors, delayed loss of muscle mass and strength, reduced tumour progression, and increased survival. CONCLUSIONS: RAGE is a molecular determinant in inducing the hallmarks of cancer cachexia, and molecular targeting of RAGE might represent a therapeutic strategy to prevent or counteract the cachectic syndrome.


Assuntos
Caquexia/prevenção & controle , Neoplasias/complicações , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Animais , Humanos , Camundongos , Neoplasias/fisiopatologia
16.
Invest Ophthalmol Vis Sci ; 61(3): 14, 2020 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-32176265

RESUMO

Purpose: We used a human corneal epithelial cell (HCE) line to determine the involvement of the advanced glycation end products (AGEs) / receptor for AGEs (RAGE) couple in corneal epithelium wound healing. Methods: After wounding, HCE cells were exposed to two major RAGE ligands (HMGB1 and AGEs), and wound healing was evaluated using the in vitro scratch assay. Following wound healing, the HCE cells were used to study the influence of the RAGE ligands on HCE proliferation, invasion, and migration. Activation of the nuclear factor (NF)-κB signaling pathway by the AGEs/RAGE couple was tested using a luciferase reporter assay. Functional transcriptional regulation by this pathway was confirmed by quantification of expression of the connexin 43 target gene. For each experiment, specific RAGE involvement was confirmed by small interfering RNA treatments. Results: AGEs treatment at a dose of 100 µg/mL significantly improved the wound healing process in a RAGE-dependent manner by promoting cell migration, whereas HMGB1 had no effect. No significant influence of the AGEs/RAGE couple was observed on cell proliferation and invasion. However, this treatment induced an early activation of the NF-κB pathway and positively regulated the expression of the target gene, connexin 43, at both the mRNA and protein levels. Conclusions: Our results demonstrate that the RAGE pathway is activated by AGEs treatment and is involved in the promotion of corneal epithelial wound healing. This positive action is observed only during the early stages of wound healing, as illustrated by the quick activation of the NF-κB pathway and induction of connexin 43 expression.


Assuntos
Lesões da Córnea/fisiopatologia , Epitélio Corneano/efeitos dos fármacos , Produtos Finais de Glicação Avançada/farmacologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Cicatrização/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Conexina 43/genética , Conexina 43/metabolismo , Lesões da Córnea/patologia , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Epitélio Corneano/citologia , Epitélio Corneano/lesões , Epitélio Corneano/fisiologia , Produtos Finais de Glicação Avançada/administração & dosagem , Produtos Finais de Glicação Avançada/fisiologia , Proteína HMGB1/administração & dosagem , Proteína HMGB1/farmacologia , Humanos , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Receptor para Produtos Finais de Glicação Avançada/genética , Transdução de Sinais/fisiologia , Cicatrização/fisiologia
17.
Curr Drug Targets ; 21(2): 148-166, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31385768

RESUMO

Alzheimer's disease (AD) is one of the most common forms of dementia and has been a global concern for several years. Due to the multi-factorial nature of the disease, AD has become irreversible, fatal and imposes a tremendous socio-economic burden. Even though experimental medicines suggested moderate benefits, AD still lacks an effective treatment strategy for the management of symptoms or cure. Among the various hypotheses that describe development and progression of AD, the amyloid hypothesis has been a long-term adherent to the AD due to the involvement of various forms of Amyloid beta (Aß) peptides in the impairment of neuronal and cognitive functions. Hence, majority of the drug discovery approaches in the past have focused on the prevention of the accumulation of Aß peptides. Currently, there are several agents in the phase III clinical trials that target Aß or the various macromolecules triggering Aß deposition. In this review, we present the state of the art knowledge on the functional aspects of the key players involved in the amyloid hypothesis. Furthermore, we also discuss anti-amyloid agents present in the Phase III clinical trials.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Inibidores Enzimáticos/uso terapêutico , Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Anticorpos Monoclonais Humanizados/uso terapêutico , Encéfalo/metabolismo , Ensaios Clínicos como Assunto , Progressão da Doença , Humanos , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/fisiologia
18.
Acta Med Okayama ; 73(5): 383-386, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31649363

RESUMO

High mobility group box-1 (HMGB1) is a non-histone, DNA-binding nuclear protein belonging to the family of damage-associated molecular patterns (DAMPs). HMGB1 has been reported to play an important role during epileptogenesis although the mechanisms of its actions are still not clear. Many hypotheses have been suggested especially about the relationship between HMGB1 and inflammation responses and blood-brain barrier disruption during epileptogenesis. In this review, we will mainly discuss the role of HMGB1 in epileptogenesis.


Assuntos
Epilepsia/etiologia , Proteína HMGB1/fisiologia , Barreira Hematoencefálica , Humanos , Inflamação/etiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Receptor 4 Toll-Like/fisiologia
19.
Brain Res ; 1723: 146379, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31415766

RESUMO

Abnormal accumulation of amyloid-ß (Aß) peptide defines progression of Alzheimer's disease (AD) pathology in brain. Here, we investigated expressive changes of two main Aß transport receptors low-density lipoprotein receptor related protein-1 (LRP1) and receptor for advanced glycation end products (RAGE) in a novel AD mice (APP23) with chronic cerebral hypoperfusion (CCH) model, moreover, examined a protective effect of a free radical scavenger edaravone (Eda). In contrast to wild type (WT) and APP23 mice, CCH strongly accelerated abnormal Aß40 depositions and cerebral amyloid angiopathy (CAA) pathology, increased both LRP1 and RAGE expressions in brain parenchyma, while a decrease of LRP1 and an increase of RAGE were observed in vascular endothelial cells at age 12 months (M) of AD mice. Furthermore, CCH strongly increased expressions of two hypoxia-related proteins hypoxia inducible factor-1α (HIF-1α) and heme oxygenase-1 (HO-1), two oxidative-related proteins 4-hydroxy-2-nonenal (4-HNE) and 8-hydroxy-2'-deoxyguanosine (8-OHdG), and decreased both two vital nutrient transporter proteins major facilitator super family domain containing 2a (Mfsd2a) and glucose transporter 1 (Glut1) expressions. Such the above abnormal pathological changes were significantly ameliorated by edaravone treatment. The present study demonstrated that CCH strongly enhanced primary AD pathology causing double imbalances of Aß efflux and influx transport related proteins in the cortical blood vessels in AD mice, and that such a neuropathologic abnormality was greatly ameliorated by Eda.


Assuntos
Doença de Alzheimer/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Isquemia Encefálica/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Edaravone/farmacologia , Células Endoteliais/metabolismo , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fármacos Neuroprotetores/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Receptores de LDL/metabolismo
20.
Inflamm Res ; 68(8): 639-642, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31115587

RESUMO

OBJECTIVE: The hypoxic milieu at tumor microenvironment is able to drive the behavior of infiltrating tumor cells. Considering that hypoxia-mediated HMGB1 release is known to promote tumor growth, as well to enhance the pro-tumoral profile of M2 macrophages by a RAGE-dependent mechanism, it is tempting to evaluate the potential contribution of HMGB1 under hypoxia to restrain M2 macrophages mobility. METHODS: CCR-2 expression was evaluated in M2 polarized macrophages by western blotting and immunocytochemistry. The secreted levels of CCL-2 and the migration capability were evaluated using an ELISA and a chemotaxis assay, respectively. RESULTS: HMGB1, under hypoxic conditions, markedly reduce both the production of CCL-2 and the expression of its receptor CCR-2; and reduced the migration capacity of M2 macrophages. CONCLUSIONS: These results provided new insights into the mechanisms that regulate M2 macrophages mobility at the tumor microenvironment.


Assuntos
Proteína HMGB1/fisiologia , Macrófagos/fisiologia , Receptores CCR2/fisiologia , Hipóxia Tumoral/fisiologia , Movimento Celular , Quimiocina CCL2/fisiologia , Humanos , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Células THP-1 , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA