1
|
Cai M, Zhang X, Gao X, Huo Q, Sun Y, Dai X. Chitooligosaccharide ameliorates cognitive deficits and neuroinflammation in APP/PS1 mice associated with the regulation of Nrf2/NF-κB axis. Int J Biol Macromol 2025; 303:140683. [PMID: 39914538 DOI: 10.1016/j.ijbiomac.2025.140683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025]
Abstract
Mounting evidence suggests that neuroinflammation is involved in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD). Amyloid β peptide (Aβ) could recruit and activate microglia, leading to the generation of pro-inflammatory factors, and ultimately neuroinflammation. Chitooligosaccharide (COS) is widely recognized as anti-inflammation bioactive substance, though whether it exerts beneficial effect on AD is unclear. In this study, we explored the effect of COS on AD prevention and treatment. We found that COS ameliorated cognitive deficiency, increased the expression of Nrf2 but decreased Aβ levels and the activation of NF-κB in APP/PS1 mice. In vitro, COS decreased the secretions of IL-6, IL-1β and TNF-α in Aβ25-35 + lipopolysaccharides (LPS) -exposed BV2 microglia. Meanwhile, COS down-regulated the expressions of iNOS, COX-2, NLRP3, caspase 1 and the nuclear translocation of NF-κB p65, while upregulated the expressions of Nrf2 and HO-1. Further, COS improved the viability of SK-N-SH cells that exposed to Aβ25-35 + LPS-stimulated microglial conditioned media, and the repressive effect of COS on NLRP3, iNOS, and phospho-NF-κB p65 expressions were markedly compromised upon Nrf2-siRNA transfection. Collectively, COS improved cognitive decline and suppressed neuroinflammation via the Nrf2/NF-κB signaling axis, suggesting COS might be a promising candidate in down-regulating inflammatory responses during AD progression.
Collapse
Affiliation(s)
- Mingyang Cai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China; Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Xiaoxia Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Xiaohan Gao
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Qing Huo
- Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China; Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China; Department of Food Science and Biomedicine, College of Biochemical Engineering, Beijing Union University, Beijing 100023, China.
| |
Collapse
|
2
|
Marasinghe CK, Je JY. Ark Shell-Derived Peptides AWLNH (P3) and PHDL (P4) Mitigate Foam Cell Formation by Modulating Cholesterol Metabolism and HO-1/Nrf2-Mediated Oxidative Stress in Atherosclerosis. Mar Drugs 2025; 23:111. [PMID: 40137297 PMCID: PMC11944032 DOI: 10.3390/md23030111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Atherosclerosis, a leading contributor to cardiovascular diseases (CVDs), is characterized by foam cell formation driven by excessive lipid accumulation in macrophages and vascular smooth muscle cells. This study elucidates the anti-atherosclerotic potential of AWLNH (P3) and PHDL (P4) peptides by assessing their effects on foam cell formation, lipid metabolism, and oxidative stress regulation. P3 and P4 effectively suppressed intracellular lipid accumulation in RAW264.7 macrophages and human aortic smooth muscle cells (hASMCs), thereby mitigating foam cell formation. Mechanistically, both peptides modulated cholesterol homeostasis by downregulating cholesterol influx mediators, cluster of differentiation 36 (CD36), and class A1 scavenger receptor (SR-A1), while upregulating cholesterol efflux transporters ATP-binding cassette subfamily A member 1 (ABCA1) and ATP-binding cassette subfamily G member 1 (ABCG1). The activation of peroxisome proliferator-activated receptor-gamma (PPAR-γ) and liver X receptor-alpha (LXR-α) further substantiated their role in promoting cholesterol efflux and restoring lipid homeostasis. Additionally, P3 and P4 peptides exhibited potent antioxidative properties by attenuating reactive oxygen species (ROS) generation through activation of the HO-1/Nrf2 signaling axis. HO-1 silencing via siRNA transfection abolished these effects, confirming HO-1-dependent regulation of oxidative stress and lipid metabolism. Collectively, these findings highlight P3 and P4 peptides as promising therapeutic agents for atherosclerosis by concurrently targeting foam cell formation, cholesterol dysregulation, and oxidative stress, warranting further exploration for potential clinical applications.
Collapse
Affiliation(s)
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
3
|
Choi JW, Choi HJ, Na CS, Lee H, Lee BJ, Shin KC, Jeong JB. Anti-Inflammatory Activity of Biotransformed Platycodon grandiflorum Root Extracts Containing 3- O-β-D-Glucopyranosyl Platycosides in LPS-Stimulated Alveolar Macrophages, NR8383 Cells. J Microbiol Biotechnol 2024; 34:2609-2617. [PMID: 39476865 PMCID: PMC11729695 DOI: 10.4014/jmb.2408.08005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 12/31/2024]
Abstract
Acute lung injury (ALI) is a severe inflammatory condition characterized by excessive immune responses and oxidative stress, leading to significant tissue damage. Given the need for novel therapeutic agents, this study aimed to explore the anti-inflammatory activity and mechanisms of biotransformed Platycodon grandiflorum root extracts (BT-PGR), which were enzymatically processed using rapidsase PL Classic from Aspergillus niger. The goal was to assess the potential of BT-PGR as a natural treatment for ALI. BT-PGR effectively inhibited the production of NO, iNOS, IL-1β, IL-6, and TNF-α induced by LPS in NR8383 cells. BT-PGR inhibited the phosphorylation of ERK1/2, p38, JNK and p65 in LPS-stimulated NR8383 cells. In addition, BT-PGR suppressed LPS-mediated activation of NF-κB luciferase activity. BT-PGR increased the levels of HO-1 and the inhibition of HO-1 by ZnPP attenuated BT-PGR-mediated inhibition of NO production. In addition, the inhibition of PI3K by LY294002 blocked the BT-PGR-mediated increase of HO-1 level. BT-PGR increased nuclear Nrf2 level and the knockdown of Nrf2 by siRNA inhibited BT-PGR-mediated increase of HO-1 level. In addition, inhibition of PI3K by LY294002 suppressed the increase of nuclear Nrf2 level. Based on these results, it can be inferred that BT-PGR exhibits anti-inflammatory activity in rat alveolar macrophages, suggesting its potential as a natural candidate for the improvement of ALI.
Collapse
Affiliation(s)
- Jeong Won Choi
- Department of Forest Science, Andong National University, Andong 36729, Republic of Korea
| | - Hyeok Jin Choi
- Department of Forest Science, Andong National University, Andong 36729, Republic of Korea
| | - Chae Sun Na
- Division of Wild Plant and Seeds, Baekdudaegan National Arboretum, Bonghwa 36029, Republic of Korea
| | - Hwan Lee
- Lifeceutical Bio Food Research Center, Seoul 06138, Republic of Korea
| | - Byung Joo Lee
- Lifeceutical Bio Food Research Center, Seoul 06138, Republic of Korea
| | - Kyung-Chul Shin
- Department of Bioscience and Biotechnology, Hankuk University of Foreign Studies, Yongin 17035, Republic of Korea
| | - Jin Boo Jeong
- Department of Forest Science, Andong National University, Andong 36729, Republic of Korea
| |
Collapse
|
4
|
Zhan ZQ, Li JX, Chen YX, Fang JY. The effects of air and transportation noise pollution-related altered blood gene expression, DNA methylation, and protein abundance levels on gastrointestinal diseases risk. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175633. [PMID: 39163931 DOI: 10.1016/j.scitotenv.2024.175633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/03/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
INTRODUCTION Air pollution and transportation noise pollution has been linked to gastrointestinal (GI) diseases, but their relationship remains unclear. METHODS We extracted the significantly modulated genes and CpG sites related to air pollution (PM2.5, PM10, and NOx) and transportation noise pollution (aircraft, railway, and traffic road noise) from previous published studies. Genome-wide methylation analysis and colocalization analysis with these CpG sites and GWAS data of GI diseases were performed to disentangle the relationship between pollution-related blood DNA methylation (DNAm) alterations and GI diseases risk. Summary-based Mendelian randomization (SMR) analysis assessed the impact of pollution-related genes on GI diseases risk across methylation, gene expression, and protein levels. Enrichment analysis investigated the implicated biological pathways and immune cell types. RESULTS DNAm at cg00227781 [CD300A] (modulated by NOx exposure) and cg19215199 [ZMIZ1] (modulated by PM2.5 exposure) was significantly linked to increased noninfective enteritis and colitis risk, while cg08500171 [BAT2] (modulated by NOx exposure) is significantly associated with an increased gastroesophageal reflux disease (GERD) risk. Colocalization analysis provides strong evidence supporting a shared causal variant between these associations. Multi-omics levels SMR analysis revealed that pollution-modulated lower DNAm at 5 specific CpG sites were associated with increased expression of 4 genes (IL21R, EVPL, SYNGR1, and WDR46), subsequently increasing the risk of GERD, ulcerative colitis, and gastric ulcer. 7 circulating proteins coded by pollution-modulated genes were observed to be associated with 6 GI diseases risk. Enrichment analysis implicates immune and inflammatory responses, MAPK signaling, and telomere maintenance in these pollution-induced effects. CONCLUSION We identified potential links between air and transportation noise pollution-related gene methylation, expression, and protein abundance with GI diseases risk, possibly revealing new therapeutic targets.
Collapse
Affiliation(s)
- Zhi-Qing Zhan
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia-Xin Li
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology; Shanghai Institute of Digestive Disease; NHC Key Laboratory of Digestive Diseases; State Key Laboratory for Oncogenes and Related Genes; Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
5
|
Le MPT, Marasinghe CK, Je JY. Chitosan oligosaccharides: A potential therapeutic agent for inhibiting foam cell formation in atherosclerosis. Int J Biol Macromol 2024; 282:137186. [PMID: 39491693 DOI: 10.1016/j.ijbiomac.2024.137186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Foam cell formation is a key hallmark in atherosclerosis and associated cardiovascular diseases (CVDs). The potential anti-atherosclerotic potential of chitosan oligosaccharides (COS) was investigated using oxLDL-treated RAW264.7 murine cells. COS treatment led to a significant inhibition of lipid accumulation, as demonstrated by Oil Red O staining, and reduced levels of total cholesterol, free cholesterol, cholesterol esters, and triglycerides in.oxLDL-treated RAW264.7 cells. COS blocked cholesterol influx through down-regulating class A1 scavenger receptors (SR-A1) and cluster of differentiation 36 (CD36) expression and stimulated cholesterol efflux through up-regulating ABC transporters ABCA-1 and ABCG-1 expressions. Additionally, COS treatment stimulated nuclear signaling pathways involving peroxisome proliferator-activated receptor-γ (PPAR-γ) and liver X receptor α (LXR-α), and also led to the phosphorylation of AMP-activated protein kinase (AMPK). COS further demonstrated anti-inflammatory effects by inhibiting the production of pro-inflammatory cytokines and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in oxLDL-treated RAW264.7 cells, through suppression of NF-κB signaling. Furthermore, COS alleviated oxidative stress induced by oxLDL by activating Nrf2 signaling and enhancing the expression of antioxidant genes, including heme oxygenase-1 (HO-1), superoxide dismutase (SOD), glutathione peroxidase (Gpx), and catalase (CAT). In conclusion, COS can be beneficial in preventing atherosclerosis and related diseases.
Collapse
Affiliation(s)
- My Phuong Thi Le
- Department of Food and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | | | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
6
|
Marasinghe CK, Yoon SD, Je JY. Two peptides LLRLTDL and GYALPCDCL inhibit foam cell formation through activating PPAR-γ/LXR-α signaling pathway in oxLDL-treated RAW264.7 macrophages. Biofactors 2024; 50:1161-1175. [PMID: 38760074 DOI: 10.1002/biof.2075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 03/12/2024] [Indexed: 05/19/2024]
Abstract
Foam cell formation plays a pivotal role in atherosclerosis-associated cardiovascular diseases. Bioactive peptides generated from marine sources have been found to provide multifunctional health advantages. In the present study, we investigated the anti-atherosclerotic effects of LLRLTDL (Bu1) and GYALPCDCL (Bu2) peptides, isolated from ark shell protein hydrolysates by assessing their inhibitory effect on oxidized LDL (oxLDL)-induced foam cell formation. The two peptides showed a promising anti-atherosclerotic effect by inhibiting foam cell formation, which was evidenced by inhibiting lipid accumulation in oxLDL-treated RAW264.7 macrophages and oxLDL-treated primary human aortic smooth muscle cells (HASMC). Two peptides effectively reduced total cholesterol, free cholesterol, cholesterol ester, and triglyceride levels by upregulating cholesterol efflux and downregulating cholesterol influx. Expression of cholesterol influx-related proteins such as SR-A1 and CD36 were reduced, whereas cholesterol efflux-related proteins such as ATP-binding cassette transporter ABCA-1 and ABCG-1 were highly expressed. In addition, Bu1 and Bu2 peptides increased PPAR-γ and LXR-α expression. However, PPAR-γ siRNA transfection reversed the foam cell formation inhibitory activity of Bu1 and Bu2 peptides. Furthermore, the synergistic effect of Bu1 and Bu2 peptides on foam cell formation inhibition was observed with PPAR-γ agonist thiazolidinediones, indicating that PPAR-γ signaling pathway plays a key role in foam cell formation of macrophages. Beyond their impact on foam cell formation, Bu1 and Bu2 peptides demonstrated anti-inflammatory potential by inhibiting the generation of pro-inflammatory cytokines and nitric oxide and NF-κB nuclear activation. Taken together, these results suggest that Bu1 and Bu2 peptides may be useful for atherosclerosis and associated anti-inflammatory therapies.
Collapse
Affiliation(s)
| | - Soon-Do Yoon
- Department of Biomolecular and Chemical Engineering, Chonnam National University, Yeosu, Jeonnam, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
7
|
Marasinghe CK, Je JY. Blue Mussel-Derived Bioactive Peptides PIISVYWK (P1) and FSVVPSPK (P2): Promising Agents for Inhibiting Foam Cell Formation and Inflammation in Cardiovascular Diseases. Mar Drugs 2024; 22:466. [PMID: 39452874 PMCID: PMC11509633 DOI: 10.3390/md22100466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Atherosclerosis is a key etiological event in the development of cardiovascular diseases (CVDs), strongly linked to the formation of foam cells. This study explored the effects of two blue mussel-derived bioactive peptides (BAPs), PIISVYWK (P1) and FSVVPSPK (P2), on inhibiting foam cell formation and mitigating inflammation in oxLDL-treated RAW264.7 macrophages. Both peptides significantly suppressed intracellular lipid accumulation and cholesterol levels while promoting cholesterol efflux by downregulating cluster of differentiation 36 (CD36) and class A1 scavenger receptors (SR-A1) and upregulating ATP binding cassette subfamily A member 1 (ABCA-1) and ATP binding cassette subfamily G member 1 (ABCG-1) expressions. The increased expression of peroxisome proliferator-activated receptor-gamma (PPAR-γ) and liver X receptor-alpha (LXR-α) further validated their role in enhancing cholesterol efflux. Additionally, P1 and P2 inhibited foam cell formation in oxLDL-treated human aortic smooth muscle cells and exerted anti-inflammatory effects by reducing pro-inflammatory cytokines, nitric oxide (NO), prostaglandin E2 (PGE2), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2), primarily through inhibiting NF-κB activation. Furthermore, P1 and P2 alleviated oxidative stress by activating the Nrf2/HO-1 pathway. Our findings demonstrate that P1 and P2 have significant potential in reducing foam cell formation and inflammation, both critical factors in atherosclerosis development. These peptides may serve as promising therapeutic agents for the prevention and treatment of CVDs associated with oxidative stress and inflammation.
Collapse
Affiliation(s)
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
8
|
Sui Y, Xu D. Isolation and identification of anti-inflammatory and analgesic polysaccharides from Coix seed ( Coix lacryma-jobi L.var. Ma-yuen (Roman.) Stapf). Nat Prod Res 2024; 38:2165-2174. [PMID: 36584288 DOI: 10.1080/14786419.2022.2162896] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/12/2022] [Accepted: 12/17/2022] [Indexed: 12/31/2022]
Abstract
Coix seed is a nutrient-rich food and traditional Chinese medicine with anti-inflammatory and analgesic properties. Polysaccharides from Coix seed have been rarely investigated for structure and activities. In this study, the analgesic and anti-inflammatory effects were investigated in vivo and in vitro. The results showed that Coix seed had a significant influence on reducing the number of writhing, increasing the pain threshold and alleviating the swelling degree caused by acute inflammation. Column chromatography was used to obtain two active compounds of Coix seed. Compound 1 was (1→6)-α-glucan with a molecular weight of 6.81 × 105 Da. The chemical connection of compound 2 was as follows: α-Frup (2→ [1)-α-Glcp (6]5→1)-α-Glcp (4→1)-α-Glcp, which was isolated in Coix seed for the first time. LPS-induced inflammation in RAW264.7 cells was well inhibited by compounds. These findings offered a preliminary investigation into the analgesic and anti-inflammatory properties of Coix seed, which may be helpful for application.
Collapse
Affiliation(s)
- Yingling Sui
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Wuxi, China
| | - Deping Xu
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Wuxi, China
| |
Collapse
|
9
|
Rashwan AG, Assar DH, Salah AS, Liu X, Al-Hawary II, Abu-Alghayth MH, Salem SMR, Khalil K, Hanafy NAN, Abdelatty A, Sun L, Elbialy ZI. Dietary Chitosan Attenuates High-Fat Diet-Induced Oxidative Stress, Apoptosis, and Inflammation in Nile Tilapia ( Oreochromis niloticus) through Regulation of Nrf2/Kaep1 and Bcl-2/Bax Pathways. BIOLOGY 2024; 13:486. [PMID: 39056682 PMCID: PMC11273726 DOI: 10.3390/biology13070486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024]
Abstract
Fatty liver injury is a prevalent condition in most farmed fish, yet the molecular mechanisms underpinning this pathology remain largely elusive. A comprehensive feeding trial spanning eight weeks was conducted to discern the potential of dietary chitosan in mitigating the deleterious effects of a high-fat diet (HFD) while concurrently exploring the underlying mechanism. Growth performance, haemato-biochemical capacity, antioxidant capacity, apoptotic/anti-apoptotic gene expression, inflammatory gene expression, and histopathological changes in the liver, kidney, and intestine were meticulously assessed in Nile tilapia. Six experimental diets were formulated with varying concentrations of chitosan. The first three groups were administered a diet comprising 6% fat with chitosan concentrations of 0%, 5%, and 10% and were designated as F6Ch0, F6Ch5, and F6Ch10, respectively. Conversely, the fourth, fifth, and sixth groups were fed a diet containing 12% fat with chitosan concentrations of 0%, 5%, and 10%, respectively, for 60 days and were termed F12Ch0, F12Ch5, and F12Ch10. The results showed that fish fed an HFD demonstrated enhanced growth rates and a significant accumulation of fat in the perivisceral tissue, accompanied by markedly elevated serum hepatic injury biomarkers and serum lipid levels, along with upregulation of pro-apoptotic and inflammatory markers. In stark contrast, the expression levels of nrf2, sod, gpx, and bcl-2 were notably decreased when compared with the control normal fat group. These observations were accompanied by marked diffuse hepatic steatosis, diffuse tubular damage, and shortened intestinal villi. Intriguingly, chitosan supplementation effectively mitigated the aforementioned findings and alleviated intestinal injury by upregulating the expression of tight junction-related genes. It could be concluded that dietary chitosan alleviates the adverse impacts of an HFD on the liver, kidney, and intestine by modulating the impaired antioxidant defense system, inflammation, and apoptosis through the variation in nrf2 and cox2 signaling pathways.
Collapse
Affiliation(s)
- Aya G. Rashwan
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (A.G.R.); (I.I.A.-H.)
| | - Doaa H. Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Abdallah S. Salah
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK
| | - Xiaolu Liu
- Single-Cell Center, Shandong Key Laboratory of Energy Genetics and Shandong Energy Institute, Qingdao Institute of Bioenergy and Bioprocess Technology, CAS Key Laboratory of Biofuels, Chinese Academy of Sciences, Qingdao 266101, China;
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Ibrahim I. Al-Hawary
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (A.G.R.); (I.I.A.-H.)
| | - Mohammed H. Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia;
| | - Shimaa M. R. Salem
- Department of Animal Nutrition and Nutritional Deficiency Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 33516, Egypt;
| | - Karim Khalil
- Department of Veterinary Medicine, College of Applied & Health Sciences, A’Sharqiyah University, P.O. Box 42, Ibra 400, Oman;
| | - Nemany A. N. Hanafy
- Group of Molecular Cell Biology and Bionanotechnology, Nanomedicine Department, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Alaa Abdelatty
- Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Luyang Sun
- Single-Cell Center, Shandong Key Laboratory of Energy Genetics and Shandong Energy Institute, Qingdao Institute of Bioenergy and Bioprocess Technology, CAS Key Laboratory of Biofuels, Chinese Academy of Sciences, Qingdao 266101, China;
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zizy I. Elbialy
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (A.G.R.); (I.I.A.-H.)
| |
Collapse
|
10
|
Liu X, Zang L, Yu J, Yu J, Wang S, Zhou L, Song H, Ma Y, Niu X, Li W. Anti-inflammatory effect of proanthocyanidins from blueberry through NF-κβ/NLRP3 signaling pathway in vivo and in vitro. Immunopharmacol Immunotoxicol 2024:1-11. [PMID: 38772618 DOI: 10.1080/08923973.2024.2358770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/18/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Systemic inflammatory response syndrome (SIRS) is an uncontrolled systemic inflammatory response. Proanthocyanidins (PC) is a general term of polyphenol compounds widely existed in blueberry fruits and can treat inflammation-related diseases. This study aimed to explore the regulatory effect of PC on lipopolysaccharide (LPS)-induced systemic inflammation and its potential mechanism, providing effective strategies for the further development of PC. METHODS Here, RAW264.7 macrophages were stimulated with LPS to establish an inflammation model in vitro, while endotoxin shock mouse models were constructed by LPS in vivo. The function of PC was investigated by MTT, ELISA kits, H&E staining, immunohistochemistry, and Western blot analysis. RESULTS Functionally, PC could demonstrate the potential to mitigate mortality in mice with endotoxin shock, as well as attenuated the levels of inflammatory cytokines (IL-6, TNF-α) and biochemical indicators (AST, ALT, CRE and BUN). Moreover, it had a significant protective effect on lung and kidney tissues damage. Mechanistically, PC exerted anti-inflammatory effects by inhibiting the activation of the NF-κB/NLRP3 signaling pathway. CONCLUSION PC might have the potential ability of anti-inflammatory effects via modulation of the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Lulu Zang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yajing Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
11
|
Marasinghe CK, Jung WK, Je JY. Phloroglucinol possesses anti-inflammatory activities by regulating AMPK/Nrf2/HO-1 signaling pathway in LPS-stimulated RAW264.7 murine macrophages. Immunopharmacol Immunotoxicol 2023; 45:571-580. [PMID: 36988555 DOI: 10.1080/08923973.2023.2196602] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/24/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Inflammation is closely related to the pathogenesis of chronic illnesses. Secondary metabolites of marine seaweeds are recognized as reliable sources of bioactive compounds due to their health benefits besides their nutritional value. The objective of this study was to determine the potential anti-inflammatory effect of phloroglucinol (Phl) in RAW264.7 murine macrophages after lipopolysaccharides (LPS) stimulation. METHODS MTT, nitric oxide (NO), and DCFH-DA assays were conducted to determine cell viability, NO production, and reactive oxygen species (ROS) generation respectively. Pro-inflammatory cytokines and prostaglandin E2 (PGE2) levels were measured using ELISA assay kits. Protein expression levels were determined by western blot analysis. RESULTS Phl treatment showed a promising anti-inflammatory effect by reducing NO production, secretion of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6), PGE2 production, protein expression levels of inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2), and ROS generation in LPS-stimulated RAW264.7 murine macrophages. Phl treatment upregulated heme oxygenase-1 (HO-1) expression by inducing nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and activating AMPK. However, Zinc protoporphyrin (ZnPP), an inhibitor of HO-1, partially reversed these effects, including NO production, pro-inflammatory cytokine secretion, iNOS, COX-2 and HO-1 expression, and ROS generation. CONCLUSION Phl has potential anti-inflammatory activities by regulating AMPK/Nrf2/HO-1 pathway in LPS-stimulated RAW264.7 murine macrophages.
Collapse
Affiliation(s)
| | - Won-Kyo Jung
- Major of Biomedical Engineering, Division of Smart Healthcare, Pukyong National University, Busan, Republic of Korea
- Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
12
|
Jang HY, Lee SO. Heme Oxygenase 1-Mediated Anti-Inflammatory Effect of Extract from the Aerial Part of Heracleum moellendorffii Hance. Foods 2023; 12:3309. [PMID: 37685243 PMCID: PMC10486398 DOI: 10.3390/foods12173309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
In this study, the anti-inflammatory effects of a methanolic extract from the aerial part of Heracleum moellendorffii Hance (HmAPE) and its underlying mechanisms were investigated. HmAPE demonstrated a significant reduction in nitric oxide production in lipopolysaccharide (LPS)-treated murine macrophage RAW264.7 cells, and HmAPE decreased the protein and mRNA expression of inducible nitric oxide synthase. Further mechanistic studies on inflammatory signaling pathways revealed that HmAPE-mediated downregulation of inflammatory gene expressions was not associated with mitogen-activated protein kinases or nuclear factor-κB signaling pathways. However, HmAPE treatment activated nuclear factor E2-related factor 2 (Nrf2) and upregulated heme oxygenase-1 (HO-1) expression, which is known to suppress pro-inflammatory cytokine production. Additionally, treatment with a selective HO-1 inhibitor, tin protoporphyrin IX, partially reversed the effects of HmAPE in LPS-treated RAW264.7 cells, indicating that HmAPE inhibited LPS-induced NO production, at least in part, through induction of Nrf2-mediated HO-1 expression. These findings suggest that HmAPE could serve as a potential edible source with anti-inflammatory properties, and further studies are required to ascertain its anti-inflammatory efficacy in vivo.
Collapse
Affiliation(s)
| | - Syng-Ook Lee
- Correspondence: ; Tel.: +82-53-580-5570; Fax: +82-53-580-5372
| |
Collapse
|
13
|
OxLDL-Induced Foam Cell Formation Inhibitory Activity of Pepsin Hydrolysate of Ark Shell (Scapharca subcrenata (Lischke, 1869)) in RAW264.7 Macrophages. J Food Biochem 2023. [DOI: 10.1155/2023/6905673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Inhibitory effect of ark shell (Scapharca subcrenata (Lischke, 1869)) proteolytic hydrolysates (ASHs) on oxidized low-density lipoprotein (oxLDL)-induced macrophage foam cell formation was investigated. Two types of ASHs were prepared by Alcalase® and pepsin, ASAH (ark shell-Alcalase® hydrolysates), and ASPH (ark shell-pepsin hydrolysate). Oil Red O staining results showed that ASPH suppressed foam cell formation and lipid accumulation more than ASAH in oxLDL-induced foam cell formation of RAW264.7 macrophages. ASPH reduced the levels of total cholesterol, cholesterol ester, and free cholesterol in oxLDL-treated RAW264.7 macrophages. It was found that ASPH increased cholesterol efflux and decreased cholesterol influx rate. In this regard, protein expressions of CD36 and scavenger receptor class A1 (SR-A1) for cholesterol influx and ATP-binding cassette transporter A1 and G1 (ABCA1 and ABCG1) for cholesterol efflux were investigated. ASPH treatment resulted in an increase of ABCA1 and ABCG1 expression but downregulated CD36 and SR-A1 expression. Furthermore, ASPH suppressed production of proinflammatory cytokines, including tumor necrosis factor-α and interleukin-6 and -1β, through regulating nuclear factor-kappa B (NF-κB) in oxLDL-induced foam cell formation of RAW264.7 macrophages. Taken together, our data indicate that ASPH might be a useful ingredient in functional foods for ameliorating atherosclerosis by preventing foam cell formation.
Collapse
|
14
|
Marasinghe CK, Jung WK, Je JY. Anti-inflammatory action of ark shell (Scapharca subcrenata) protein hydrolysate in LPS-stimulated RAW264.7 murine macrophages. J Food Biochem 2022; 46:e14493. [PMID: 36309949 DOI: 10.1111/jfbc.14493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 01/14/2023]
Abstract
Potential anti-inflammatory effects of ark shell (Scapharca subcrenata) protein hydrolysates were investigated. Ark shell protein hydrolysates were prepared using Alcalase® and pepsin and were designated ASAH and ASPH, respectively. The nitric oxide (NO) inhibitory activity of ASAH and ASPH was determined in lipopolysaccharides (LPS)-stimulated RAW264.7 murine macrophages, and the results showed that ASAH inhibited better NO inhibitory activity than ASPH. ASAH suppressed inflammatory mediator, a prostaglandin E2, secretion of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6), and production of reactive oxygen species (ROS) dose dependently. It inhibited the protein expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) and simulated heme oxygenase-1 (HO-1) protein expression. However, the pharmacological approach revealed that pretreatment with zinc protoporphyrin ІX (ZnPP), an inhibitor of HO-1, reversed the anti-inflammatory effect of ASAH. Moreover, ASAH upregulated phosphorylation of mitogen-activated protein kinases (MAPKs) including ERK1/2, JNK1/2, and p38 MAPK. To find out the role of MAPKs phosphorylation, MAPKs inhibitors were used, and the results showed that ASAH-mediated HO-1 protein expression and Nrf2 nuclear translocation were abolished. Taken all together, this study revealed that ASAH has a potential anti-inflammatory activity through regulation of the MAPK-dependent HO-1/Nrf2 pathway. PRACTICAL APPLICATIONS: Food-derived marine bioactive peptides, due to their pivotal role in biological activities, are gaining much attention recently. However, the anti-inflammatory activities of ark shell protein hydrolysates still remain to be investigated. This study investigated that ASAH shows potential anti-inflammatory activities through regulation of the MAPK-dependent HO-1/Nrf2 pathway in RAW264.7 murine macrophages. These findings indicated that ASAH may be used as a dietary supplement, functional food, and medicinal drug for the management of inflammation and inflammation-associated diseases.
Collapse
Affiliation(s)
| | - Won-Kyo Jung
- Major of Biomedical Engineering, Division of Smart Healthcare, Pukyong National University, Busan, Republic of Korea.,Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
15
|
Bangar SP, Dunno K, Dhull SB, Kumar Siroha A, Changan S, Maqsood S, Rusu AV. Avocado seed discoveries: Chemical composition, biological properties, and industrial food applications. Food Chem X 2022; 16:100507. [PMID: 36573158 PMCID: PMC9789361 DOI: 10.1016/j.fochx.2022.100507] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
The processing industry discards avocado seeds, which increases production and ultimately pollutes the environment. It would be advantageous to handle these waste by-products both economically and environmentally. Avocado seeds are rich in polysaccharides, proteins, lipids, vitamins, minerals, and other bioactive substances. The nutritional and phytochemical composition of avocado seeds has been well studied and discussed. Avocado-seed extracts also have many health-related bioactive properties, such as anti-hyperglycaemic, anticancer, anti-hypercholesterolemia, antioxidant, anti-inflammatory, and anti-neurogenerative effects are clearly demonstrated how these properties can be used to formulate or fortify food. The health-promoting properties of avocado seeds have been studied. These properties are attributed to various phytochemicals, such as acetogenin, catechin, epicatechin, procyanidin B1, estragole, etc. Additionally, items made from valorized avocado seeds that people can consume have been explored. The best applications of valorized by-products have been created for the pharmaceutical, functional food, and nutraceutical sectors while considering quality and safety. More clinical testing and product development research are required to prove the effectiveness of avocado seeds.
Collapse
Affiliation(s)
- Sneh Punia Bangar
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, SC 29634, USA,Corresponding authors at: Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania.
| | - Kyle Dunno
- Department of Packaging Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Sanju Bala Dhull
- Department of Food Science and Technology, Chaudhary Devi Lal University, Sirsa-125055, India
| | - Anil Kumar Siroha
- Department of Food Science and Technology, Chaudhary Devi Lal University, Sirsa-125055, India
| | - Sushil Changan
- Division of Crop Physiology, Biochemistry and Post-Harvest Technology, ICAR – Central Potato Research Institute, Shimla 171001, India
| | - Sajid Maqsood
- Department of Food Science, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Alexandru Vasile Rusu
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania,Animal Science and Biotechnology Faculty, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania,Corresponding authors at: Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania.
| |
Collapse
|
16
|
Liu R, Qin S, Li W. Phycocyanin: Anti-inflammatory effect and mechanism. Biomed Pharmacother 2022; 153:113362. [DOI: 10.1016/j.biopha.2022.113362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/28/2022] Open
|
17
|
V. Lima B, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Harnessing chitosan and poly-(γ-glutamic acid)-based biomaterials towards cancer immunotherapy. MATERIALS TODAY ADVANCES 2022; 15:100252. [DOI: 10.1016/j.mtadv.2022.100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
18
|
Guan Z, Feng Q. Chitosan and Chitooligosaccharide: The Promising Non-Plant-Derived Prebiotics with Multiple Biological Activities. Int J Mol Sci 2022; 23:ijms23126761. [PMID: 35743209 PMCID: PMC9223384 DOI: 10.3390/ijms23126761] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 12/24/2022] Open
Abstract
Biodegradable chitin is the second-most abundant natural polysaccharide, widely existing in the exoskeletons of crabs, shrimps, insects, and the cell walls of fungi. Chitosan and chitooligosaccharide (COS, also named chitosan oligosaccharide) are the two most important deacetylated derivatives of chitin. Compared with chitin, chitosan and COS not only have more satisfactory physicochemical properties but also exhibit additional biological activities, which cause them to be widely applied in the fields of food, medicine, and agriculture. Additionally, due to their significant ability to improve gut microbiota, chitosan and COS are deemed prospective prebiotics. Here, we introduced the production, physicochemical properties, applications, and pharmacokinetic characteristics of chitosan and COS. Furthermore, we summarized the latest research on their antioxidant, anti-inflammatory, and antimicrobial activities. Research progress on the prebiotic functions of chitosan and COS is particularly reviewed. We creatively analyzed and discussed the mechanisms and correlations underlying these activities of chitosan and COS and their physicochemical properties. Our work enriched people's understanding of these non-plant-derived prebiotics. Based on this review, the future directions of research on chitosan and COS are explored. Collectively, optimizing the production technology of chitin derivatives and enriching understanding of their biological functions will shed more light on their capability to improve human health.
Collapse
Affiliation(s)
- Zhiwei Guan
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Human Microbiome, School of Stomatology, Shandong University, Jinan 250012, China;
- School of Life Science, Qilu Normal University, Jinan 250200, China
| | - Qiang Feng
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Human Microbiome, School of Stomatology, Shandong University, Jinan 250012, China;
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266347, China
- Correspondence:
| |
Collapse
|
19
|
Pharmacological mechanisms of chitotriose as a redox regulator in the treatment of rat inflammatory bowel disease. Biomed Pharmacother 2022; 150:112988. [PMID: 35468583 DOI: 10.1016/j.biopha.2022.112988] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/15/2022] [Accepted: 04/17/2022] [Indexed: 11/20/2022] Open
Abstract
Although extensive development has been made in the treatment of inflammatory bowel disease (IBD), adverse effects and incomplete efficacy of currently used medications are continuous challenge. Accumulated reports on the benefits of chitosan oligosaccharides in intestinal disorders make chitotriose (COS) a breakthrough in the development of new IBD drugs. This study aimed to investigate the biosafety, efficacy and pharmacological mechanisms of COS in the treatment of experimental IBD in compare with the commercial 5-Aminosalicylic acid (5-ASA). In this study, COS effectively relieved active inflammation, restored epithelial function, and reduced intestinal fibrosis. Further investigation demonstrated that COS treatment regulated redox state of the colon tissue by stimulating the transcription factor nuclear factor E2-related factor 2 (Nrf2), increasing production of endogenous antioxidants, and alleviating oxidative stress. The offset of oxidative stress shut down the nuclear factor kappa-B (NF-ĸB) inflammatory pathway, mitophagy of epithelial cells, M2 macrophage polarization in pre-fibrotic inflammation, and myofibroblast activation in intestinal fibrogenesis. In conclusion, COS is a safe and effective therapeutic agent for experimental IBD as a redox regulator. Our results expand the current understanding of the pharmacology of chitosan oligosaccharides for IBD treatment and provides experimental basis for the medicinal development of small molecule carbohydrates.
Collapse
|
20
|
Punia Bangar S, Dunno K, Kumar M, Mostafa H, Maqsood S. A comprehensive review on lotus seeds (Nelumbo nucifera Gaertn.): Nutritional composition, health-related bioactive properties, and industrial applications. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
21
|
Shan M, Yu X, Li Y, Fu C, Zhang C. Vitamin B6 Alleviates Lipopolysaccharide-induced Myocardial Injury by Ferroptosis and Apoptosis Regulation. Front Pharmacol 2022; 12:766820. [PMID: 35002705 PMCID: PMC8740299 DOI: 10.3389/fphar.2021.766820] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/04/2021] [Indexed: 01/15/2023] Open
Abstract
Vitamin B6 (VitB6) is a water-soluble vitamin and includes pyridoxine, pyridoxal, pyridoxamine, and their phosphorylated forms. In the current study, we demonstrated that VitB6 could improve lipopolysaccharide (LPS)-induced myocardial injury. We demonstrated that VitB6 can suppress LPS-induced oxidative stress and lipid peroxidation that lead to ferroptosis and apoptosis in vivo and in vitro. Moreover, we found that VitB6 can regulate the expression of iron regulatory proteins, maintaining intracellular iron homeostasis. To confirm that VitB6 could inhibit LPS-induced ferroptosis and apoptosis, we pretreated mice with ferrostatin-1 (Fer-1) and emricasan that efficiently mimicked VitB6 pharmacological effects. This improved the survival rate of mice challenged with a high LPS dose. In addition, VitB6 regulated the expression of LPS-induced apoptosis-related proteins and iron regulatory proteins. It mediated the expression of Nrf2, transcription factor NF-E2-related factor 2, which promoted the expression of antioxidant enzymes and restrained LPS-induced ferroptosis and apoptosis. Overall, our results indicated that VitB6 can be used on novel therapies to relieve LPS-induced myocardial injury.
Collapse
Affiliation(s)
- Meirong Shan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China.,Department of Geriatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xujie Yu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Yajie Li
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changning Fu
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
22
|
Gu YF, Chen YP, Jin R, Wang C, Wen C, Zhou YM. Dietary chitooligosaccharide supplementation alleviates intestinal barrier damage, and oxidative and immunological stress in lipopolysaccharide-challenged laying hens. Poult Sci 2022; 101:101701. [PMID: 35150943 PMCID: PMC8844238 DOI: 10.1016/j.psj.2022.101701] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/15/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023] Open
Abstract
This study aimed to investigate the effects of chitooligosaccharide (COS) on intestinal barrier, antioxidant capacity, and immunity of lipopolysaccharide (LPS)-challenged laying hens. A total of 360 Hy-line Brown laying hens (80-wk-old) were randomly divided into 5 groups with 6 replicates of 12 birds. Hens were fed a corn-soybean meal basal diet supplemented with different COS levels (0; 5; 10; 15; 20 mg/kg) for 8 wk. The results showed that 15 mg/kg COS administration elevated albumen height and Haugh unit (P < 0.05), and numerically optimized productive performance (P > 0.05), therefore, the dosage of 15 mg/kg was chosen for the subsequent experiment. Thereafter, 12 birds from non-supplemented group were randomly selected and assigned into 2 groups, and birds in each group were administered (1.5 mg/kg BW, i.p.) with saline (control group) or LPS (challenge group). Another 6 hens from 15 mg/kg COS-supplemented group were selected and injected with LPS in the same way. Compared with the control group, LPS-challenged birds exhibited elevated circulating diamine oxidase activity, and reduced jejunal villus height and ratio of villus height to crypt depth, and these indices were reversed to control levels by COS (P < 0.05). Also, LPS increased malondialdehyde accumulation and reduced several antioxidant enzyme activities in the intestinal mucosa (P < 0.05). Additionally, LPS increased jejunal secretory IgA and interferon-γ (IFN-γ), and ileal secretory IgA, IgM, and interleukin-1β (IL-1β) concentrations, whereas COS reduced jejunal IFN-γ and IL-1β, and ileal IgM levels (P < 0.05). Moreover, LPS down-regulated mRNA abundance of jejunal occludin and claudin 2, and upregulated expression of jejunal nuclear factor erythroid-2 related factor 2, superoxide dismutase 1, and IFN-γ as well as ileal IL-1β (P < 0.05). Besides, COS increased jejunal occludin and ileal claudin 2, nuclear factor erythroid-2 related factor 2, and heme oxygenase-1 expression, and decreased jejunal IFN-γ and IL-1β abundance (P < 0.05). These results suggested that COS could alleviate LPS-induced intestinal barrier impairment, and oxidative and immunological stress in laying hens.
Collapse
Affiliation(s)
- Y F Gu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Y P Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - R Jin
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - C Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - C Wen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Y M Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.
| |
Collapse
|
23
|
Lynch RI, Lavelle EC. Immuno-modulatory biomaterials as anti-inflammatory therapeutics. Biochem Pharmacol 2022; 197:114890. [PMID: 34990595 DOI: 10.1016/j.bcp.2021.114890] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022]
Abstract
Biocompatible and biodegradable biomaterials are used extensively in regenerative medicine and serve as a tool for tissue replacement, as a platform for regeneration of injured tissue, and as a vehicle for delivery of drugs. One of the key factors that must be addressed in developing successful biomaterial-based therapeutics is inflammation. Whilst inflammation is initially essential for wound healing; bringing about clearance of debris and infection, prolonged inflammation can result in delayed wound healing, rejection of the biomaterial, further tissue damage and increased scarring and fibrosis. In this context, the choice of biomaterial must be considered carefully to minimise further induction of inflammation. Here we address the ability of the biomaterials themselves to modulate inflammatory responses and outline how the physico-chemical properties of the materials impact on their pro and anti-inflammatory properties (Fig. 1).
Collapse
Affiliation(s)
- Roisin I Lynch
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590, Dublin 2, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590, Dublin 2, Ireland.
| |
Collapse
|
24
|
Zhang X, Liang S, Gao X, Huang H, Lao F, Dai X. Protective Effect of Chitosan Oligosaccharide against Hydrogen Peroxide-Mediated Oxidative Damage and Cell Apoptosis via Activating Nrf2/ARE Signaling Pathway. Neurotox Res 2021; 39:1708-1720. [PMID: 34622385 DOI: 10.1007/s12640-021-00419-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022]
Abstract
Chitosan oligosaccharide (COS), hydrolyzed and deacetylated from chitosan, has been reported to possess varieties of biological activities. Alzheimer's disease (AD) is a multifactorial progressive neurodegenerative disorder characterized by cognitive decline and memory loss, where oxidative stress was reported to be an overwhelming cause of the occurrence of AD. We have previously reported that COS could significantly decrease cell death, ROS generation, and lipid peroxidation, though the potential mechanism was yet to be determined. This study was designed to investigate the neuroprotective effect of COS against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis in neuronal SH-SY5Y cells. Our results indicated that COS could dose-dependently scavenge H2O2 in the cell-free systems. Accordingly, COS markedly decreased H2O2-induced cell apoptosis and intracellular ROS generation, while increased antioxidant capacity in SH-SY5Y cells. Further, COS significantly reduced the expression of Bax and upregulated Bcl-2. The mRNA and protein expression levels of nuclear Nrf2, heme oxygenase 1 (HO-1), and NAD(P)H: quinone oxidoreductase 1 (NQO1) were significantly increased upon COS treatment. Moreover, Nrf2-siRNA evidently reversed the promotive effect of COS on expression levels of HO-1 and NQO1, and ARE-driven transcriptional activity as determined by double-luciferase reporter gene assay. Besides, COS reversed H2O2-mediated increased phosphorylation of ERK1/2 and p38 MAPK. In conclusion, our findings indicate that COS could protect SH-SY5Y cells from oxidative damage and apoptosis via regulating Nrf2/ARE signaling pathway, which may provide new applications for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Xiaoxia Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Shuang Liang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Xiaohan Gao
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Hanchang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Fengxue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing, 100191, China.
| |
Collapse
|
25
|
Chang Q, Cai H, Wei L, Lan R. Chitosan oligosaccharides alleviate acute heat stress-induced oxidative damage by activating ERK1/2-mediated HO-1 and GSH-Px gene expression in breast muscle of broilers. Poult Sci 2021; 101:101515. [PMID: 34826744 PMCID: PMC8626842 DOI: 10.1016/j.psj.2021.101515] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022] Open
Abstract
The purpose of this study was to evaluate the effects of chitosan oligosaccharides (COS) on acute heat stress (AHS) induced poor meat quality by alleviating oxidative damage through mitogen-activated protein kinase-nuclear factor-erythroid 2-related factor 2-antioxidant responsive element (MAPK-Nrf2-ARE) signaling pathway. A total of 108 thirty-five-day-old Chinese indigenous broilers (Luhua chicken) was used for this 42-d experiment. The broilers were randomly allocated to 3 treatments: control group (CON), AHS group, and AHS with 400 mg/kg COS supplementation (AHS-C) group. Both CON and AHS groups given the basal diet, and the AHS-C group given the basal diet with 400 mg/kg COS supplementation. On d 42, broilers in the AHS and AHS-C groups treated with AHS (increasing temperature from 24 to 34°C in 2-h and held for another 2-h), and the CON group under normal temperature (24°C). AHS exposure elevated (P < 0.05) body temperature (rectal, comb, eyelids, and feet) of broilers, increased (P < 0.05) breast muscle lightness (L*), drip loss, share force, hydrogen peroxide (H2O2) scavenging activity, reactive oxygen species (ROS) production, malondialdehyde (MDA) content, and catalase (CAT) activity, however, decreased (P < 0.05) pH45min, pH24h, redness (a*), and relative expression of heme oxygenase-1 (HO-1). Compared to the AHS group, dietary COS supplementation increased (P < 0.05) breast muscle pH45min, pH24h, and a*, H2O2 scavenging activity, as well as relative expression of HO-1 and glutathione peroxidase (GSH-Px), however, decreased (P < 0.05) drip loss, share force, superoxide anion free radicals (O2•−) scavenging activity, ROS production, and MDA content. It was concluded that AHS impaired meat quality, which may be related to oxidative damage, as evidenced by increasing ROS production, MDA content, and decreasing the relative expression of HO-1. Dietary COS supplementation could effectively elevate the meat quality of broilers exposed to AHS via decreasing ROS production, activating the Nrf2 pathway, and Nrf2-mediated HO-1 and GSH-Px gene expression.
Collapse
Affiliation(s)
- Qingqing Chang
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524-088, P.R. China
| | - Haoan Cai
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524-088, P.R. China
| | - Linlin Wei
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524-088, P.R. China
| | - Ruixia Lan
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524-088, P.R. China.
| |
Collapse
|
26
|
Cytoprotective Peptides from Blue Mussel Protein Hydrolysates: Identification and Mechanism Investigation in Human Umbilical Vein Endothelial Cells Injury. Mar Drugs 2021; 19:md19110609. [PMID: 34822480 PMCID: PMC8620150 DOI: 10.3390/md19110609] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease represents a leading cause of mortality and is often characterized by the emergence of endothelial dysfunction (ED), a physiologic condition that takes place in the early progress of atherosclerosis. In this study, two cytoprotective peptides derived from blue mussel chymotrypsin hydrolysates with the sequence of EPTF and FTVN were purified and identified. Molecular mechanisms underlying the cytoprotective effects against oxidative stress which lead to human umbilical vein endothelial cells (HUVEC) injury were investigated. The results showed that pretreatment of EPTF, FTVN and their combination (1:1) in 0.1 mg/mL significantly reduced HUVEC death due to H2O2 exposure. The cytoprotective mechanism of these peptides involves an improvement in the cellular antioxidant defense system, as indicated by the suppression of the intracellular ROS generation through upregulation of the cytoprotective enzyme heme oxygenase-1. In addition, H2O2 exposure triggers HUVEC damage through the apoptosis process, as evidenced by increased cytochrome C release, Bax protein expression, and the elevated amount of activated caspase-3, however in HUVEC pretreated with peptides and their combination, the presence of those apoptotic stimuli was significantly decreased. Each peptide showed similar cytoprotective effect but no synergistic effect. Taken together, these peptides may be especially important in protecting against oxidative stress-mediated ED.
Collapse
|
27
|
Yu J, Li W, Xiao X, Huang Q, Yu J, Yang Y, Han T, Zhang D, Niu X. (-)-Epicatechin gallate blocks the development of atherosclerosis by regulating oxidative stress in vivo and in vitro. Food Funct 2021; 12:8715-8727. [PMID: 34365492 DOI: 10.1039/d1fo00846c] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
(-)-Epicatechin gallate (ECG), as a compound in green tea extract polyphenols, has specific therapeutic effects against oxidative stress. Oxidative stress exists throughout the pathological development of atherosclerosis. In this study, two atherosclerosis models, oxidized low-density lipoprotein (ox-LDL)-induced vascular smooth muscle cells (VSMCs) and high fat diet (HFD)-induced ApoE-/- mice, were applied to explore the mechanism of ECG intervention on AS. In vivo and in vitro studies showed that ECG reduced the level of MDA and increased the activity of SOD, which are oxidative stress factors. ECG also improved HFD-induced disorder of lipid factor expression in the serum of ApoE-/- mice and alleviated oxidative stress by enhancing the antioxidant activity. The potential mechanism was supposed to be the inhibition of the phosphorylation of p65 by ECG in the NF-κB pathway in the aorta, thereby blocking the expression of inflammatory mediators. In addition, ECG increased the stability of atherosclerosis plaques by reducing the expression of MMP-2 and ICAM-1 in atherosclerosis diseased tissues. ECG reduced lipid accumulation in the aorta and its roots and also plaque neoplasia. Western blotting experiments indicated that ECG increased the nuclear transfer of Nrf2 and the expression of heme oxygenase 1 (HO-1) was increased. These results demonstrated that ECG significantly reduced the formation of aortic plaque in ApoE-/- mice which was possibly triggered by the inhibition of hyperlipidemia and oxidative stress that exhibited the anti-atherosclerotic potential.
Collapse
Affiliation(s)
- Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, P.R. China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, P.R. China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P.R. China.
| |
Collapse
|
28
|
Agostini SBN, Malta IHS, Rodrigues RF, Freitas JTJ, Lino MEDS, Dos Santos RS, Elisei LS, Moraes TR, Giusto LADR, de Oliveira MK, Bassi da Silva J, Bruschi ML, Santos AMD, Nogueira DA, Novaes RD, Pereira GR, Galdino G, Carvalho FC. Preclinical evaluation of methotrexate-loaded polyelectrolyte complexes and thermosensitive hydrogels as treatment for rheumatoid arthritis. Eur J Pharm Sci 2021; 163:105856. [PMID: 33882329 DOI: 10.1016/j.ejps.2021.105856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/29/2022]
Abstract
This work proposes new methotrexate (MTX) loaded drug delivery systems (DDS) to treat rheumatoid arthritis via the intra-articular route: a poloxamer based thermosensitive hydrogel (MTX-HG), oligochitosan and hypromellose phthalate-based polyelectrolyte complexes (MTX-PEC) and their association (MTX-PEC-HG). MTX-PEC showed 470 ± 166 nm particle size, 0.298 ± 0.108 polydispersity index, +26 ± 2 mV and 74.3 ± 5.8% MTX efficiency entrapment and particle formation was confirmed by infrared spectroscopy and thermal analysis. MTX-HG and MTX-PEC-HG gelled at 36.7°C. MTX drug release profile was prolonged for MTX-HG and MTX-PEC-HG, and faster for MTX-PEC and free MTX. The in vivo effect of the MTX-DDSs systems was evaluated in induced arthritis rats as single intra-articular dose. The assessed parameters were the mechanical nociceptive threshold, the plasmatic IL-1β level and histological analysis of the tibiofemoral joint. MTX-HG and MTX-PEC-HG performance were similar to free MTX and worse than oral MTX, used as positive control. All DDSs showed some irritative effect, for which further studies are required. MTX-PEC was the best treatment on recovering cartilage damage and decreasing allodynia. Thus, MTX-PEC demonstrated potential to treat rheumatoid arthritis, with the possibility of decreasing the systemic exposure to the drug.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Merelym Ketterym de Oliveira
- Instituto de Ciências Biomédicas, Departamento de Ciências Fisiológicas, Universidade Federal de Alfenas, Brazil
| | - Jéssica Bassi da Silva
- Laboratório de Pesquisa e Desenvolvimento de Sistemas de Liberação de Fármacos, Departamento de Farmácia, Universidade Estadual de Maringá, Brazil
| | - Marcos Luciano Bruschi
- Laboratório de Pesquisa e Desenvolvimento de Sistemas de Liberação de Fármacos, Departamento de Farmácia, Universidade Estadual de Maringá, Brazil
| | - Aline Martins Dos Santos
- Faculdade de Ciências Farmacêuticas, Universidade Estadual Paulista "Julio de Mesquita Filho". UNESP, Araraquara, Brazil
| | | | - Rômulo Dias Novaes
- Instituto de Ciências Biomédicas, Departamento de Biologia Estrutural, Universidade Federal de Alfenas, Brazil
| | - Gislaine Ribeiro Pereira
- Escola de Farmácia, Departamento de Fármacos e Alimentos, Universidade Federal de Alfenas, Brazil
| | - Giovane Galdino
- Instituto de Ciência da Motricidade, Universidade Federal de Alfenas, Brazil
| | - Flávia Chiva Carvalho
- Escola de Farmácia, Departamento de Fármacos e Alimentos, Universidade Federal de Alfenas, Brazil
| |
Collapse
|
29
|
Wu YH, Chueh KS, Chuang SM, Long CY, Lu JH, Juan YS. Bladder Hyperactivity Induced by Oxidative Stress and Bladder Ischemia: A Review of Treatment Strategies with Antioxidants. Int J Mol Sci 2021; 22:ijms22116014. [PMID: 34199527 PMCID: PMC8199707 DOI: 10.3390/ijms22116014] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023] Open
Abstract
Overactive bladder (OAB) syndrome, including frequency, urgency, nocturia and urgency incontinence, has a significantly negative impact on the quality-of-life scale (QoL) and can cause sufferer withdrawal from social activities. The occurrence of OAB can result from an imbalance between the production of pro-oxidants, such as free radicals and reactive species, and their elimination through protective mechanisms of antioxidant-induced oxidative stress. Several animal models, such as bladder ischemia/reperfusion (I/R), partial bladder outlet obstruction (PBOO) and ovarian hormone deficiency (OHD), have suggested that cyclic I/R during the micturition cycle induces oxidative stress, leading to bladder denervation, bladder afferent pathway sensitization and overexpression of bladder-damaging molecules, and finally resulting in bladder hyperactivity. Based on the results of previous animal experiments, the present review specifically focuses on four issues: (1) oxidative stress and antioxidant defense system; (2) oxidative stress in OAB and biomarkers of OAB; (3) OAB animal model; (4) potential nature/plant antioxidant treatment strategies for urinary dysfunction with OAB. Moreover, we organized the relationships between urinary dysfunction and oxidative stress biomarkers in urine, blood and bladder tissue. Reviewed information also revealed the summary of research findings for the effects of various antioxidants for treatment strategies for OAB.
Collapse
Affiliation(s)
- Yi-Hsuan Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-H.W.); (K.-S.C.)
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Urology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung 80661, Taiwan
| | - Kuang-Shun Chueh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-H.W.); (K.-S.C.)
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Shu-Mien Chuang
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Cheng-Yu Long
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jian-He Lu
- Emerging Compounds Research Center, Department of Environmental Science and Engineering, College of Engineering, National Pingtung University of Science and Technology, Pintung 91201, Taiwan;
| | - Yung-Shun Juan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (Y.-H.W.); (K.-S.C.)
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-7-3121101; Fax: +886-7-3506269
| |
Collapse
|
30
|
Mei QX, Hu JH, Huang ZH, Fan JJ, Huang CL, Lu YY, Wang XP, Zeng Y. Pretreatment with chitosan oligosaccharides attenuate experimental severe acute pancreatitis via inhibiting oxidative stress and modulating intestinal homeostasis. Acta Pharmacol Sin 2021; 42:942-953. [PMID: 33495520 PMCID: PMC8149410 DOI: 10.1038/s41401-020-00581-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022]
Abstract
Severe acute pancreatitis (SAP) is a severe acute abdominal disease. Recent evidence shows that intestinal homeostasis is essential for the management of acute pancreatitis. Chitosan oligosaccharides (COS) possess antioxidant activity that are effective in treating various inflammatory diseases. In this study we explored the potential therapeutic effects of COS on SAP and underlying mechanisms. Mice were treated with COS (200 mg·kg-1·d-1, po) for 4 weeks, then SAP was induced in the mice by intraperitoneal injection of caerulein. We found that COS administration significantly alleviated the severity of SAP: the serum amylase and lipase levels as well as pancreatic myeloperoxidase activity were significantly reduced. COS administration suppressed the production of proinflammatory cytokines (TNF-α, IL-1β, CXCL2 and MCP1) in the pancreas and ileums. Moreover, COS administration decreased pancreatic inflammatory infiltration and oxidative stress in SAP mice, accompanied by activated Nrf2/HO-1 and inhibited TLR4/NF-κB and MAPK pathways. We further demonstrated that COS administration restored SAP-associated ileal damage and barrier dysfunction. In addition, gut microbiome analyses revealed that the beneficial effect of COS administration was associated with its ability to improve the pancreatitis-associated gut microbiota dysbiosis; in particular, probiotics Akkermansia were markedly increased, while pathogenic bacteria Escherichia-Shigella and Enterococcus were almost eliminated. The study demonstrates that COS administration remarkably attenuates SAP by reducing oxidative stress and restoring intestinal homeostasis, suggesting that COS might be a promising prebiotic agent for the treatment of SAP.
Collapse
Affiliation(s)
- Qi-Xiang Mei
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
| | - Jun-Hui Hu
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
| | - Ze-Hua Huang
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
| | - Jun-Jie Fan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
| | - Chun-Lan Huang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
| | - Ying-Ying Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China
| | - Xing-Peng Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China.
| | - Yue Zeng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201600, China.
| |
Collapse
|
31
|
Wang YR, Zhang XN, Meng FG, Zeng T. Targeting macrophage polarization by Nrf2 agonists for treating various xenobiotics-induced toxic responses. Toxicol Mech Methods 2021; 31:334-342. [PMID: 33627030 DOI: 10.1080/15376516.2021.1894624] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Macrophages can polarize into different phenotypes in response to different microenvironmental stimuli. Macrophage polarization has been assigned to two extreme states, namely proinflammatory M1 and anti-inflammatory M2. Accumulating evidences have demonstrated that M1 polarized macrophages contribute to various toxicants-induced deleterious effects. Switching macrophages from proinflammatory M1 phenotype toward anti-inflammatory M2 phenotype could be a promising approach for treating various inflammatory diseases. Studies in the past few decades have revealed that nuclear factor erythroid 2-related factor 2 (Nrf2) can modulate the polarization of macrophages. Specifically, activation of Nrf2 could block M1 stimuli-induced production of proinflammatory cytokines and chemokines, and shift the polarization of macrophages toward M2 by cross-talking with nuclear factor kappa-B (NF-κB), mitogen-activated protein kinases (MAPKs), peroxisome proliferator-activated receptor γ (PPARγ), and autophagy. Importantly, a great number of studies have confirmed the beneficial effects of natural and synthesized Nrf2 agonists on various inflammatory diseases; however, most of these compounds are far away from clinical application due to lack of characterization and defects of study designs. Interestingly, some endogenous Nrf2 inducers and compounds with dual activities (such as the Nrf2 inducing and CO releasing effects) exhibit potent anti-inflammatory effects, which points out an important direction for future researches.
Collapse
Affiliation(s)
- Yi-Ran Wang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiu-Ning Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fan-Ge Meng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
32
|
Wei L, Li Y, Chang Q, Guo G, Lan R. Effects of chitosan oligosaccharides on intestinal oxidative stress and inflammation response in heat stressed rats. Exp Anim 2021; 70:45-53. [PMID: 32921697 PMCID: PMC7887628 DOI: 10.1538/expanim.20-0085] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
This study was to verify the effects of chitosan oligosaccharides (COS) on intestinal integrity, oxidative status, and inflammatory response in a heat-stressed rat model. A total of 24 male Sprague Dawley rats were randomly divided into 3 treatment: CON, the control group; HS, the heat stress group; HSC, the heat stress group with 200 mg/kg COS. Rats in the HS and HSC group exposed to a cyclical heat stress for 7 consecutive days. The CON and HS group provided basal diet, and the HSC group provided the same diet with 200 mg/kg COS. Compared with the HS group, rats in the HSC group had lower serum diamine oxidase and D-lactate acid level, higher villus height of jejunum and ileum, lower malondialdehyde (MDA) content in duodenum, jejunum, and ileum mucosa, higher glutathione peroxidase (GSH-Px), catalase (CAT) and total antioxidant capacity (T-AOC) activity in duodenum mucosa, higher T-AOC activity in jejunum mucosa, and higher glutathione (GSH) level in ileum mucosa. Compared with the HS group, rats in the HSC group had higher interleukin-10 (IL-10) level, but lower tumor necrosis factor-α (TNF-α) level in duodenum, jejunum, and ileum mucosa. These results indicated that COS may alleviate intestinal damage under heat stress condition, probably by modulating intestinal inflammatory response and oxidative status.
Collapse
Affiliation(s)
- Linlin Wei
- College of Coastal Agriculture Science, Guangdong Ocean University, No. 1 Haida Road, Mazhang District, Zhanjiang 524-088, Guangdong, P.R. China
| | - Yaxuan Li
- College of Coastal Agriculture Science, Guangdong Ocean University, No. 1 Haida Road, Mazhang District, Zhanjiang 524-088, Guangdong, P.R. China
| | - Qingqing Chang
- College of Coastal Agriculture Science, Guangdong Ocean University, No. 1 Haida Road, Mazhang District, Zhanjiang 524-088, Guangdong, P.R. China
| | - Guangzhen Guo
- College of Coastal Agriculture Science, Guangdong Ocean University, No. 1 Haida Road, Mazhang District, Zhanjiang 524-088, Guangdong, P.R. China
| | - Ruixia Lan
- College of Coastal Agriculture Science, Guangdong Ocean University, No. 1 Haida Road, Mazhang District, Zhanjiang 524-088, Guangdong, P.R. China
| |
Collapse
|
33
|
The Protect Effects of Chitosan Oligosaccharides on Intestinal Integrity by Regulating Oxidative Status and Inflammation under Oxidative Stress. Mar Drugs 2021; 19:md19020057. [PMID: 33503912 PMCID: PMC7911331 DOI: 10.3390/md19020057] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to evaluate the effects of the dietary supplementation of chitosan oligosaccharides (COS) on intestinal integrity, oxidative status, and the inflammation response with hydrogen peroxide (H2O2) challenge. In total, 30 rats were randomly assigned to three groups with 10 replications: CON group, basal diet; AS group, basal diet + 0.1% H2O2 in drinking water; ASC group, basal diet + 200 mg/kg COS + 0.1% H2O2 in drinking water. The results indicated that COS upregulated (p < 0.05) villus height (VH) of the small intestine, duodenum, and ileum; mucosal glutathione peroxidase activity; jejunum and ileum mucosal total antioxidant capacity; duodenum and ileum mucosal interleukin (IL)-6 level; jejunum mucosal tumor necrosis factor (TNF)-α level; duodenum and ileum mucosal IL-10 level; the mRNA expression level of zonula occludens (ZO)-1 in the jejunum and ileum, claudin in the duodenum, nuclear factor-erythroid 2-like 2 in the jejunum, and heme oxygenase-1 in the duodenum and ileum; and the protein expression of ZO-1 and claudin in jejunum; however, it downregulated (p < 0.05) serum diamine oxidase activity and D-lactate level; small intestine mucosal malondialdehyde content; duodenum and ileum mucosal IL-6 level; jejunum mucosal TNF-α level; and the mRNA expression of IL-6 in the duodenum and jejunum, and TNF-α in the jejunum and ileum. These results suggested COS could maintain intestinal integrity under oxidative stress by modulating the intestinal oxidative status and release of inflammatory cytokines.
Collapse
|
34
|
Dietary chitosan oligosaccharides alleviate heat stress-induced intestinal oxidative stress and inflammatory response in yellow-feather broilers. Poult Sci 2020; 99:6745-6752. [PMID: 33248590 PMCID: PMC7705058 DOI: 10.1016/j.psj.2020.09.050] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/04/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023] Open
Abstract
The purpose of this study was to evaluate the effects of chitosan oligosaccharides (COS) on intestinal permeability, morphology, antioxidant status, and inflammatory response in heat-stressed broilers. A total of 108 thirty-five-day-old Chinese yellow-feather broilers (body weight 470.31 ± 13.15 g) were randomly allocated to 3 dietary treatments as follows: CON group, basal diet and raised under normal temperature (24°C); HS group, basal diet and raised under cycle heat stress (34°C from 10:00–18:00 and 24°C for the rest time); HSC group, basal diet with 200 mg/kg COS supplementation and raised under cycle heat stress. Each treatment had 6 replication pens and 6 broilers per pen. Compared with the CON group, heat stress decreased (P < 0.05) the relative weight of duodenum and jejunum; the relative length and villus height (VH) of duodenum, jejunum, and ileum; the ileum VH to crypt depth ratio; duodenum mucosal catalase (CAT) activity; and jejunum mucosal glutathione peroxidase (GSH-Px) and CAT activity, whereas it increased (P < 0.05) serum diamine oxidase (DAO) activity and D-lactate acid (D-LA) content, duodenum and jejunum mucosal malondialdehyde (MDA) and interleukin-1β (IL-1β) content, and ileum mucosal tumor necrosis factor-α content. Compared to the HS group, dietary COS supplementation increased (P < 0.05) the relative length of duodenum, jejunum, and ileum; the VH of jejunum and ileum; and duodenum and jejunum mucosal GSH-Px activity, whereas it decreased (P < 0.05) serum DAO activity and D-LA concentration and duodenum and jejunum mucosal MDA and IL-1β content. These results suggested that dietary COS supplementation had beneficial effects on intestinal morphology by increasing jejunum and ileum VH; permeability by decreasing serum DAO activity and D-LA content; antioxidant capacity by decreasing duodenum and jejunum mucosal MDA content and by increasing duodenum and jejunum GSH-Px activity; and inflammatory response by decreasing duodenum and jejunum mucosal IL-1β content.
Collapse
|
35
|
Yi Z, Luo X, Zhao L. Research Advances in Chitosan Oligosaccharides: From Multiple Biological Activities to Clinical Applications. Curr Med Chem 2020; 27:5037-5055. [PMID: 31309881 DOI: 10.2174/0929867326666190712180147] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/12/2019] [Accepted: 06/16/2019] [Indexed: 12/14/2022]
Abstract
Chitosan oligosaccharides (COS), hydrolysed products of chitosan, are low-molecular weight polymers with a positive charge and good biocompatibility. COS have recently been reported to possess various biological activities, including hypoglycaemic, hypolipidaemic, antioxidantantioxidant, immune regulation, anti-inflammatory, antitumour, antibacterial, and tissue engineering activities, exhibiting extensive application prospects. Currently, the biological processes and mechanisms of COS are attractive topics of study, ranging from the genetic, molecular and protein levels. This article reviews the recent discoveries about COS, especially in metabolic regulation, immune function and tissue repair, providing important insights into their multiple biological activities, medical benefits, and therapeutic mechanisms.
Collapse
Affiliation(s)
- Zhen Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiao Luo
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
36
|
Oh Y, Ahn CB, Je JY. Low molecular weight blue mussel hydrolysates inhibit adipogenesis in mouse mesenchymal stem cells through upregulating HO-1/Nrf2 pathway. Food Res Int 2020; 136:109603. [PMID: 32846625 DOI: 10.1016/j.foodres.2020.109603] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/08/2020] [Accepted: 07/24/2020] [Indexed: 10/23/2022]
Abstract
Blue mussel proteins are a good source of bioactive peptides. In this study, blue mussel hydrolysate (BMH) with anti-adipogenic effect in mouse mesenchymal stem cells (mMSC) was produced by peptic hydrolysis at 1:500 of pepsin/substrate ratio for 120 min. Additionally, BMH with below 1 kDa (BMH < 1 kDa) showed the highest anti-adipogenic effect in mMSC. BMH < 1 kDa increased lipolysis and down-regulated adipogenic transcription factors including peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein α (C/EBPα), and sterol regulatory element-binding protein 1 (SREBP1). Generation of intracellular reactive oxygen species during adipogenesis was markedly decreased by BMH < 1 kDa treatment, which is attributed to the up-regulation of heme oxygenase-1 (HO-1) through Nrf2 translocation into the nucleus. Moreover, ZnPP, HO-1 inhibitor, treatment abolished BMH < 1 kDa-mediated HO-1 expression and anti-adipogenic effect in mMSCs through down-regulating adipogenic transcription factors. Taken together, BMH < 1 kDa may be a potential ingredient of nutraceuticals and/or functional foods in ameliorating obesity.
Collapse
Affiliation(s)
- Yunok Oh
- Institute of Marine Life Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Chang-Bum Ahn
- Division of Food and Nutrition, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jae-Young Je
- Department of Marine-Bio Convergence Science, Pukyong National University, Busan 48547, Republic of Korea.
| |
Collapse
|
37
|
Wang Y, Xiong Y, Zhang A, Zhao N, Zhang J, Zhao D, Yu Z, Xu N, Yin Y, Luan X, Xiong Y. Oligosaccharide attenuates aging-related liver dysfunction by activating Nrf2 antioxidant signaling. Food Sci Nutr 2020; 8:3872-3881. [PMID: 32724648 PMCID: PMC7382186 DOI: 10.1002/fsn3.1681] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/08/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
Chitosan oligosaccharide (COS) is the depolymerized product of chitosan possessing various biological activities and protective effects against inflammation and oxidative injury. The aim of the present study was to investigate the antioxidant effects of COS supplements on aging-related liver dysfunction. We found that COS treatment significantly attenuated elevated liver function biomarkers and oxidative stress biomarkers and decreased antioxidative enzyme activities in liver tissues in D-galactose (D-gal)-treated mice. Furthermore, COS treatment significantly upregulated the expression of Nrf2 and its downstream target genes HO-1, NQO1, and CAT. Moreover, in vitro experiments showed that COS treatment played a vital role in protecting H2O2-exposed L02 cells against oxidative stress by activating Nrf2 antioxidant signaling. These data indicate that COS could protect against D-gal-induced hepatic aging by activating Nrf2 antioxidant signaling, which may provide novel applications for the prevention and treatment of aging-related hepatic dysfunction.
Collapse
Affiliation(s)
- Yueming Wang
- Department of AnatomySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Yanlei Xiong
- Department of PathologyXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Aiping Zhang
- Department of ImmunologySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Nannan Zhao
- Department of ImmunologySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Jiashen Zhang
- Department of AnatomySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Dongmei Zhao
- Department of AnatomySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Zhenhai Yu
- Department of AnatomySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Ning Xu
- Department of AnatomySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Yancun Yin
- Department of AnatomySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Xiying Luan
- Department of ImmunologySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| | - Yanlian Xiong
- Department of AnatomySchool of Basic MedicineBinzhou Medical UniversityYantaiChina
| |
Collapse
|
38
|
Gu L, Li S, Bai J, Zhang Q, Han Z. α-Lipoic acid protects against microcystin-LR induced hepatotoxicity through regeneration of glutathione via activation of Nrf2. ENVIRONMENTAL TOXICOLOGY 2020; 35:738-746. [PMID: 32061150 DOI: 10.1002/tox.22908] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 06/10/2023]
Abstract
Microcystins (MCs), as the most dominant bloom-forming strains in eutrophic surface water, can induce hepatotoxicity by oxidative stress. Alpha-lipoic acid (α-LA) is a super antioxidant that can induce the synthesis of antioxidants, such as glutathione (GSH), by nuclear factor erythroid 2-related factor 2 (Nrf2). However, the potential molecular mechanism of α-LA regeneration of GSH remains unclear. The present study aimed to investigate whether α-LA could reduce the toxicity of MCs induced in human hepatoma (HepG2), Bel7420 cells, and BALB/c mice by activating Nrf2 to regenerate GSH. Results showed that exposure to 10 μM microcystin-leucine arginine (MC-LR) reduced viability of HepG2 and Bel7402 cells and promoted the formation of reactive oxygen species (ROS) compared with untreated cells. Moreover, the protection of α-LA included reducing the level of ROS, increasing superoxide dismutase activity, and decreasing malondialdehyde. Levels of reduced glutathione (rGSH) and rGSH/oxidized glutathione were significantly increased in cells cotreated with α-LA and MC-LR compared to those treated with MC-LR alone, indicating an ability of α-LA to attenuate oxidative stress and MC-LR-induced cytotoxicity by increasing the amount of rGSH. α-LA can mediate GSH regeneration through the Nrf2 pathway under the action of glutathione reductase in MC-LR cell lines. Furthermore, the data also showed that α-LA-induced cytoprotection against MC-LR is associated with Nrf2 mediate pathway in vivo. These findings demonstrated the potential of α-LA to resist MC-LR-induced oxidative damage of liver.
Collapse
Affiliation(s)
- Lihong Gu
- Department of labor hygiene and environmental hygiene, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Shangchun Li
- Department of labor hygiene and environmental hygiene, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Bai
- Department of labor hygiene and environmental hygiene, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Qingbi Zhang
- Department of labor hygiene and environmental hygiene, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhixia Han
- Department of labor hygiene and environmental hygiene, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
39
|
Shen K, Tang Q, Fang X, Zhang C, Zhu Z, Hou Y, Lai M. The sustained release of dexamethasone from TiO 2 nanotubes reinforced by chitosan to enhance osteoblast function and anti-inflammation activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111241. [PMID: 32806259 DOI: 10.1016/j.msec.2020.111241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 01/07/2023]
Abstract
Controlling macrophage response to biomaterials is critical for the reduction of inflammation after implantation. Here we designed a sustained release system from TiO2 nanotubes (TNTs) to improve osteogenesis on titanium implants with anti-inflammatory properties. TNTs (around 70 nm diameter) were first fabricated on titanium surfaces by anodization, directly filled with the anti-inflammatory drug, dexamethasone (DEX) and then covered by chitosan (CHI) multilayer films. Primary osteoblast and macrophage (RAW 264.7) cells were cultured on untreated and treated titanium surfaces in vitro. Osteoblasts grown on CHI-coated Dex-filled TNTs surfaces displayed higher alkaline phosphatase (ALP) and mineralization, which was consistent with qRT-PCR analysis of osteoblastic genes including collagen type I (Col I), osteocalcin (OCN), osteopontin (OPN) and runt related transcription factor 2 (Runx2). In contrast, protein levels of nitric oxide (NO) and proinflammatory cytokines (TNF-α and IL-1β) from macrophages on Dex-filled TNTs, CHI-coated TNTs and CHI-coated Dex-filled TNTs were significantly lower, especially on CHI-coated Dex-filled TNTs surfaces compared to levels on titanium and TNTs. These results indicate that CHI-coated Dex-filled TNTs enhanced osteoblast differentiation and decreased the inflammatory response of macrophages. The approach presented here provides new insight into the modification of TNTs for the development of titanium-based implants.
Collapse
Affiliation(s)
- Ke Shen
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Qiang Tang
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Xingtang Fang
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Chunlei Zhang
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China
| | - Zhaojing Zhu
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Yanhua Hou
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Min Lai
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China.
| |
Collapse
|
40
|
Chitoheptaose Promotes Heart Rehabilitation in a Rat Myocarditis Model by Improving Antioxidant, Anti-Inflammatory, and Antiapoptotic Properties. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2394704. [PMID: 32351668 PMCID: PMC7171680 DOI: 10.1155/2020/2394704] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 12/25/2022]
Abstract
Background Myocarditis is one of the important causes of dilated cardiomyopathy, cardiac morbidity, and mortality worldwide. Chitosan oligosaccharides (COS) may have anti-inflammatory and cardioprotective effects on myocarditis. However, the exact molecular mechanism for the effects of functional COS on myocarditis remains unclear. Methods Anti-inflammatory activities of COS (chitobiose, chitotriose, chitotetraose, chitopentaose, chitohexaose, chitoheptaose, and chitooctaose) were measured in lipopolysaccharide- (LPS-) stimulated RAW264.7 cells. A rat model with myocarditis was established and treated with chitopentaose, chitohexaose, chitoheptaose, and chitooctaose. Serum COS were measured by using high-performance liquid chromatography (HPLC) in all rats. Myocarditis injury, the levels of reactive oxygen species (ROS), reactive nitrogen species (RNS), inflammatory factors, and apoptotic factors were also measured. Pearson's correlation coefficient test was used to explore the relationship between the levels of ROS/RNS and cardiac parameters. Results Among all chitosan oligosaccharides, the COS > degrees of polymerization (DP) 4 showed anti-inflammatory activities (the activity order was chitopentaose<chitohexaose<chitoheptaose<chitooctaose) by reducing the levels of interleukin- (IL-) 1β, IL-17A, and interferon- (IFN-) γ and increasing the level of IL-10. However, the serum level of chitooctaose was low whereas it showed significant therapeutic effects on myocarditis by improving cardiac parameters (left ventricular internal dimension, both end-systolic and end-diastolic, ejection fraction, and fractional shortening), inflammatory cytokines (IL-1β, IL-10, IL-17A, and IFN-γ), oxidative factors (ROS and RNS), and apoptotic factors (caspase 3, BAX, and BCL-2) when compared with chitopentaose, chitohexaose, and chitooctaose (COS DP > 4). The levels of ROS/RNS had a strong relationship with cardiac parameters. Conclusions Chitoheptaose plays a myriad of cardioprotective roles in the myocarditis model via its antioxidant, anti-inflammatory, and antiapoptotic activities.
Collapse
|
41
|
Poria cocos polysaccharides attenuated ox-LDL-induced inflammation and oxidative stress via ERK activated Nrf2/HO-1 signaling pathway and inhibited foam cell formation in VSMCs. Int Immunopharmacol 2020; 80:106173. [DOI: 10.1016/j.intimp.2019.106173] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/24/2019] [Accepted: 12/28/2019] [Indexed: 12/31/2022]
|
42
|
Tian L, Sun SS, Cui LB, Wang SQ, Peng ZW, Tan QR, Hou WG, Cai M. Repetitive Transcranial Magnetic Stimulation Elicits Antidepressant- and Anxiolytic-like Effect via Nuclear Factor-E2-related Factor 2-mediated Anti-inflammation Mechanism in Rats. Neuroscience 2020; 429:119-133. [PMID: 31918011 DOI: 10.1016/j.neuroscience.2019.12.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/03/2019] [Accepted: 12/13/2019] [Indexed: 12/26/2022]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) treatment is widely accepted as an evidence-based treatment option for depression and anxiety. However, the underlying mechanism of this treatment maneuver has not been clearly understood. The chronic unpredictable mild stress (CUMS) procedure was used to establish depression and anxiety-like behavior in rats. The rTMS was performed with a commercially available stimulator for seven consecutive days, and then depression and anxiety-like behaviors were subsequently measured. The expression of nuclear factor-E2-related factor 2 (Nrf2) was measured by western-blot, and the level of tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS), interleukin-1β (IL-1β), and interleukin-6 (IL-6) was measured with Enzyme-linked immunesorbent assay (ELISA) analyzing kits. Furthermore, a small interfering RNA was employed to knockdown Nrf2, after which the neurobehavioral assessment, Nrf2 nuclear expression, and the amount of inflammation factors were evaluated. Application of rTMS exhibited a significant antidepressant and anxiolytic-like effect, which was associated with the increased Nrf2 nuclear translocation and reduced level of TNF-α, iNOS, IL-1β, and IL-6 in the hippocampus. Following Nrf2 silencing, the antidepressant and anxiolytic-like effect produced by rTMS was abolished. Moreover, the elevated Nrf2 nuclear translocation, and the reduced production of TNF-α, iNOS, IL-1β, and IL-6 in hippocampus mediated by rTMS, were reversed by Nrf2 knockdown. Together, these results reveal that the Nrf2-induced anti-inflammation effect is crucial in regulating antidepressant-related behaviors produced by rTMS.
Collapse
Affiliation(s)
- Li Tian
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China
| | - Si-Si Sun
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China; Medical Department of Xi'an Emergency Center, the 111th of Fengcheng 4th Road, Xi'an 718900, Shaanxi, China
| | - Long-Biao Cui
- School of Medical Psychology, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China
| | - Shi-Quan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China
| | - Zheng-Wu Peng
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China
| | - Qing-Rong Tan
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China
| | - Wu-Gang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China
| | - Min Cai
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, the 127th of Changle Road, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
43
|
Zhang M, Xu Y, Jiang L. Sulforaphane attenuates angiotensin II-induced human umbilical vein endothelial cell injury by modulating ROS-mediated mitochondrial signaling. Hum Exp Toxicol 2020; 39:734-747. [PMID: 31957488 DOI: 10.1177/0960327119893414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The study aimed to investigate whether sulforaphane (SFN) protects against angiotensin II (Ang II)-mediated human umbilical vein endothelial cell (HUVEC) injury. Ang II treatment decreased HUVEC viability, increased cell apoptosis, decreased mitochondria membrane potential (MMP), impaired cytochrome c release, activated caspase 3/9, and induced reactive oxygen species (ROS) production, and nicotinamide adenine dinucleotide phosphate oxidase activity. Moreover, SFN treatment blunted Ang II-stimulated oxidative stress and mitochondria-related apoptosis in HUVECs. The ROS scavenger N-acetyl-l-cysteine reduced Ang II-induced oxidative stress and apoptosis, indicating that ROS generation is involved in the Ang II-induced mitochondria-mediated apoptotic pathway. SFN induced nuclear factor erythroid 2 (Nrf2) activation and expression in Ang II-stimulated HUVECs. Downregulation of Nrf2 expression by a target-specific siRNA revealed an Nrf2-dependent effect on the SFN-mediated attenuation of Ang II-induced apoptosis in HUVECs. Pretreatment with brusatol, an Nrf2-specific inhibitor, reversed the protective effects of SFN on Ang II-induced HUVEC injury. SFN treatment protected HUVECs from Ang II-induced damage by decreasing oxidative stress and ameliorating mitochondrial injury.
Collapse
Affiliation(s)
- M Zhang
- Division of Cardiology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Y Xu
- Division of Cardiology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - L Jiang
- Division of Cardiology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Kim HN, Kim JD, Park SB, Son HJ, Park GH, Eo HJ, Kim HS, Jeong JB. Anti-inflammatory activity of the extracts from Rodgersia podophylla leaves through activation of Nrf2/HO-1 pathway, and inhibition of NF-κB and MAPKs pathway in mouse macrophage cells. Inflamm Res 2020; 69:233-244. [PMID: 31907559 DOI: 10.1007/s00011-019-01311-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 12/13/2019] [Accepted: 12/23/2019] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE Recently, Rodgersia podophylla has been reported to exhibit anti-inflammatory activity. However, little is known about the potential mechanisms about its anti-inflammatory activity. We elucidated the anti-inflammatory mechanisms of leaves extracts from Rodgersia podophylla (RP-L) in RAW264.7 cells. MATERIALS AND METHODS LPS-induced NO was measured by Griess and mRNA of pro-inflammatory mediators was analyzed by RT-PCR. Cell viability was measured using MTT assay. The protein level was analyzed by Western blot. RESULTS RP-L significantly inhibited the production of the pro-inflammatory mediators such as NO, iNOS, IL-1β and IL-6 in LPS-stimulated RAW264.7 cells. RP-L increased HO-1 expression in RAW264.7 cells, and the inhibition of HO-1 by ZnPP reduced the inhibitory effect of RP-L against LPS-induced NO production in RAW264.7 cells. Inhibition of p38, ROS and GSK3β attenuated RP-L-mediated HO-1 expression. Inhibition of ROS inhibited p38 phosphorylation and GSK3β expression induced by RP-L. In addition, inhibition of GSK3β blocked RP-L-mediated p38 phosphorylation. RP-L induced nuclear accumulation of Nrf2, and inhibition of p38, ROS and GSK3β abolished RP-L-mediated nuclear accumulation of Nrf2. Furthermore, RP-L blocked LPS-induced degradation of IκB-α and nuclear accumulation of p65. RP-L also attenuated LPS-induced phosphorylation of ERK1/2 and p38. In GC/MS analysis of RP-L, pyrogallol was detected as bioactive compound for anti-inflammatory activity of RP-L. Pyrogallol was observed to activate HO-1 expression through ROS/GSK3β/p38/Nrf2/HO-1 signaling. CONCLUSIONS Our results suggest that RP-L exerts potential anti-inflammatory activity by activating ROS/GSK3β/p38/Nrf2/HO-1 signaling and inhibiting NF-κB and MAPK signaling in RAW264.7 cells. These findings suggest that RP-L may have great potential for the development of anti-inflammatory drug.
Collapse
Affiliation(s)
- Ha Na Kim
- Department of Medicinal Plant Resources, Andong National University, Andong, 36729, Republic of Korea
| | - Jeong Dong Kim
- Department of Medicinal Plant Resources, Andong National University, Andong, 36729, Republic of Korea
| | - Su Bin Park
- Department of Medicinal Plant Resources, Andong National University, Andong, 36729, Republic of Korea
| | - Ho-Jun Son
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yongju, 36040, Republic of Korea
| | - Gwang Hun Park
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yongju, 36040, Republic of Korea
| | - Hyun Ji Eo
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yongju, 36040, Republic of Korea
| | - Hyun-Seok Kim
- Department of Food Science and Biotechnology, Kyonggi University, Suwon, 16227, Republic of Korea
| | - Jin Boo Jeong
- Department of Medicinal Plant Resources, Andong National University, Andong, 36729, Republic of Korea. .,Agricultural Science and Technology Research Institute, Andong National University, Andong, 36729, Republic of Korea.
| |
Collapse
|
45
|
Chitosan Oligosaccharides Protect Sprague Dawley Rats from Cyclic Heat Stress by Attenuation of Oxidative and Inflammation Stress. Animals (Basel) 2019; 9:ani9121074. [PMID: 31816916 PMCID: PMC6940990 DOI: 10.3390/ani9121074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Heat stress has negative effects on animal health and performance, and chitosan oligosaccharides (COS) exhibits antioxidant and anti-inflammatory properties. The aim of this study was to evaluate the effects of COS alleviation of oxidative stress and inflammatory response in heat-stressed rats. The results indicated heat stress decreased (p < 0.05) growth performance; the relative weight of spleen and kidney; and the level of antioxidant enzymes and IL-10 in liver, spleen, and kidney, while it increased (p < 0.05) the MDA and inflammatory cytokines concentration. Dietary COS supplementation enhanced (p < 0.05) ADG, the relative weight of spleen and kidney, and the level of antioxidant enzymes and IL-10 in liver, spleen, and kidney. Collectively, COS was beneficial to heat-stressed rats by alleviating oxidative damage and inflammatory response. Abstract Chitosan oligosaccharides (COS) exhibits antioxidant and anti-inflammatory properties. The aim of this study was to evaluate the effects of COS on antioxidant system and inflammatory response in heat-stressed rats. A total of 30 male rats were randomly divided to three groups and reared at either 24 °C or 35 °C for 4 h/d for this 7-day experiment: CON, control group with basal diet; HS, heat stress group with basal diet; HSC, heat stress with 200mg/kg COS supplementation. Compared with the CON group, HS significantly decreased (p < 0.05) average daily gain (ADG); average daily feed intake (ADFI); the relative weight of spleen and kidney; the level of liver CAT, GSH-Px, T-AOC, and IL-10; spleen SOD, GSH-Px, GSH, and IL-10; and kidney SOD, GSH-Px, T-AOC, and IL-10, while significantly increased the MDA concentration in liver, spleen, and kidney; the liver IL-1β concentration; and spleen and kidney IL-6 and TNF-α concentration. In addition, dietary COS supplementation significantly improved (p < 0.05) ADG; the relative weight of spleen and kidney; the level of liver GSH-Px, spleen GSH-Px, GSH, and IL-10; and kidney GSH-Px, while significantly decreased (p < 0.05) liver IL-1β concentration under heat stress condition. Collectively, COS was beneficial to heat-stressed rats by alleviating oxidative damage and inflammatory response.
Collapse
|
46
|
The Bioprotective Effects of Polyphenols on Metabolic Syndrome against Oxidative Stress: Evidences and Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6713194. [PMID: 31885810 PMCID: PMC6914975 DOI: 10.1155/2019/6713194] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/11/2019] [Accepted: 11/01/2019] [Indexed: 12/25/2022]
Abstract
Polyphenols are the general designation of various kinds of phytochemicals, mainly classified as flavonoids and nonflavonoids. Polyphenolic compounds have been confirmed to exhibit numerous bioactivities and potential health benefits both in vivo and in vitro. Dietary polyphenols have been shown to significantly alleviate several manifestations of metabolic syndrome, namely, central obesity, hypertension, dyslipidemia, and high blood sugar. This review is aimed at discussing the bioprotective effects and related molecular mechanisms of polyphenols, mainly by increasing antioxidant capacity or oxygen scavenging capacity. Polyphenols can exert their antioxidative activity by balancing the organic oxidoreductase enzyme system, regulating antioxidant responsive signaling pathways, and restoring mitochondrial function. These data are helpful for providing new insights into the potential biological effects of polyphenolic compounds and the development of future antioxidant therapeutics.
Collapse
|
47
|
Recent Updates in Pharmacological Properties of Chitooligosaccharides. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4568039. [PMID: 31781615 PMCID: PMC6875261 DOI: 10.1155/2019/4568039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/26/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
Chemical structures derived from marine foods are highly diverse and pharmacologically promising. In particular, chitooligosaccharides (COS) present a safe pharmacokinetic profile and a great source of new bioactive polymers. This review describes the antioxidant, anti-inflammatory, and antidiabetic properties of COS from recent publications. Thus, COS constitute an effective agent against oxidative stress, cellular damage, and inflammatory pathogenesis. The mechanisms of action and targeted therapeutic pathways of COS are summarized and discussed. COS may act as antioxidants via their radical scavenging activity and by decreasing oxidative stress markers. The mechanism of COS antidiabetic effect is characterized by an acceleration of pancreatic islets proliferation, an increase in insulin secretion and sensitivity, a reduction of postprandial glucose, and an improvement of glucose uptake. COS upregulate the GLUT2 and inhibit digestive enzyme and glucose transporters. Furthermore, they resulted in reduction of gluconeogenesis and promotion of glucose conversion. On the other hand, the COS decrease inflammatory mediators, suppress the activation of NF-κB, increase the phosphorylation of kinase, and stimulate the proliferation of lymphocytes. Overall, this review brings evidence from experimental data about protective effect of COS.
Collapse
|
48
|
Moon SW, Ahn CB, Oh Y, Je JY. Lotus (Nelumbo nucifera) seed protein isolate exerts anti-inflammatory and antioxidant effects in LPS-stimulated RAW264.7 macrophages via inhibiting NF-κB and MAPK pathways, and upregulating catalase activity. Int J Biol Macromol 2019; 134:791-797. [DOI: 10.1016/j.ijbiomac.2019.05.094] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
49
|
Xu D, Liao S, Li P, Zhang Q, Lv Y, Fu X, Yang M, Wang J, Kong L. Metabolomics Coupled with Transcriptomics Approach Deciphering Age Relevance in Sepsis. Aging Dis 2019; 10:854-870. [PMID: 31440390 PMCID: PMC6675524 DOI: 10.14336/ad.2018.1027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/27/2018] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a severe disease frequently occurred in the Intenisive Care Unit (ICU), which has a very high morbidity and mortality, especially in patients aged over 65 years. Owing to the aging effect and the ensuing deterioration of body function, the elder patients may have atypical responses to sepsis. Diagnosis and pathogenesis of sepsis in this population are thus difficult, which hindered effective treatment and management in clinic. To investigated age effects on sepsis, 158 elderly septic patients and 71 non-septic elderly participants were enrolled, and their plasma samples were collected for transcriptomics (RNA-seq) and metabolomics (NMR and GC-MS) analyses, which are both increasingly being utilized to discover key molecular changes and potential biomarkers for various diseases. Protein-protein interaction (PPI) analysis was subsequently performed to assist cross-platform integration. Real time polymerase chain reaction (RT-PCR) was used for validation of RNA-seq results. For further understanding of the mechanisms, cecal ligation and puncture (CLP) experiment was performed both in young and middle-aged rats, which were subjected to NMR-based metabolomics study and validated for several key inflammation pathways by western blot. Comprehensive analysis of data from the two omics approaches provides a systematic perspective on dysregulated pathways that could facilitate the development of therapy and biomarkers for elderly sepsis. Additionally, the metabolites of lactate, arginine, histamine, tyrosine, glutamate and glucose were shown to be highly specific and sensitive in distinguishing septic patients from healthy controls. Significant increases of arginine, trimethylamine N-oxide and allantoin characterized elderly patient incurred sepsis. Further analytical and biological validations in different subpopulations of septic patients should be carried out, allowing accurate diagnostics and precise treatment of sepsis in clinic.
Collapse
Affiliation(s)
- Dingqiao Xu
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shanting Liao
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Pei Li
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qian Zhang
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yan Lv
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaowei Fu
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Minghua Yang
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Junsong Wang
- 2Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, China
| | - Lingyi Kong
- 1Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
50
|
Kim HN, Park GH, Park SB, Kim JD, Eo HJ, Son HJ, Song JH, Jeong JB. Sageretia thea Inhibits Inflammation through Suppression of NF-κB and MAPK and Activation of Nrf2/HO-1 Signaling Pathways in RAW264.7 Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:385-403. [DOI: 10.1142/s0192415x19500198] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sageretia thea (S. thea) commonly known as Chinese sweet plum or Chinese bird plum has been used for treating hepatitis and fevers in Korea and China. S. thea has been reported to exert anti-oxidant, anticancer and anti-human immunodeficiency virus activity. However, there is little study on the anti-inflammatory activity of S. thea. Thus, we evaluated the anti-inflammatory effect of extracts of leaves (ST-L) and branches (ST-B) from Sageretia thea in LPS-stimulated RAW264.7 cells. ST-L and ST-B significantly inhibited the production of the pro-inflammatory mediators such as NO, iNOS, COX-2, IL-1[Formula: see text] and IL-6 in LPS-stimulated RAW264.7 cells. ST-L and ST-B blocked LPS-induced degradation of I[Formula: see text]B-[Formula: see text] and nuclear accumulation of p65, which resulted in the inhibition of NF-[Formula: see text]B activation in RAW264.7 cells. ST-L and ST-B also attenuated the phosphorylation of ERK1/2, p38 and JNK in LPS-stimulated RAW264.7 cells. In addition, ST-L and ST-B increased HO-1 expression in RAW264.7 cells, and the inhibition of HO-1 by ZnPP reduced the inhibitory effect of ST-L and ST-B against LPS-induced NO production in RAW264.7 cells. Inhibition of p38 activation and ROS elimination attenuated HO-1 expression by ST-L and ST-B, and ROS elimination inhibited p38 activation induced by ST-L and ST-B. ST-L and ST-B dramatically induced nuclear accumulation of Nrf2, but this was significantly reversed by the inhibition of p38 activation and ROS elimination. Collectively, our results suggest that ST-L and ST-B exerts potential anti-inflammatory activity by suppressing NF-[Formula: see text]B and MAPK signaling activation, and activating HO-1 expression through the nuclear accumulation of Nrf2 via ROS-dependent p38 activation. These findings suggest that ST-L and ST-B may have great potential for the development of anti-inflammatory drug to treat acute and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Ha Na Kim
- Department of Medicinal Plant Resources, National Institute of Forest Science, Yongju 36040, Republic of Korea
| | - Gwang Hun Park
- Agricultural Science and Technology Research Institute, Andong National University, Andong 36729, Republic of Korea
| | - Su Bin Park
- Department of Medicinal Plant Resources, National Institute of Forest Science, Yongju 36040, Republic of Korea
| | - Jeong Dong Kim
- Department of Medicinal Plant Resources, National Institute of Forest Science, Yongju 36040, Republic of Korea
| | - Hyun Ji Eo
- Agricultural Science and Technology Research Institute, Andong National University, Andong 36729, Republic of Korea
| | - Ho-Jun Son
- Agricultural Science and Technology Research Institute, Andong National University, Andong 36729, Republic of Korea
| | - Jeong Ho Song
- Agricultural Science and Technology Research Institute, Andong National University, Andong 36729, Republic of Korea
| | - Jin Boo Jeong
- Department of Medicinal Plant Resources, National Institute of Forest Science, Yongju 36040, Republic of Korea
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yongju 36040, Republic of Korea
| |
Collapse
|