1
|
Ding C, Wu Y, Zhan C, Naseem A, Chen L, Li H, Yang B, Liu Y. Research progress on the role and inhibitors of Keap1 signaling pathway in inflammation. Int Immunopharmacol 2024; 141:112853. [PMID: 39159555 DOI: 10.1016/j.intimp.2024.112853] [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: 06/18/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
Inflammation is a protective mechanism against endogenous and exogenous pathogens. It is a typical feature of numerous chronic diseases and their complications. Keap1 is an essential target in oxidative stress and inflammatory diseases. Among them, the Keap1-Nrf2-ARE pathway (including Keap1-Nrf2-HO-1) is the most significant pathway of Keap1 targets, which participates in the control of inflammation in multiple organs (including renal inflammation, lung inflammation, liver inflammation, neuroinflammation, etc.). Identifying new Keap1 inhibitors is crucial for new drug discovery. However, most drugs have specificity issues as they covalently bind to cysteine residues of Keap1, causing off-target effects. Therefore, direct inhibition of Keap1-Nrf2 PPIs is a new research idea. Through non-electrophilic and non-covalent binding, its inhibitors have better specificity and ability to activate Nrf2, and targeting therapy against Keap1-Nrf2 PPIs has become a new method for drug development in chronic diseases. This review summarizes the members and downstream genes of the Keap1-related pathway and their roles in inflammatory disease models. In addition, we summarize all the research progress of anti-inflammatory drugs targeting Keap1 from 2010 to 2024, mainly describing their biological functions, molecular mechanisms of action, and therapeutic roles in inflammatory diseases.
Collapse
Affiliation(s)
- Chao Ding
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Ying Wu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| | - Chaochao Zhan
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Anam Naseem
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Yan Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| |
Collapse
|
2
|
Ahmed RF, Elbaset MA, Farouk H, Shabana ME, Ahmed-Farid OA, Nasr M. Nanoliposomal amino acids counteracting protein malnutrition induced hematopoietic and hepatic complications. J Drug Deliv Sci Technol 2023; 89:105051. [DOI: 10.1016/j.jddst.2023.105051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Liu M, Wen H, Zuo L, Song X, Geng Z, Ge S, Ge Y, Wu R, Chen S, Yu C, Gao Y. Bryostatin-1 attenuates intestinal ischemia/reperfusion-induced intestinal barrier dysfunction, inflammation, and oxidative stress via activation of Nrf2/HO-1 signaling. FASEB J 2023; 37:e22948. [PMID: 37130016 DOI: 10.1096/fj.202201540r] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 03/27/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Bryostatin-1 (Bryo-1) exerts antioxidative stress effects in multiple diseases, and we confirmed that it improves intestinal barrier dysfunction in experimental colitis. Nevertheless, there are few reports on its action on intestinal ischemia/reperfusion (I/R). In this study, we mainly explored the effect of Bryo-1 on intestinal I/R injury and determined the mechanism. C57BL/6J mice underwent temporary superior mesenteric artery (SMA) obturation to induce I/R, on the contrary, Caco-2 cells suffered to oxygen and glucose deprivation/reperfusion (OGD/R) to establish the in vitro model. RAW264.7 cells were stimulated with LPS to induce macrophage inflammation. The drug gradient experiment was used to demonstrate in vivo and in vitro models. Bryo-1 ameliorated the intestinal I/R-induced injury of multiple organs and epithelial cells. It also alleviated intestinal I/R-induced barrier disruption of intestines according to the histology, intestinal permeability, intestinal bacterial translocation rates, and tight junction protein expression results. Bryo-1 significantly inhibited oxidative stress damages and inflammation, which may contribute to the restoration of intestinal barrier function. Further, Bryo-1 significantly activated Nrf2/HO-1 signaling in vivo. However, the deletion of Nrf2 in Caco-2 and RAW264.7 cells attenuated the protective functions of Bryo-1 and significantly abolished the anti-inflammatory effect of Bryo-1 on LPS-induced macrophage inflammation. Bryo-1 protects intestines against I/R-induced injury. It is associated with intestinal barrier protection, as well as inhibition of inflammation and oxidative stress partly through Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Mulin Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Hexin Wen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijun Geng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Sitang Ge
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yuanyuan Ge
- Department of Colorectal Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Rong Wu
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, China
| | - Shiyuan Chen
- Department of Vascular Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Chaowen Yu
- Department of Vascular Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yong Gao
- Department of Vascular Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
4
|
Ahmed RF, Nasr M, Abd Elbaset M, Hussein AF, Ahmed-Farid OAH, Shafee N, Shabana ME. Combating hematopoietic and hepatocellular abnormalities resulting from administration of cisplatin: Role of liver targeted glycyrrhetinic acid nanoliposomes loaded with amino acids. Pharm Dev Technol 2022; 27:925-941. [PMID: 36168910 DOI: 10.1080/10837450.2022.2129687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The effectiveness of cisplatin in cancer treatment renders its use vital to clinicians. However, the accompanying side effects as cachexia, emesis and liver damage necessitate the use of a dietary supplement which is capable of hindering such undesirable complications. The branched chain amino acids as well as glutamine and arginine have been proven to be effective nutritional co-adjuvant therapeutic agents. Furthermore, new pharmaceutical approaches encompass designing organ-targeted nanoformulations to increase the medicinal efficacy. Therefore, the aim of the present study was to investigate the beneficial effects of liver-targeted amino acids-loaded nanoliposomes in counteracting the adverse hematopoietic and hepatic complications associated with cisplatin. Results revealed the use of the combination of two nanoliposomal formulations (one loading leucine + isolecuine + valine, and the other loading glutamine and arginine) given orally at a dose of 200 mg/kg for twelve days was effective against cisplatin-induced toxicities represented by improvement in the complete blood picture parameters, decrease in the serum hepatic enzymes levels, amelioration of the hepatic oxidative stress and cellular energy imbalance along with reduction in the histopathological abnormalities. It can be concluded that amino acids loaded nanoliposomes could be considered a new strategy in preventing cisplatin's adverse effects.
Collapse
Affiliation(s)
- Rania F Ahmed
- Department of Pharmacology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Marawan Abd Elbaset
- Department of Pharmacology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Alyaa F Hussein
- Department of Pharmacology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Omar A H Ahmed-Farid
- Department of Physiology, National Organization for Drug Control and Research, 12553, Giza, Egypt
| | - Nermin Shafee
- Department of Pathology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Marwa E Shabana
- Department of Pathology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| |
Collapse
|
5
|
Dengler F, Sternberg F, Grages M, Kästner SBR, Verhaar N. Adaptive mechanisms in no flow vs. low flow ischemia in equine jejunum epithelium: Different paths to the same destination. Front Vet Sci 2022; 9:947482. [PMID: 36157182 PMCID: PMC9493374 DOI: 10.3389/fvets.2022.947482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/17/2022] [Indexed: 01/18/2023] Open
Abstract
Intestinal ischemia reperfusion injury (IRI) is a frequent complication of equine colic. Several mechanisms may be involved in adaptation of the intestinal epithelium to IRI and might infer therapeutic potential, including hypoxia-inducible factor (HIF) 1α, AMP-activated protein kinase (AMPK), nuclear factor-erythroid 2-related factor 2 (NRF2), and induction of autophagy. However, the mechanisms supporting adaptation and thus cellular survival are not completely understood yet. We investigated the activation of specific adaptation mechanisms in both no and low flow ischemia and reperfusion simulated in equine jejunum epithelium in vivo. We found an activation of HIF1α in no and low flow ischemia as indicated by increased levels of HIF1α target genes and phosphorylation of AMPKα tended to increase during ischemia. Furthermore, the protein expression of the autophagy marker LC3B in combination with decreased expression of nuclear-encoded mitochondrial genes indicates an increased rate of mitophagy in equine intestinal IRI, possibly preventing damage by mitochondria-derived reactive oxygen species (ROS). Interestingly, ROS levels were increased only shortly after the onset of low flow ischemia, which may be explained by an increased antioxidative defense, although NFR2 was not activated in this setup. In conclusion, we could demonstrate that a variety of adaptation mechanisms manipulating different aspects of cellular homeostasis are activated in IRI irrespective of the ischemia model, and that mitophagy might be an important factor for epithelial survival following small intestinal ischemia in horses that should be investigated further.
Collapse
Affiliation(s)
- Franziska Dengler
- Department of Biochemical Sciences, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
- *Correspondence: Franziska Dengler
| | - Felix Sternberg
- Department of Biochemical Sciences, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Marei Grages
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sabine BR Kästner
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole Verhaar
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
6
|
Shen J, Zhan Y, He Q, Deng Q, Li K, Wen S, Huang W. Remifentanil Promotes PDIA3 Expression by Activating p38MAPK to Inhibit Intestinal Ischemia/Reperfusion-Induced Oxidative and Endoplasmic Reticulum Stress. Front Cell Dev Biol 2022; 10:818513. [PMID: 35155431 PMCID: PMC8826554 DOI: 10.3389/fcell.2022.818513] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Remifentanil protects against intestinal ischemia/reperfusion (I/R) injury; however, its exact mechanism remains to be elucidated. The objective of this study was to investigate the underlying molecular mechanism of remifentanil in intestinal I/R injury in mice.Methods: We evaluated the intestine-protective effect of remifentanil in adult male mice with 45 min superior mesenteric artery occlusion followed by 4 h reperfusion by determining the following: intestinal Chiu’s scores, diamine oxidase, and intestinal fatty acid binding protein in serum; the apoptotic index, lipid peroxidation product malondialdehyde (MDA), and superoxide dismutase (SOD) activity in the intestinal mucosa; and the intestinal mRNA and protein expressions of Bip, CHOP, caspase-12, and cleaved caspase-3, reflecting endoplasmic reticulum (ER) stress. Furthermore, conditional knockout mice, in which the protein disulfide isomerase A3 (PDIA3) gene was deleted from the intestinal epithelium, and SB203580 (a selective p38MAPK inhibitor) were used to determine the role of PDIA3 and p38MAPK in I/R progression and intestinal protection by remifentanil.Results: Our data showed that intestinal I/R induced obvious oxidative stress and endoplasmic reticulum stress–related cell apoptosis, as evidenced by an increase in the intestinal mucosal malondialdehyde, a decrease in the intestinal mucosal SOD, and an increase in the apoptotic index and the mRNA and protein expression of Bip, CHOP, caspase-12, and cleaved caspase-3. Remifentanil significantly improved these changes. Moreover, the deletion of intestinal epithelium PDIA3 blocked the protective effects of remifentanil. SB203580 also abolished the intestinal protection of remifentanil and downregulated the mRNA and protein expression of PDIA3.Conclusion: Remifentanil appears to act via p38MAPK to protect the small intestine from intestinal I/R injury by its PDIA3-mediated antioxidant and anti-ER stress properties.
Collapse
Affiliation(s)
| | | | | | | | | | - Shihong Wen
- *Correspondence: Shihong Wen, ; Wenqi Huang,
| | - Wenqi Huang
- *Correspondence: Shihong Wen, ; Wenqi Huang,
| |
Collapse
|
7
|
Fernandes JC, Schemitt EG, Da Silva J, Marroni NP, Lima A, Ferreira RB. Combination of Trans-Resveratrol and ε-Viniferin Induces a Hepatoprotective Effect in Rats with Severe Acute Liver Failure via Reduction of Oxidative Stress and MMP-9 Expression. Nutrients 2021; 13:nu13113677. [PMID: 34835933 PMCID: PMC8622851 DOI: 10.3390/nu13113677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 01/11/2023] Open
Abstract
Stilbenes are a major grapevine class of phenolic compounds, known for their biological activities, including anti-inflammatory and antioxidant, but never studied in combination. We aimed to evaluate the effect of trans-resveratrol + ε-viniferin as an antioxidant mixture and its role in inflammatory development an in vivo model of severe acute liver failure induced with TAA. Trans-resveratrol + trans-ε-viniferin (5 mg/kg each) was administered to Wistar rats. Resveratrol + ε-viniferin significantly decreased TBARS and SOD activity and restored CAT and GST activities in the treated group. This stilbene combination reduced the expression of TNFα, iNOS, and COX-2, and inhibited MMP-9. The combination of resveratrol + ε-viniferin had a hepatoprotective effect, reducing DNA damage, exhibiting a protective role on the antioxidant pathway by altering SOD, CAT, and GST activities; by downregulating TNFα, COX-2, and iNOS; and upregulating IL-10. Our results suggested that adding viniferin to resveratrol may be more effective in hepatoprotection than resveratrol alone, opening a new perspective on using this stilbene combination in functional diets.
Collapse
Affiliation(s)
- João C. Fernandes
- Linking Landscape, Environment, Agriculture and Food (LEAF), Universidade de Lisboa, Instituto Superior de Agronomia, Tapada da Ajuda, 1349-017 Lisbon, Portugal; (A.L.); (R.B.F.)
- Correspondence: (J.C.F.); Tel.: +351-213-653-414
| | - Elizângela G. Schemitt
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040-060, Brazil; (E.G.S.); (N.P.M.)
| | - Juliana Da Silva
- Laboratório de Genética Toxicológica, Universidade Luterana do Brasil, Canoas 92425-900, Brazil;
| | - Norma P. Marroni
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040-060, Brazil; (E.G.S.); (N.P.M.)
| | - Ana Lima
- Linking Landscape, Environment, Agriculture and Food (LEAF), Universidade de Lisboa, Instituto Superior de Agronomia, Tapada da Ajuda, 1349-017 Lisbon, Portugal; (A.L.); (R.B.F.)
- Faculty of Veterinary Medicine, Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisbon, Portugal
| | - Ricardo B. Ferreira
- Linking Landscape, Environment, Agriculture and Food (LEAF), Universidade de Lisboa, Instituto Superior de Agronomia, Tapada da Ajuda, 1349-017 Lisbon, Portugal; (A.L.); (R.B.F.)
| |
Collapse
|
8
|
Eryilmaz S, Turkyilmaz Z, Karabulut R, Gulburun MA, Poyraz A, Gulbahar O, Arslan B, Sonmez K. The effects of hydrogen-rich saline solution on intestinal anastomosis performed after intestinal ischemia reperfusion injury. J Pediatr Surg 2020; 55:1574-1578. [PMID: 31466816 DOI: 10.1016/j.jpedsurg.2019.07.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/02/2019] [Accepted: 07/19/2019] [Indexed: 01/31/2023]
Abstract
AIM We investigated the effects of hydrogen-rich saline solution (HRSS) on intestinal anastomosis performed after intestinal ischemia reperfusion injury (IRI). MATERIALS AND METHODS Thirty Wistar albino female rats were randomly divided into five groups. Only laparotomy was performed in the Sham group. In the other four groups, an intestinal IRI was performed for 45 min by clamping the superior mesenteric artery. After intestinal IRI, anastomosis was performed by cutting the intestine from the proximal 15 cm of the ileocecal valve at the first and 24th hours. HRSS was given intraperitoneally 5 ml/kg before reperfusion and for four more days in the HRSS1 and HRSS24groups, while no treatment was given to the I/R1 and I/R24 groups. After 5 days, all groups underwent relaparotomy. The anastomotic bursting pressures were measured in all groups, except the Sham group. The tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), myeloperoxidase (MPO) and malondialdehyde (MDA) levels were measured in the tissues taken from the anastomosis line. The tissue sections were evaluated histopathologically and the apoptosis index was determined by applying the TUNEL method. The results were analyzed one-way analysis of variance (ANOVA) and Pearson's chi-squared test. RESULTS Although the MPO, MDA, IL-6 and TNF-α tissue values were not statistically significant among the groups, the degree of tissue damage and apoptosis levels were lower and the anastomotic bursting pressures values were higher in the HRSS1 and HRSS24 groups compared to the I/R1 and I/R24 groups. CONCLUSION HRSS is effective in reducing the intestinal damage caused by an IRI: HRSS has the potential to reduce the detrimental effects of intestinal anastomosis performed after an intestinal IRI.
Collapse
Affiliation(s)
- Sibel Eryilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Zafer Turkyilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ramazan Karabulut
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey.
| | - Merve Altin Gulburun
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Aylar Poyraz
- Department of Pediatric Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ozlem Gulbahar
- Department of Pediatric Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Burak Arslan
- Department of Pediatric Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Kaan Sonmez
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
9
|
Costanzo M, Caterino M, Cevenini A, Jung V, Chhuon C, Lipecka J, Fedele R, Guerrera IC, Ruoppolo M. Proteomics Reveals that Methylmalonyl-CoA Mutase Modulates Cell Architecture and Increases Susceptibility to Stress. Int J Mol Sci 2020; 21:ijms21144998. [PMID: 32679819 PMCID: PMC7403994 DOI: 10.3390/ijms21144998] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Methylmalonic acidemia (MMA) is a rare inborn error of metabolism caused by deficiency of the methylmalonyl-CoA mutase (MUT) enzyme. Downstream MUT deficiency, methylmalonic acid accumulates together with toxic metabolites from propionyl-CoA and other compounds upstream of the block in the enzyme pathway. The presentation is with life-threatening acidosis, respiratory distress, brain disturbance, hyperammonemia, and ketosis. Survivors develop poorly understood multi-organ damage, notably to the brain and kidneys. The HEK 293 cell line was engineered by CRISPR/Cas9 technology to knock out the MUT gene (MUT-KO). Shotgun label-free quantitative proteomics and bioinformatics analyses revealed potential damaging biological processes in MUT-deficient cells. MUT-KO induced alteration of cellular architecture and morphology, and ROS overproduction. We found the alteration of proteins involved in cytoskeleton and cell adhesion organization, cell trafficking, mitochondrial, and oxidative processes, as validated by the regulation of VIM, EXT2, SDC2, FN1, GLUL, and CHD1. Additionally, a cell model of MUT-rescuing was developed in order to control the specificity of MUT-KO effects. Globally, the proteomic landscape of MUT-KO suggests the cell model to have an increased susceptibility to propionate- and H2O2-induced stress through an impairment of the mitochondrial functionality and unbalances in the oxidation-reduction processes.
Collapse
Affiliation(s)
- Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy;
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy;
| | - Armando Cevenini
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy;
| | - Vincent Jung
- Proteomics Platform Necker, Université de Paris—Structure Fédérative de Recherche Necker, Inserm US24/CNRS UMS3633, 75015 Paris, France; (V.J.); (C.C.); (J.L.)
| | - Cerina Chhuon
- Proteomics Platform Necker, Université de Paris—Structure Fédérative de Recherche Necker, Inserm US24/CNRS UMS3633, 75015 Paris, France; (V.J.); (C.C.); (J.L.)
| | - Joanna Lipecka
- Proteomics Platform Necker, Université de Paris—Structure Fédérative de Recherche Necker, Inserm US24/CNRS UMS3633, 75015 Paris, France; (V.J.); (C.C.); (J.L.)
| | - Roberta Fedele
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy;
| | - Ida Chiara Guerrera
- Proteomics Platform Necker, Université de Paris—Structure Fédérative de Recherche Necker, Inserm US24/CNRS UMS3633, 75015 Paris, France; (V.J.); (C.C.); (J.L.)
- Correspondence: (I.C.G.); (M.R.); Tel.: +39-081-373-7850 (M.R.)
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (M.C.); (M.C.); (A.C.)
- CEINGE—Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy;
- Correspondence: (I.C.G.); (M.R.); Tel.: +39-081-373-7850 (M.R.)
| |
Collapse
|
10
|
KARAKAŞ S, KAYA C, GÜRASLAN H, SAKIZ D, SÜZEN ÇAYPINAR S, CENGİZ H, EKİN M, YAŞAR L. Effect of metformin and detorsion treatment on serum anti-Müllerian hormonelevels and ovarian histopathology in a rat ovarian torsion model. Turk J Med Sci 2020; 50:455-463. [PMID: 31999406 PMCID: PMC7164755 DOI: 10.3906/sag-1803-196] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 01/29/2020] [Indexed: 12/31/2022] Open
Abstract
Background/aim Adnexal torsion is a common gynaecological emergency, and considered to be a problem mostly in reproductive-age women. To evaluatethe effect of metformin and detorsion treatment on reducing ovarian reserve in an ovarian torsion model. Materials and methods Twenty-four nonpregnant, Wistar Hannover rats were included in the study. Animals were divided into 3 groups: the control group, the detorsion only group, and the metformin + detorsion group. The first group received only laparotomy. In the second group, ovaries were fixed to the abdominal wall after performing 360° ovarian torsion, followed by detorsion after a 3-h period of ischemia. The third group underwent the same torsion and detorsion procedures as the second group, and received 50 mg/kg metformin by gavage for 14 days. Ovarian damage scores, follicle counts, and AMH levels were evaluated. Results The total damage score was significantly increased in the detorsion only group compared to the metformin+detorsion and control groups. Pre-operative/post-operative AMH decreases were statistically significant in negative direction in the detorsion only group when compared to the metformin+detorsion and control groups (P = 0.001). Conclusion Metformin+detorsion treatment may be effective in protecting the ovarian reserve after ovarian torsion.
Collapse
Affiliation(s)
- Sema KARAKAŞ
- Department of Obstetrics and Gynecology, University of Health Sciences,Gaziosmanpaşa Taksim Training and Research Hospital, İstanbulTurkey
| | - Cihan KAYA
- Department of Obstetrics and Gynecology, University of Health Sciences,Bakirköy Dr. Sadi Konuk Training and Research Hospital, İstanbulTurkey
| | - Hakan GÜRASLAN
- Department of Obstetrics and Gynecology, University of Health Sciences,Bakirköy Dr. Sadi Konuk Training and Research Hospital, İstanbulTurkey
| | - Damlanur SAKIZ
- Department of Pathology, University of Health Sciences,Bakirköy Dr Sadi Konuk Training and Research Hospital, İstanbulTurkey
| | - Sema SÜZEN ÇAYPINAR
- Department of Obstetrics and Gynecology, University of Health Sciences,Bakirköy Dr. Sadi Konuk Training and Research Hospital, İstanbulTurkey
| | - Hüseyin CENGİZ
- Department of Obstetrics and Gynecology, University of Health Sciences,Bakirköy Dr. Sadi Konuk Training and Research Hospital, İstanbulTurkey
| | - Murat EKİN
- Department of Obstetrics and Gynecology, University of Health Sciences,Bakirköy Dr. Sadi Konuk Training and Research Hospital, İstanbulTurkey
| | - Levent YAŞAR
- Department of Obstetrics and Gynecology, University of Health Sciences,Bakirköy Dr. Sadi Konuk Training and Research Hospital, İstanbulTurkey
| |
Collapse
|
11
|
Xu H, Liu G, Gu H, Wang J, Li Y. Glutamine protects intestine against ischemia-reperfusion injury by alleviating endoplasmic reticulum stress induced apoptosis in rats. Acta Cir Bras 2020; 35:e202000104. [PMID: 32159588 PMCID: PMC7065443 DOI: 10.1590/s0102-865020200010000004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/08/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose Glutamine, as an essential part of enteral nutrition and parenteral nutrition agent, has been widely recognized to be a kind of important intestinal mucosa protectant in clinical practice and experimental research. However, the mechanisms of its protective effects are still not fully understand. Consequently, this study aimed to explore the potential mechanism of glutamine on ischemia-reperfusion (I/R) injury induced endoplasmic reticulum (ER) stress in intestine. Methods An experimental model of intestinal I/R in rats was established by 1 hour occlusion of the superior mesenteric artery followed by 3 hours of reperfusion. Morphologic changes of intestinal mucosa, apoptosis of epithelial cells, and expression of intestinal Grp78, Gadd153, Caspase-12, ATF4, PERK phosphorylation (P-PERK) and elF2αphosphorylation(P-elF2α) were determined. Results After I/R, the apoptotic index of intestinal mucosa epithelial cells observably increased with notable necrosis of intestinal mucosa, and the expressions of Grp78, Gadd153, Caspase-12, ATF4, P-PERK and P-elF2αall were increased. However, treatment with glutamine could significantly relieve intestinal I/R injury and apoptosis index. Moreover, glutamine could clearly up-regulate the expression of Grp78, restrain P-PERK and P-elF2α, and reduce ATF4, Gadd153 and Caspase-12 expressions. Conclusion Glutamine may be involved in alleviating ER stress induced intestinal mucosa cells apoptosis.
Collapse
Affiliation(s)
- Hao Xu
- People’s Hospital of Kaizhou District, China
| | | | | | | | - Yang Li
- Chongqing Medical University, China
| |
Collapse
|
12
|
Liu Z, Wu Q, Jiao C, Cheng B, Zhu D, Ma Y, Li Y, Li W. Effects of Glutamine on the Mucosal Structure and Immune Cells in the Intestines of Broiler Chickens Challenged with Salmonella Enteritidis. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2020. [DOI: 10.1590/1806-9061-2020-1270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Z Liu
- Henan University of Science and Technology, PR China
| | - Q Wu
- Henan University of Science and Technology, PR China
| | - C Jiao
- Henan University of Science and Technology, PR China
| | - B Cheng
- Henan University of Science and Technology, PR China
| | - D Zhu
- Henan University of Science and Technology, PR China
| | - Y Ma
- Henan University of Science and Technology, PR China
| | - Y Li
- Henan University of Science and Technology, PR China
| | - W Li
- Henan University of Science and Technology, PR China
| |
Collapse
|
13
|
Hu H, Dai S, Li J, Wen A, Bai X. Glutamine improves heat stress-induced oxidative damage in the broiler thigh muscle by activating the nuclear factor erythroid 2-related 2/Kelch-like ECH-associated protein 1 signaling pathway. Poult Sci 2019; 99:1454-1461. [PMID: 32115031 PMCID: PMC7587763 DOI: 10.1016/j.psj.2019.11.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/07/2019] [Accepted: 11/04/2019] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to evaluate the effect of glutamine (Gln) on modulating heat stress–induced oxidative damage in the broiler thigh muscle through nuclear factor erythroid 2–related 2/Kelch-like ECH-associated protein 1 (Nrf2-Keap1) pathway. Three-hundred 22-day-old Arbor Acres broilers were reallocated into 5 groups: a control group (24 °C) fed with basal diet and 4 heat stress (HS) groups (34 °C for 8 h/D) fed with basal diet containing 0, 0.5, 1.0, and 1.5% Gln. This experiment lasted 21 D. Heat stress decreased (P < 0.05) pH, redness, and Gln levels, and increased (P < 0.05) luminance, water loss rate, and cooking loss (CL) values of the thigh meat. Compared with the HS group, supplementation with 1.5% Gln increased (P < 0.05) pH, redness, and Gln levels, but decreased (P < 0.05) luminance and CL values in the thigh meat. There were significant decreases (P < 0.05) in glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), total antioxidant capacity (T-AOC), and Nrf2 levels, but significant increases (P < 0.05) in the malondialdehyde (MDA) and Keap1 levels of the thigh muscle after HS treatment. Compared with the HS group, supplementation with 1.0, and 1.5% Gln decreased (P < 0.05) MDA and Keap1 levels; supplementation with 1.5% Gln increased (P < 0.05) GSH, GSH-Px, T-AOC, CAT, SOD, and Nrf2 levels in the thigh muscle of heat-stressed broilers. Furthermore, HS decreased (P < 0.05) Nrf2, SOD, CAT, and GSH-Px mRNA expression levels, but increased (P < 0.05) Keap1 mRNA level in the thigh muscle of broiler. Dietary supplementation with 1.5% Gln increased (P < 0.05) Nrf2, GSH-Px, CAT, and SOD mRNA expression levels, but decreased (P < 0.05) Keap1 mRNA level in the thigh muscle of heat-stressed broilers. In conclusion, dietary Gln improved the resistance of heat-stressed broiler muscles to oxidative damage possibly through reversing the muscle Gln level and inducing the expression of the Nrf2-Keap1 pathway.
Collapse
Affiliation(s)
- Hong Hu
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100 People's Republic of China
| | - Sifa Dai
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100 People's Republic of China; Department of Pharmaceutical and Life Sciences, Jiujiang University, Jiujiang 332005, People's Republic of China
| | - Jiaqi Li
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100 People's Republic of China
| | - Aiyou Wen
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100 People's Republic of China
| | - Xi Bai
- College of Animal Science, Anhui Science and Technology University, Fengyang, 233100 People's Republic of China.
| |
Collapse
|
14
|
Huang L, Yin P, Liu F, Liu Y, Liu Y, Xia Z. Protective effects of L-arginine on the intestinal epithelial barrier under heat stress conditions in rats and IEC-6 cell line. J Anim Physiol Anim Nutr (Berl) 2019; 104:385-396. [PMID: 31709652 DOI: 10.1111/jpn.13246] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 09/03/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
Heat stress (HS) and the associated restricted blood flow to the intestine have been proven to destroy intestinal integrity. Considering the beneficial properties of L-arginine on gut function, we investigated the protective effects of L-arginine on the intestine under HS conditions. In vivo, the serum cortisol level and the rectal temperature increased in response to HS. Under HS, the intestinal damage showed obvious morphological changes. Furthermore, HS decreased the mRNA and protein expression levels of Nurr1, ZO-1, occludin, claudin-6 and E-cadherin, increased the mRNA expression of NF-κB and IL-1β, and increased the protein expression of cleaved caspase-3. In contrast, L-arginine supplementation maintained intestinal integrity and increased the villus/crypt ratio. L-arginine also suppressed the expression of inflammation-related genes and the protein expression of cleaved caspase-3, whereas it upregulated the mRNA and protein expression of tight junction proteins and LC3B protein expression. In vitro, L-arginine attenuated HS-induced apoptosis as demonstrated by flow cytometry and decreased cleaved caspase-3 protein expression. L-arginine induced autophagy, which was demonstrated by decreased expression of p62 and p-mTOR/mTOR, and increased expression of LC3B. The protein expression levels of TJ proteins also enhanced by L-arginine in IEC-6 cells. Taken together, these results suggest that L-arginine can alleviate intestinal damage and protect the intestinal integrity by suppressing local inflammation response, promoting the production of TJs and facilitating autophagy under HS conditions.
Collapse
Affiliation(s)
- Liqing Huang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Peng Yin
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Fenghua Liu
- College of Animal Science and Technology, Beijing Agricultural University, Beijing, China
| | - Yilin Liu
- College of Animal Science and Technology, Beijing Agricultural University, Beijing, China
| | - Yanhan Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhaofei Xia
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Schemitt EG, Hartmann RM, Colares JR, Licks F, Salvi JO, Marroni CA, Marroni NP. Protective action of glutamine in rats with severe acute liver failure. World J Hepatol 2019; 11:273-286. [PMID: 30967905 PMCID: PMC6447424 DOI: 10.4254/wjh.v11.i3.273] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute liver failure (SALF) is a rare, but high-mortality, rapidly evolving syndrome that leads to hepatocyte degeneration with impaired liver function. Thioacetamide (TAA) is a known xenobiotic, which promotes the increase of the formation of reactive oxygen species. Erythroid 2-related factor 2 (Nrf2) activates the antioxidant protection of cells. Studies have evidenced the involvement of inflammatory mediators in conditions of oxidative stress. AIM To evaluate the antioxidant effects of glutamine on Nrf2 activation and NFκB-mediated inflammation in rats with TAA-induced IHAG. METHODS Male Wistar rats (n = 28) were divided into four groups: control, control+glutamine, TAA, and TAA + glutamine. Two TAA doses (400 mg/kg) were administered intraperitoneally, 8 h apart. Glutamine (25 mg/kg) was administered at 30 min, 24 h, and 36 h. At 48 h, blood was collected for liver integrity analysis [aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP)]. The liver was harvested for histology and assessment of oxidative stress [thiobarbituric acid-reactive substances (TBARS), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), glutathione (GSH), Nrf2, Kelch-like ECH-associated protein 1 (Keap1), NADPH quinone oxidoreductase1 (NQO1), superoxide dismutase (SOD)] and inflammatory process. RESULTS TAA caused disruption of the hepatic parenchyma, with inflammatory infiltration, massive necrosis, and ballooning degeneration. Glutamine mitigated this tissue damage, with visible regeneration of hepatic parenchyma; decreased TBARS (P < 0.001), GSH (P < 0.01), IL-1β, IL6, and TNFα levels (P <0.01) in hepatic tissue; and decreased blood levels of AST, ALT, and ALP (P <0.05). In addition, CAT, GPx, and GST activities were restored in the glutamine group (P <0.01, P <0.01, and P <0.001, respectively vs TAA alone). Glutamine increased expression of Nrf2 (P < 0.05), NQO1, and SOD (P < 0.01), as well as levels of IL-10 (P <0.001), while decreasing expression of Keap1, TLR4, NFκB (P < 0.001), COX-2 and iNOS, (P < 0.01), and reducing NO2 and NO3 levels (P < 0.05). CONCLUSION In the TAA experimental model of IHAG, glutamine activated the Nrf2 pathway, thus promoting antioxidant protection, and blunted the NFκB-mediated pathway, reducing inflammation.
Collapse
Affiliation(s)
- Elizângela G Schemitt
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Renata M Hartmann
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Josieli R Colares
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Francielli Licks
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Jéferson O Salvi
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Cláudio A Marroni
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil.
| | - Norma P Marroni
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| |
Collapse
|