1
|
Berrino E, Guglielmi P, Carta F, Carradori S, Campestre C, Angeli A, Arrighi F, Pontecorvi V, Chimenti P, Secci D, Supuran CT, Gallorini M. In Vitro CO-Releasing and Antioxidant Properties of Sulfonamide-Based CAI-CORMs in a H 2O 2-Stimulated Human Achilles Tendon-Derived Cell Model. Molecules 2025; 30:593. [PMID: 39942697 PMCID: PMC11819963 DOI: 10.3390/molecules30030593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Tendinopathy is often described as a complex and multifactorial condition which affects tendons. Tendon disorders are marked by a reduction in mechanical function, accompanied by pain and swelling. At the molecular level, tendinopathy leads to oxidative stress-driven inflammation, increased cell death, disruption of extracellular matrix balance, abnormal growth of capillaries and arteries, and degeneration of collagen formation. Here, we report an innovative approach to modulate oxidative stress during tendinopathy based on sulfonamide-based Carbonic Anhydrase Inhibitors-carbon monoxide releasing molecules (CAI-CORMs) hybrids endowed with dual carbon monoxide (CO) releasing activity and carbonic anhydrase (CA) inhibition. The synthesised compounds have been studied in a model of human Achilles tendon-derived cells stimulated by H2O2. Among the library, compound 1c and, to a greater extent, compound 1a, showed to be extremely effective in terms of restoration of cell metabolic activity and cell proliferation due to their capacity to release CO and inhibit the CA isoforms involved in inflammatory processes in the nanomolar range. Moreover, 1a can restore collagen type 1 secretion under pro-oxidant conditions.
Collapse
Affiliation(s)
- Emanuela Berrino
- Department of Life Science, Health, and Health Professions, Link Campus University, Via del Casale di San Pio V, 44, 00165 Rome, Italy;
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (V.P.); (P.C.); (D.S.)
| | - Paolo Guglielmi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (V.P.); (P.C.); (D.S.)
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, University of Florence, Sesto Fiorentino, 50019 Florence, Italy; (F.C.); (A.A.); (C.T.S.)
| | - Simone Carradori
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.C.); (C.C.); (M.G.)
| | - Cristina Campestre
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.C.); (C.C.); (M.G.)
| | - Andrea Angeli
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, University of Florence, Sesto Fiorentino, 50019 Florence, Italy; (F.C.); (A.A.); (C.T.S.)
| | - Francesca Arrighi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (V.P.); (P.C.); (D.S.)
| | - Virginia Pontecorvi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (V.P.); (P.C.); (D.S.)
| | - Paola Chimenti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (V.P.); (P.C.); (D.S.)
| | - Daniela Secci
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (F.A.); (V.P.); (P.C.); (D.S.)
| | - Claudiu T. Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, University of Florence, Sesto Fiorentino, 50019 Florence, Italy; (F.C.); (A.A.); (C.T.S.)
| | - Marialucia Gallorini
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.C.); (C.C.); (M.G.)
| |
Collapse
|
2
|
Chen SY, Xu H, Qin Y, He TQ, Shi RR, Xing YR, Xu J, Cong RC, Wang MR, Yang JS, Gu JH, He BS. Nicotinamide adenine dinucleotide phosphate alleviates intestinal ischemia/reperfusion injury via Nrf2/HO-1 pathway. Int Immunopharmacol 2024; 143:113478. [PMID: 39471691 DOI: 10.1016/j.intimp.2024.113478] [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: 05/24/2024] [Revised: 09/30/2024] [Accepted: 10/20/2024] [Indexed: 11/01/2024]
Abstract
Intestinal ischemia-reperfusion (I/R) injury is a critical condition in the abdomen that has significant morbidity and fatality rates. Prior studies have noted the defensive role of the coenzymatic antioxidant reduced nicotinamide adenine dinucleotide phosphate (NADPH) in heart and brain I/R damage, yet its impact on intestinal I/R trauma required further exploration. Through the application of an in vitro oxygen-glucose deprivation-reoxygenation model and a mouse model of short-term intestinal I/R, this study clarified the defensive mechanisms of NADPH against intestinal I/R injury. We demonstrated that intraperitoneal NADPH administration markedly reduced interleukin-1β (IL-1β) levels and blocked NLRP3 inflammasome activation, hence reducing inflammation. The antioxidative properties of NADPH were established by the reduction of oxidative stress markers and enhancement of glutathione levels. Importantly, NADPH improved intestinal barrier integrity, indicated by an upregulation of zonula occludens-1 and the promotion of a balanced gut microbiome profile. Furthermore, we identified the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1(HO-1) pathway as a crucial conduit for NADPH's beneficence. When this pathway was inhibited by ML385, the favorable outcomes conferred by NADPH were significantly abrogated. These results demonstrate that NADPH functions as an antioxidative, anti-inflammatory, microbiota-balancing, barrier-strengthening, and anti-inflammatory agent against intestinal I/R damage through activation of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Su-Ying Chen
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China; Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Ultrasonography, Wuxi City Rehabilitation Hospital, Liangxi District Chinese Medicine Hospital, Wuxi 214000, China
| | - Hui Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China.
| | - Yan Qin
- Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Tian-Qi He
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Rui-Rui Shi
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Yu-Run Xing
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China; Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Jian Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Ruo-Chen Cong
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Mei-Rong Wang
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Ju-Shun Yang
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jin-Hua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China.
| | - Bo-Sheng He
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China; Translational Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, China.
| |
Collapse
|
3
|
Fan Q, Chang H, Tian L, Zheng B, Liu R, Li Z. Methane saline suppresses ferroptosis via the Nrf2/HO-1 signaling pathway to ameliorate intestinal ischemia-reperfusion injury. Redox Rep 2024; 29:2373657. [PMID: 39023011 PMCID: PMC11259071 DOI: 10.1080/13510002.2024.2373657] [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] [Indexed: 07/20/2024] Open
Abstract
OBJECTIVES Intestinal ischemia-reperfusion (I/R) injury is a multifactorial and complex clinical pathophysiological process. Current research indicates that the pathogenesis of intestinal I/R injury involves various mechanisms, including ferroptosis. Methane saline (MS) has been demonstrated to primarily exert anti-inflammatory and antioxidant effects in I/R injury. In this study, we mainly investigated the effect of MS on ferroptosis in intestinal I/R injury and determined its potential mechanism. METHODS In vivo and in vitro intestinal I/R injury models were established to validate the relationship between ferroptosis and intestinal I/R injury. MS treatment was applied to assess its impact on intestinal epithelial cell damage, intestinal barrier disruption, and ferroptosis. RESULTS MS treatment led to a reduction in I/R-induced intestinal epithelial cell damage and intestinal barrier disruption. Moreover, similar to treatment with ferroptosis inhibitors, MS treatment reduced ferroptosis in I/R, as indicated by a decrease in the levels of intracellular pro-ferroptosis factors, an increase in the levels of anti-ferroptosis factors, and alleviation of mitochondrial damage. Additionally, the expression of Nrf2/HO-1 was significantly increased after MS treatment. However, the intestinal protective and ferroptosis inhibitory effects of MS were diminished after the use of M385 to inhibit Nrf2 in mice or si-Nrf2 in Caco-2 cells. DISCUSSION We proved that intestinal I/R injury was mitigated by MS and that the underlying mechanism involved modulating the Nrf2/HO-1 signaling pathway to decrease ferroptosis. MS could be a promising treatment for intestinal I/R injury.
Collapse
Affiliation(s)
- Qingrui Fan
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Xi’an Medical University, Xi’an, People’s Republic of China
| | - Hulin Chang
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
- Department of Hepatobiliary Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Lifei Tian
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Bobo Zheng
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Ruiting Liu
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| | - Zeyu Li
- Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, People’s Republic of China
| |
Collapse
|
4
|
Tan S, Kong Y, Xian Y, Gao P, Xu Y, Wei C, Lin P, Ye W, Li Z, Zhu X. The Mechanisms of Ferroptosis and the Applications in Tumor Treatment: Enemies or Friends? Front Mol Biosci 2022; 9:938677. [PMID: 35911967 PMCID: PMC9334798 DOI: 10.3389/fmolb.2022.938677] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis, as a newly discovered non-apoptotic cell death mode, is beginning to be explored in different cancer. The particularity of ferroptosis lies in the accumulation of iron dependence and lipid peroxides, and it is different from the classical cell death modes such as apoptosis and necrosis in terms of action mode, biochemical characteristics, and genetics. The mechanism of ferroptosis can be divided into many different pathways, so it is particularly important to identify the key sites of ferroptosis in the disease. Herein, based on ferroptosis, we analyze the main pathways in detail. More importantly, ferroptosis is linked to the development of different systems of the tumor, providing personalized plans for the examination, treatment, and prognosis of cancer patients. Although some mechanisms and side effects of ferroptosis still need to be studied, it is still a promising method for cancer treatment.
Collapse
Affiliation(s)
- Shuzheng Tan
- School of Laboratory Medicine and Biological Engineering, Hangzhou Medical College, Hangzhou, China
- Department of Dermatology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Kong
- Department of Clinical Laboratory, Hubei No.3 People’s Hospital of Jianghan University, Wuhan, China
| | - Yongtong Xian
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Pengbo Gao
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Yue Xu
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Chuzhong Wei
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Peixu Lin
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Weilong Ye
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
- *Correspondence: Zesong Li, ; Xiao Zhu,
| | - Xiao Zhu
- School of Laboratory Medicine and Biological Engineering, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Zesong Li, ; Xiao Zhu,
| |
Collapse
|
5
|
Wang Y, Wen J, Almoiliqy M, Wang Y, Liu Z, Yang X, Lu X, Meng Q, Peng J, Lin Y, Sun P. Sesamin Protects against and Ameliorates Rat Intestinal Ischemia/Reperfusion Injury with Involvement of Activating Nrf2/HO-1/NQO1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5147069. [PMID: 34630849 PMCID: PMC8494576 DOI: 10.1155/2021/5147069] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023]
Abstract
Intestinal ischemia-reperfusion (I/R) may induce cell/tissue injuries, leading to multiple organ failure. Based on our preexperiments, we proposed that sesamin could protect against and ameliorate intestinal I/R injuries and related disorders with involvement of activating Nrf2 signaling pathway. This proposal was evaluated using SD intestinal I/R injury rats in vivo and hypoxia/reoxygenation- (H/R-) injured rat small intestinal crypt epithelial cell line (IEC-6 cells) in vitro. Sesamin significantly alleviated I/R-induced intestinal histopathological injuries and significantly reduced serum biochemical indicators ALT and AST, alleviating I/R-induced intestinal injury in rats. Sesamin also significantly reversed I/R-increased TNF-α, IL-6, IL-1β, and MPO activity in serum and MDA in tissues and I/R-decreased GSH in tissues and SOD in both tissues and IEC-6 cells, indicating its anti-inflammatory and antioxidative stress effects. Further, sesamin significantly decreased TUNEL-positive cells, downregulated the increased Bax and caspase-3 protein expression, upregulated the decreased protein expression of Bcl-2 in I/R-injured intestinal tissues, and significantly reversed H/R-reduced IEC-6 cell viability as well as reduced the number of apoptotic cells among H/R-injured IEC-6 cell, showing antiapoptotic effects. Activation of Nrf2 is known to ameliorate tissue/cell injuries. Consistent with sesamin-induced ameliorations of both intestinal I/R injuries and H/R injuries, transfection of Nrf2 cDNA significantly upregulated the expression of Nrf2, HO-1, and NQO1, respectively. On the contrary, either Nrf2 inhibitor (ML385) or Nrf2 siRNA transfection significantly decreased the expression of these proteins. Our results suggest that activation of the Nrf2/HO-1/NQO1 signaling pathway is involved in sesamin-induced anti-inflammatory, antioxidative, and antiapoptotic effects in protection against and amelioration of intestinal I/R injuries.
Collapse
Affiliation(s)
- Yilin Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001 Liaoning, China
| | - Jin Wen
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Marwan Almoiliqy
- Key Lab of Aromatic Plant Resources Exploitation and Utilization in Sichuan Higher Education, Yibin University, Yibin, 644000 Sichuan, China
| | - Yaojia Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Zhihao Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Xiaobo Yang
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Xiaolong Lu
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Yuan Lin
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Pengyuan Sun
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, 116044 Liaoning, China
| |
Collapse
|
6
|
Gendy A, Soubh A, Al-Mokaddem A, Kotb El-Sayed M. Dimethyl fumarate protects against intestinal ischemia/reperfusion lesion: Participation of Nrf2/HO-1, GSK-3β and Wnt/β-catenin pathway. Biomed Pharmacother 2021; 134:111130. [PMID: 33348309 DOI: 10.1016/j.biopha.2020.111130] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Dimethyl fumarate (DMFU), a known Nrf2 activator, has proven its positive effect in different organs against ischemia/reperfusion (Is/Re) injury. Nevertheless, its possible impact to modulate intestinal Is/Re-induced injury has not been previously demonstrated before. Hence, this study aimed to investigate DMFU mechanistic maneuver against intestinal Is/Re. METHODS To accomplish this goal, Wistar rats were allocated into four groups; Sham-operated (SOP), intestinal Is/Re (1 h/6 h), and 14 days pre-treated DMFU (15 and 25 mg/kg/day, p.o). RESULTS The mechanistic maneuver divulged that DMFU safeguarded the intestine partly via amplifying the expression/content of Nrf2 along with enhancing its downstream, HO-1 expression/content. In addition, DMFU lessened GSK-3β expression/content accompanied by enriching β-catenin expression/content. The antioxidant action was affirmed by enhancing total antioxidant capacity, besides reducing MDA, iNOS, and its by-product, NOx. The DMFU action entailed anti-inflammatory character manifested by down-regulation of expression/content NF-κB with subsequent rebating the contents of TNF-α, IL-1β, and P-selectin, as well as MPO activity. Moreover, DMFU had anti-apoptotic nature demonstrated through enriching Bcl-2 level and diminishing that of caspase-3. CONCLUSION DMFU purveyed tenable novel protective mechanisms and mitigated events associated with intestinal Is/Re mischief either in the lower or the high dose partly by amending of oxidative stress and inflammation through the modulation of Nrf2/HO-1, GSK-3β, and Wnt/β-catenin pathways.
Collapse
Affiliation(s)
- Abdallah Gendy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, October 6 University, Giza, 12585, Egypt.
| | - Ayman Soubh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Giza, 12566, Egypt
| | - Asmaa Al-Mokaddem
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed Kotb El-Sayed
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Helwan, 11790, Egypt
| |
Collapse
|
7
|
Xie J, He X, Fang H, Liao S, Liu Y, Tian L, Niu J. Identification of heme oxygenase-1 from golden pompano (Trachinotus ovatus) and response of Nrf2/HO-1 signaling pathway to copper-induced oxidative stress. CHEMOSPHERE 2020; 253:126654. [PMID: 32464761 DOI: 10.1016/j.chemosphere.2020.126654] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 03/28/2020] [Accepted: 03/29/2020] [Indexed: 06/11/2023]
Abstract
Heme oxygenase-1(HO-1) is a stress-inducible enzyme that mediates antioxidative and cytoprotective effects to maintain cellular redox homeostasis. In the present study, the full sequence of HO-1 was cloned from golden pompano(Trachinotus ovatus) by RT-PCR and RACE-PCR. The full cDNA sequence of HO-1 was 1349 bp in length which comprised of a 726 bp open reading frame (ORF) preceded by 262 bp 5'-untranslated region (UTR), and followed by a 360 bp 3'UTR, encoding 241 amino acid residues. Phylogenetic analysis revealed that HO-1 showed highest similarity to that of Takifugu rubripes. Tissue distribution analysis showed that the expression level of HO-1 was relatively high in heart, liver and spleen. A trial was conducted to investigate the response of Nrf2/HO-1 signaling pathway to oxidative stress induced by copper. The results showed that mRNA expression of NF-E2-related nuclear factor2 (Nrf2), Kelch-like-ECH-associated protein1 (keap1), superoxide dismutase (SOD), catalase (CAT), HO-1, NAD(P)H quinone oxidoreductase 1 (NQO1) and Glutathione peroxidase (GSH-PX) all significantly increased in copper treated group than that in the control group. This work provides new insight into the molecular mechanism underlying the Nrf2/HO-1 pathway in oxidative response in T. ovatus.
Collapse
Affiliation(s)
- Jiajun Xie
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animal and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Xuanshu He
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animal and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Haohang Fang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animal and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Shiyu Liao
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animal and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Yongjian Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animal and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Lixia Tian
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animal and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Jin Niu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animal and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Science, Sun Yat-sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
8
|
Zhang FH, Liu Y, Dong XB, Hao H, Fan KL, Meng XQ, Kong L. Shenmai Injection Upregulates Heme Oxygenase-1 to Confer Protection Against Severe Acute Pancreatitis. J Surg Res 2020; 256:295-302. [PMID: 32712444 DOI: 10.1016/j.jss.2020.06.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 06/01/2020] [Accepted: 06/16/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND To explore the mechanism of Shenmai injection (SMI) on severe acute pancreatitis (SAP) through heme oxygenase-1 (HO-1) signaling. METHODS A total of 40 male Sprague-Dawley (SD) rats (220-260 g) were grouped into the following four categories (n = 10): SAP + SMI + Zinc protoporphyrin (ZnPP), SAP + SMI, SAP, and sham surgery groups. ZnPP is a specific inhibitor of HO-1. Four percent of sodium taurocholate (1 mL/kg) was retrogradely injected via the pancreatic duct to induce the SAP model. The SAP group rats received 1.6 mL/kg saline by intravenous injection 30 min after the induction of SAP. The SAP + SMI group rats received 1.6 mL/kg SMI by intravenous injection 30 min after the induction of SAP. The SAP + SMI + ZnPP group rats received an intravenous injection of 1.6 mL/kg SMI and intraperitoneal administration of 30 mg/kg ZnPP 30 min after the SAP induction. Twenty-four hours after the SAP induction, blood samples were collected for the measurement of amylase, lipase, creatinine, myeloperoxidase, interleukin-10 (IL-10), tumor necrosis factor-α (TNF-α), and HO-1 level, while tissue specimens were harvested for the determination of HO-1, TNF-α, and IL-10 mRNA level. Meanwhile, histopathological changes in organs (pancreas, lung, and kidney) were stored. RESULTS The serum concentration of amylase, lipase, creatinine, and myeloperoxidase was higher in the SAP group than in the SAP + SMI group. Treatment with SMI increased HO-1 and IL-10 level and reduced TNF-α level in serum and tissues compared to the SAP group (P < 0.05). Treatment with SMI abolished the organ-damaging effects of SAP (P < 0.05). Furthermore, suppression of HO-1 expression by ZnPP canceled the aforementioned effects. CONCLUSIONS SMI confers protection against the SAP-induced systemic inflammatory response and multiple organs damage via HO-1 upregulation.
Collapse
Affiliation(s)
- Fei-Hu Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yang Liu
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Bin Dong
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Hao
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kai-Liang Fan
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xian-Qing Meng
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Li Kong
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Silva RCMC, Travassos LH, Paiva CN, Bozza MT. Heme oxygenase-1 in protozoan infections: A tale of resistance and disease tolerance. PLoS Pathog 2020; 16:e1008599. [PMID: 32692767 PMCID: PMC7373268 DOI: 10.1371/journal.ppat.1008599] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Heme oxygenase (HO-1) mediates the enzymatic cleavage of heme, a molecule with proinflammatory and prooxidant properties. HO-1 activity deeply impacts host capacity to tolerate infection through reduction of tissue damage or affecting resistance, the ability of the host to control pathogen loads. In this Review, we will discuss the contribution of HO-1 in different and complex protozoan infections, such as malaria, leishmaniasis, Chagas disease, and toxoplasmosis. The complexity of these infections and the pleiotropic effects of HO-1 constitute an interesting area of study and an opportunity for drug development.
Collapse
Affiliation(s)
- Rafael C. M. C. Silva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Leonardo H. Travassos
- Laboratório de Imunoreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia N. Paiva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- * E-mail:
| |
Collapse
|
10
|
Van Dingenen J, Pieters L, Van Nuffel E, Lefebvre RA. Hemin reduces postoperative ileus in a heme oxygenase 1-dependent manner while dimethyl fumarate does without heme oxygenase 1-induction. Neurogastroenterol Motil 2020; 32:e13624. [PMID: 31121086 DOI: 10.1111/nmo.13624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 03/28/2019] [Accepted: 04/26/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Postoperative ileus (POI), the impairment of gastrointestinal motility after abdominal surgery, is mainly due to intestinal muscular inflammation. Carbon monoxide (CO)-releasing compounds were shown to exert an anti-inflammatory effect in murine POI partially through induction of heme oxygenase-1 (HO-1). The influence of hemin and dimethyl fumarate (DMF), currently used for multiple sclerosis (MS), was therefore tested in murine POI. METHODS C57BL/6J mice were anesthetized and after laparotomy, POI was induced via intestinal manipulation (IM). Animals were treated with either 30 mg kg-1 hemin intraperitoneally (ip), 30 mg kg-1 DMF ip, or 100 mg kg-1 intragastrically (ig) 24 hours before IM. Intestinal transit was assessed 24 hours postoperatively and mucosa-free muscularis or whole segments of the small intestine were stored for later analysis. Intestinal HO-1 protein expression was studied at 6, 12, and 24 hours after administration of hemin or DMF in non-manipulated mice. KEY RESULTS Pretreatment with hemin and DMF, both ig and ip, prevented the delayed transit seen after IM. Concomitantly, both hemin and DMF significantly reduced the increased interleukin-6 levels and the elevated leukocyte infiltration in the muscularis. Hemin but not DMF caused a significant increase in intestinal HO-1 protein expression and co-administration of the HO-1 inhibitor chromium mesoporphyrin abolished the protective effects of hemin on POI; DMF reduced the IM-induced activation of NF-κB and ERK 1/2. CONCLUSIONS AND INFERENCES Both hemin and DMF improve the delayed transit and inflammation seen in murine POI, but only hemin does so in a HO-1-dependent manner.
Collapse
Affiliation(s)
- Jonas Van Dingenen
- Department of Basic and Applied Medical Sciences, Faculty of Medicine & Health Sciences, Ghent University, Ghent, Belgium
| | - Leen Pieters
- Department of Human Structure and Repair, Faculty of Medicine & Health Sciences, Ghent University, Ghent, Belgium
| | - Elien Van Nuffel
- Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Romain A Lefebvre
- Department of Basic and Applied Medical Sciences, Faculty of Medicine & Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Liu T, Mukosera GT, Blood AB. The role of gasotransmitters in neonatal physiology. Nitric Oxide 2019; 95:29-44. [PMID: 31870965 DOI: 10.1016/j.niox.2019.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 11/07/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
The gasotransmitters, nitric oxide (NO), hydrogen sulfide (H2S), and carbon monoxide (CO), are endogenously-produced volatile molecules that perform signaling functions throughout the body. In biological tissues, these small, lipid-permeable molecules exist in free gaseous form for only seconds or less, and thus they are ideal for paracrine signaling that can be controlled rapidly by changes in their rates of production or consumption. In addition, tissue concentrations of the gasotransmitters are influenced by fluctuations in the level of O2 and reactive oxygen species (ROS). The normal transition from fetus to newborn involves a several-fold increase in tissue O2 tensions and ROS, and requires rapid morphological and functional adaptations to the extrauterine environment. This review summarizes the role of gasotransmitters as it pertains to newborn physiology. Particular focus is given to the vasculature, ventilatory, and gastrointestinal systems, each of which uniquely illustrate the function of gasotransmitters in the birth transition and newborn periods. Moreover, given the relative lack of studies on the role that gasotransmitters play in the newborn, particularly that of H2S and CO, important gaps in knowledge are highlighted throughout the review.
Collapse
Affiliation(s)
- Taiming Liu
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - George T Mukosera
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Arlin B Blood
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA; Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
12
|
Ahmad AF, Dwivedi G, O'Gara F, Caparros-Martin J, Ward NC. The gut microbiome and cardiovascular disease: current knowledge and clinical potential. Am J Physiol Heart Circ Physiol 2019; 317:H923-H938. [PMID: 31469291 DOI: 10.1152/ajpheart.00376.2019] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The human body is populated by a diverse community of microbes, dominated by bacteria, but also including viruses and fungi. The largest and most complex of these communities is located in the gastrointestinal system and, with its associated genome, is known as the gut microbiome. Gut microbiome perturbations and related dysbiosis have been implicated in the progression and pathogenesis of CVD, including atherosclerosis, hypertension, and heart failure. Although there have been advances in the characterization and analysis of the gut microbiota and associated bacterial metabolites, the exact mechanisms through which they exert their action are not well understood. This review will focus on the role of the gut microbiome and associated functional components in the development and progression of atherosclerosis. Potential treatments to alter the gut microbiome to prevent or treat atherosclerosis and CVD are also discussed.
Collapse
Affiliation(s)
- Adilah F Ahmad
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia.,Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Fergal O'Gara
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,BIOMERIT Research Centre, School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland.,Telethon Kids Institute, Children's Hospital, Perth, Western Australia, Australia
| | - Jose Caparros-Martin
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Natalie C Ward
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,School of Public Health, Curtin University, Perth Western Australia, Australia
| |
Collapse
|
13
|
Nunes NS, Chandran P, Sundby M, Visioli F, da Costa Gonçalves F, Burks SR, Paz AH, Frank JA. Therapeutic ultrasound attenuates DSS-induced colitis through the cholinergic anti-inflammatory pathway. EBioMedicine 2019; 45:495-510. [PMID: 31253515 PMCID: PMC6642284 DOI: 10.1016/j.ebiom.2019.06.033] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/05/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ulcerative Colitis (UC) is an Inflammatory Bowel Disease (IBD) characterized by uncontrolled immune response, diarrhoea, weight loss and bloody stools, where sustained remission is not currently achievable. Dextran Sulphate Sodium (DSS)-induced colitis is an animal model that closely mimics human UC. Ultrasound (US) has been shown to prevent experimental acute kidney injury through vagus nerve (VN) stimulation and activation of the cholinergic anti-inflammatory pathway (CAIP). Since IBD patients may present dysfunctional VN activity, our aim was to determine the effects of therapeutic ultrasound (TUS) in DSS-induced colitis. METHODS Acute colitis was induced by 2% DSS in drinking water for 7 days and TUS was administered to the abdominal area for 7 min/day from days 4-10. Clinical symptoms were analysed, and biological samples were collected for proteomics, macroscopic and microscopic analysis, flow cytometry and immunohistochemistry. FINDINGS TUS attenuated colitis by reducing clinical scores, colon shortening and histological damage, inducing proteomic tolerogenic response in the gut during the injury phase and early recovery of experimental colitis. TUS did not improve clinical and pathological outcomes in splenectomised mice, while α7nAChR (α7 nicotinic acetylcholine receptor - indicator of CAIP involvement) knockout animals presented with disease worsening. Increased levels of colonic F4/80+α7nAChR+ macrophages in wild type mice suggest CAIP activation. INTERPRETATION These results indicate TUS improved DSS-induced colitis through stimulation of the splenic nerve along with possible contribution by VN with CAIP activation. FUND: Intramural Research Programs of the Clinical Centre, the National Institute of Biomedical Imaging and Bioengineering at the NIH and CAPES/Brazil.
Collapse
Affiliation(s)
- Natalia Schneider Nunes
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States; Gastroenterology and Hepatology Sciences Graduate Program, UFRGS, Porto Alegre, RS, Brazil.
| | - Parwathy Chandran
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States
| | - Maggie Sundby
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States
| | - Fernanda Visioli
- Faculty of Dentistry, Oral Pathology, UFRGS, Porto Alegre, RS, Brazil
| | | | - Scott Robert Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States
| | - Ana Helena Paz
- Gastroenterology and Hepatology Sciences Graduate Program, UFRGS, Porto Alegre, RS, Brazil
| | - Joseph Alan Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Centre, NIH, Bethesda, MD, United States; National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, MD, United States
| |
Collapse
|
14
|
Effect of heme oxygenase-1 on the protection of ischemia reperfusion injury of bile duct in rats after liver transplantation. Clin Res Hepatol Gastroenterol 2018; 42:245-254. [PMID: 29174380 DOI: 10.1016/j.clinre.2017.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/13/2017] [Accepted: 09/19/2017] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To investigate the effect of heme oxygenase-1 (HO-1) on the ischemic reperfusion injury (IRI) of bile duct in rat models after liver transplantation. METHODS 320 SD rats were equally and randomly divided into 5 groups, which were group A receiving injection of 3×108/pfu/ml adenovirus (adv), group B with donor receiving Adv-HO-1 and recipient receiving Adv-HO-1-siRNA, group C with donor and recipient both receiving Adv-HO-1, group D with donor receiving Adv-HO-1-siRNA and recipient receiving Adv-HO-1, and group E with donor and recipient both receiving Adv-HO-1-siRNA at 24h before liver transplantation. Donor liver was stored in UW liquid at 4°C followed by measuring HO-1 level by western blot before transplantation. On d1, d3, d7 and d14, serum and liver was isolated for analysis of liver function, inflammatory cell infiltration by H&E staining, ultrastructure of liver by transmission electron microscopy as well as the expression of HO-1, Bsep, Mrp2 and Ntcp by western blot. RESULTS Compared with group D and E, group B and C displayed improved liver function as demonstrated by lower level of ALT, AST, LDH, TBIL, ALP and GGT, increased secretion of TBA and PL as well as expression of transporter proteins (Bsep, Mrp2 and Ntcp), reduced inflammatory cells infiltration and liver injury. CONCLUSION Our study demonstrated that overexpression of HO-1 in donor liver can ameliorate the damage to bile duct and liver, and improved liver function, suggesting HO-1 might be a new therapeutic target in the treatment of IRI after liver transplantation.
Collapse
|
15
|
Nadatani Y, Watanabe T, Shimada S, Otani K, Tanigawa T, Fujiwara Y. Microbiome and intestinal ischemia/reperfusion injury. J Clin Biochem Nutr 2018; 63:26-32. [PMID: 30087540 PMCID: PMC6064812 DOI: 10.3164/jcbn.17-137] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/22/2018] [Indexed: 12/12/2022] Open
Abstract
Intestinal ischemia/reperfusion injury is a severe disease associated with a high mortality. The mechanisms that cause ischemia/reperfusion injury are complex and many factors are involved in the injury formation process; however, the only available treatment is surgical intervention. Recent studies demonstrated that the intestinal microbiome plays a key role in intestinal ischemia/reperfusion injury and there are many factors associated with intestinal bacteria during the formation of the intestinal ischemia/reperfusion injury. Among the Toll-like receptors (TLR), TLR2, TLR4, and their adaptor protein, myeloid differentiation primary-response 88 (MyD88), have been reported to be involved in intestinal ischemia/reperfusion injury. Oxidative stress and nitric oxide are also associated with intestinal bacteria during the formation of the intestinal ischemia/reperfusion injury. This review focuses on our current understanding of the impact of the microbiome, including the roles of the TLRs, oxidative stress, and nitric oxide, on intestinal ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Yuji Nadatani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka City, Osaka 545-8585, Japan
| | - Toshio Watanabe
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka City, Osaka 545-8585, Japan
| | - Sunao Shimada
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka City, Osaka 545-8585, Japan
| | - Koji Otani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka City, Osaka 545-8585, Japan
| | - Tetsuya Tanigawa
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka City, Osaka 545-8585, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka City, Osaka 545-8585, Japan
| |
Collapse
|
16
|
Strand-Amundsen RJ, Reims HM, Reinholt FP, Ruud TE, Yang R, Høgetveit JO, Tønnessen TI. Ischemia/reperfusion injury in porcine intestine - Viability assessment. World J Gastroenterol 2018; 24:2009-2023. [PMID: 29760544 PMCID: PMC5949714 DOI: 10.3748/wjg.v24.i18.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate viability assessment of segmental small bowel ischemia/reperfusion in a porcine model.
METHODS In 15 pigs, five or six 30-cm segments of jejunum were simultaneously made ischemic by clamping the mesenteric arteries and veins for 1 to 16 h. Reperfusion was initiated after different intervals of ischemia (1-8 h) and subsequently monitored for 5-15 h. The intestinal segments were regularly photographed and assessed visually and by palpation. Intraluminal lactate and glycerol concentrations were measured by microdialysis, and samples were collected for light microscopy and transmission electron microscopy. The histological changes were described and graded.
RESULTS Using light microscopy, the jejunum was considered as viable until 6 h of ischemia, while with transmission electron microscopy the ischemic muscularis propria was considered viable until 5 h of ischemia. However, following ≥ 1 h of reperfusion, only segments that had been ischemic for ≤ 3 h appeared viable, suggesting a possible upper limit for viability in the porcine mesenteric occlusion model. Although intraluminal microdialysis allowed us to closely monitor the onset and duration of ischemia and the onset of reperfusion, we were unable to find sufficient level of association between tissue viability and metabolic markers to conclude that microdialysis is clinically relevant for viability assessment. Evaluation of color and motility appears to be poor indicators of intestinal viability.
CONCLUSION Three hours of total ischemia of the small bowel followed by reperfusion appears to be the upper limit for viability in this porcine mesenteric ischemia model.
Collapse
Affiliation(s)
- Runar J Strand-Amundsen
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Oslo 0424, Norway
- Department of Physics, University of Oslo, Oslo 0316, Norway
| | - Henrik M Reims
- Department of Pathology, Oslo University Hospital, Oslo 0424, Norway
| | - Finn P Reinholt
- Department of Pathology, Oslo University Hospital, Oslo 0424, Norway
| | - Tom E Ruud
- Institute for Surgical Research, Oslo University Hospital, Oslo 0424, Norway
- Department of Surgery, Baerum Hospital, Vestre Viken Hospital Trust, Drammen 3004, Norway
| | - Runkuan Yang
- Department of Emergencies and Critical Care, Oslo University Hospital, Oslo 0424, Norway
| | - Jan O Høgetveit
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Oslo 0424, Norway
- Department of Physics, University of Oslo, Oslo 0316, Norway
| | - Tor I Tønnessen
- Department of Emergencies and Critical Care, Oslo University Hospital, Oslo 0424, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo 0424, Norway
| |
Collapse
|
17
|
Ren QG, Yang SL, Li PD, Xiong JR, Fang X, Hu JL, Wang QS, Chen RW, Chen YS, Wen L, Peng M. Low heme oxygenase-1 expression promotes gastric cancer cell apoptosis, inhibits proliferation and invasion, and correlates with increased overall survival in gastric cancer patients. Oncol Rep 2017; 38:2852-2858. [PMID: 29048628 DOI: 10.3892/or.2017.5967] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/11/2017] [Indexed: 11/06/2022] Open
Abstract
Heme oxygenase-1 (HO-1) plays a key role in anti-oxidation, anti-apoptosis, and anti-proliferation in various types of cancers. However, the relationship between HO-1 expression and gastric cancer development remains largely unknown. In this study, the protein expression of HO-1 in human gastric cancer was measured by immunohistochemistry on paraffin sections of 89 paired gastric carcinoma tissues and adjacent non-cancer tissues. The correlation of HO-1 expression with 5-year overall survival rate was estimated. The effects of decreased HO-1 expression by two strands of small interfered RNAs (siRNAs) on cell apoptosis, proliferation, and invasion of gastric cancer cell lines were examined by flow cytometry, the MTT assay, and the cell migration assay, respectively. High expression of HO-1 was detected in 11.2% (10/89) of gastric carcinoma tissues, compared with 1.1% (1/89) in matched adjacent normal tissues, and correlated with a decreased survival rate in gastric cancer patients. There were no significant correlations between HO-1 expression and clinical characteristics. Downregulation of HO-1 expression using two strands of siRNAs promoted apoptosis and inhibited the proliferation and invasion of two gastric cancer cell lines, SGC7901 and MKN-28 cells. This study demonstrated that HO-1 plays a vital role in the development of gastric cancer and may serve as a therapeutic target of this type of cancer.
Collapse
Affiliation(s)
- Quan-Guang Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Sheng-Li Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Pin-Dong Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | | | - Xiefan Fang
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jian-Li Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Qiu-Shuang Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Ren-Wang Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Ye-Shan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Lu Wen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Miao Peng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| |
Collapse
|
18
|
Zhang FH, Sun YH, Fan KL, Dong XB, Han N, Zhao H, Kong L. Protective effects of heme oxygenase-1 against severe acute pancreatitis via inhibition of tumor necrosis factor-α and augmentation of interleukin-10. BMC Gastroenterol 2017; 17:100. [PMID: 28836936 PMCID: PMC5571505 DOI: 10.1186/s12876-017-0651-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023] Open
Abstract
Background Heme oxygenase-1 (HO-1) is an inducible defense gene which plays a significant role in inflammation. HO-1 protects cells and tissues through the mechanism of anti-oxidation, maintaining microcirculation and anti-inflammation. The aim of the current study is to investigate the role of HO-1 on systemic inflammatory response in severe acute pancreatitis (SAP). Methods Forty male Sprague-Dawley (SD) rats were randomly assigned into four groups: control group (n = 10); SAP group (n = 10), SAP model was induced by retrograde injection of 3% sodium taurocholate through pancreatic duct; HO-1 stimulation group (n = 10), SD rats were injected 75 μg/kg hemin intraperitoneally 30 min after induction of SAP; HO-1 inhibition group (n = 10), SD rats were injected 20 μg/kg Zinc porphyrin (Zn-PP) intraperitoneally 30 min after induction of SAP. After 24 h of SAP establishment, tissues were collected for HO-1, tumor necrosis factor-α (TNF-α) and interleukin-10 (IL-10) mRNA expression, and blood samples were collected for cytokines and biochemical measurements. Meanwhile, the histopathological changes of pancreas and liver tissues were observed. Results The expression of HO-1 mRNA and protein were significantly induced by SAP in rat pancreas and liver. Hemin treatment significantly decreased oxidative stress and TNF-α in plasma and tissues, while the IL-10 was significantly increased. Pancreas and liver injury induced by SAP was markedly attenuated by Hemin treatment. Moreover, inhibition of HO-1 expression by Zn-PP administration aggravated the injury caused by SAP. Conclusions Induction of HO-1 in early SAP may modulate systemic inflammatory response and prevent pancreas and nearby organs such as liver injury through inhibition of TNF-α and augmentation of IL-10.
Collapse
Affiliation(s)
- Fei-Hu Zhang
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369, Jinan, Shandong Province, 250011, China
| | - Yu-Han Sun
- Department of Traditional Chinese Medicine, Jinan Municipal Organs Hospital, Jianguoxiaojingsan Road No.35, Jinan, Shandong Province, 250001, China
| | - Kai-Liang Fan
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369, Jinan, Shandong Province, 250011, China
| | - Xiao-Bin Dong
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369, Jinan, Shandong Province, 250011, China
| | - Ning Han
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369, Jinan, Shandong Province, 250011, China
| | - Hao Zhao
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369, Jinan, Shandong Province, 250011, China
| | - Li Kong
- Department of Emergency Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369, Jinan, Shandong Province, 250011, China.
| |
Collapse
|
19
|
Fan X, Mu L. The role of heme oxygenase-1 (HO-1) in the regulation of inflammatory reaction, neuronal cell proliferation and apoptosis in rats after intracerebral hemorrhage (ICH). Neuropsychiatr Dis Treat 2017; 13:77-85. [PMID: 28096675 PMCID: PMC5207456 DOI: 10.2147/ndt.s120496] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To investigate the role of heme oxygenase-1 (HO-1) in the regulation of inflammatory reaction, neuronal cell proliferation and apoptosis in rats after intracerebral hemorrhage (ICH). METHODS Thirty-six adult Sprague Dawley (SD) male rats were randomly divided into sham operation, ICH and zinc protoporphyrin (ZPP) group. Rats (except for the sham operation group) were given 50 μL stereotactic injection of autologous blood from the femoral artery into the caudate nucleus, to establish an ICH model. In addition, rats in the ZPP group were given 10 mg/kg intraperitoneal injection of ZPP. At day 3 postoperative, neurobehavioral changes and brain water content were evaluated, brain tissue HO-1 expression was detected with immunohistochemistry and reverse transcription polymerase chain reaction (RT-PCR), brain tissue apoptosis was evaluated with TUNEL method, Caspase 3, Caspase 8 and Caspase 9 activity were detected with colorimetric method, level of TNF-α, IL-1β, IL-6 and IL-8 were measured with the enzyme-linked immunosorbent assay (ELISA), while Bcl-2, Bax, p-NF-κB p65 and p-IκBα protein expression were detected with Western blot. RESULTS ICH group compared to sham operation: HO-1 positive rate and mRNA expression were increased, neurological deficit score and cell apoptosis rate were increased, Caspase 3, Caspase 8 and Caspase 9 activity were increased, level of TNF-α, IL-1β, IL-6 and IL-8 were increased, Bcl-2 expression was downregulated, Bax, p-NF-κB p65 and p-IκBα expression were upregulated. The differences were statistically significant (P<0.01). ZPP group compared to ICH: HO-1 positive rate and mRNA expression were decreased, neurological deficit score and cell apoptosis rate were decreased, Caspase 3, Caspase 8, Caspase 9 activity were decreased, level of TNF-α, IL-1β, IL-6 and IL-8 were decreased, Bcl-2 expression was upregulated, Bax, p-NF-κB p65 and p-IκBα expression were downregulated, and the differences were statistically significant (P<0.01). CONCLUSION HO-1 inhibitor, ZPP does have a protective effect on ICH rats. This might be due to its inhibition to the inflammatory reaction and neuronal cell apoptosis.
Collapse
Affiliation(s)
- Xuezheng Fan
- Department of Neurosurgery, People's Hospital of Guangming New District, Shenzhen, Guangdong, People's Republic of China
| | - Linshen Mu
- Department of Neurosurgery, Guangzhou Medical University Affiliated Brain Hospital, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
20
|
Corcoran TB, Mas E, Barden AE, Roberts LJ, Mori TA, O'Loughlin E. Controlled moderate hypovolaemia in healthy volunteers is not associated with the development of oxidative stress assessed by plasma F2-isoprostanes and isofurans. Prostaglandins Other Lipid Mediat 2016; 124:34-8. [PMID: 27381810 DOI: 10.1016/j.prostaglandins.2016.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/23/2016] [Accepted: 07/01/2016] [Indexed: 02/07/2023]
Abstract
Hypovolaemia can be associated with substantial morbidity, particularly when it occurs in the setting of trauma and in patients with comorbid diseases. Hypovolaemia and inflammation such as occur in the setting of trauma and surgery, are associated with systemic oxidative stress and free-radical injury. Free-radical injury that results from hypovolaemia-induced organ reperfusion may further augment inflammatory processes. It is unknown exactly what proportion of free-radical injury is associated with isolated hypovolaemia as opposed to the contribution from inflammation from surgery or trauma. In the first human study of its kind, we exposed 8 adult male volunteers to venesection-induced hypovolaemia in progressive aliquots of 5% of total blood volume until 20% had been removed. This blood was subsequently reinfused. Plasma F2-isoprostanes and isofurans, markers of in vivo lipid oxidation, were measured by gas chromatography-mass spectrometry at each 5% aliquot venesected and at each 5% reinfused. Between baseline and maximal blood loss there was a minor fall in haemoglobin concentration from 143.9g/l to 138.8g/l (p=0.004, 95% CI 2.2, 8.0g/L). No significant change from baseline occurred in the concentrations of either plasma F2-isoprostanes or isofurans during venesection (p=0.116 and p=0.152, respectively) or blood reinfusion (p=0.553 and p=0.736, respectively). We can conclude that in healthy adult volunteers, isolated hypovolaemia to 20% total blood volume loss is not associated with detectable systemic oxidative stress. The free-radical injury identified in surgical and trauma patients may represent the effects of tissue damage and inflammation, with an uncertain contribution from tissue ischemia as may occur with hypovolaemia.
Collapse
Affiliation(s)
- Tomas B Corcoran
- Department of Anaesthesia & Pain Medicine, Royal Perth Hospital, Perth, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Emilie Mas
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Anne E Barden
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia.
| | | | - Trevor A Mori
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Edmond O'Loughlin
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; Department of Anaesthesia, Fremantle Hospital, Fremantle, Western Australia, Australia
| |
Collapse
|
21
|
Ma T, Chen T, Li P, Ye Z, Zhai W, Jia L, Chen W, Sun A, Huang Y, Wei S, Li Z. Heme oxygenase-1 (HO-1) protects human lens epithelial cells (SRA01/04) against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis. Exp Eye Res 2016; 146:318-329. [DOI: 10.1016/j.exer.2016.02.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/02/2016] [Accepted: 02/29/2016] [Indexed: 02/07/2023]
|
22
|
Su D, Wu S, Guo J, Wu X, Yang Q, Xiong X. Protective effect of resveratrol against pseudorabies virus-induced reproductive failure in a mouse model. Food Sci Biotechnol 2016; 25:103-106. [PMID: 30263493 DOI: 10.1007/s10068-016-0105-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 02/27/2016] [Accepted: 02/29/2016] [Indexed: 12/14/2022] Open
Abstract
Resveratrol (RES), a natural polyphenol that is abundant in grapes, exerts anti-inflammatory, anti-oxidative, and antiviral bioactive effects. Protective effects of RES against pseudorabies virus (PrV)- induced reproductive failure were investigated in a mice model. Injection of PrV partially induced stillbirth and abortion, and caused poor growth of progeny. Treatment with RES attenuated the reproductive failure induced by the virus with recovery of the serum progesterone level. RES improved the growth performance of newborn mice. RES can attenuate the reproductive failure induced by PrV in mice.
Collapse
Affiliation(s)
- Dingding Su
- 1College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128 China
| | - Shusong Wu
- 1College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128 China
| | - Jing Guo
- 1College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128 China
| | - Xiaosong Wu
- 1College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128 China
| | - Qing Yang
- 2College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, 410128 China
| | - Xingyao Xiong
- Hunan Province Key Laboratory of Crop Germplasm Innovation and Utilization, Changsha, Hunan, 410128 China
| |
Collapse
|
23
|
Gordeeva AE, Temnov AA, Charnagalov AA, Sharapov MG, Fesenko EE, Novoselov VI. Protective Effect of Peroxiredoxin 6 in Ischemia/Reperfusion-Induced Damage of Small Intestine. Dig Dis Sci 2015; 60:3610-9. [PMID: 26233545 DOI: 10.1007/s10620-015-3809-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/13/2015] [Indexed: 12/09/2022]
Abstract
BACKGROUND Strong oxidative stress starting in the epithelium upon restoration of blood cell circulation is a major cause of necrosis of the intestinal epithelium in ischemia/reperfusion-induced damage. AIM The purpose of this study was to investigate the tissue-protective effect of exogenous peroxiredoxin 6 (Prx6) in ischemia/reperfusion-induced damage of small intestine. METHODS The research was carried out using a model of acute superior mesenteric artery occlusion in Wistar male rats. Exogenous Prx6 was administrated intravenously 15 min prior to small intestine ischemia. The distribution of endogenous Prx6 in the small intestine was determined by immunohistochemical analysis. The expression level of antioxidant enzymes was evaluated by RT-PCR in real time. RESULTS Exogenous Prx6 injected to animals intravenously was detected in blood vessel lumens, and its diffuse distribution was subsequently confirmed in the intestinal epithelium. Expression analysis of genes coding for major antioxidant enzymes demonstrated a significant activation of SOD 1, SOD 3, Prx6, GPx2, GPx7 expression during I/R-induced damage of the small intestine. Injection of exogenous Prx6 prior to induced ischemia resulted in minimization of oxidative injury by reducing necrosis and apoptosis, by normalization of gene activity of antioxidant enzyme. It eventually led to a reduction of epithelium destruction in the small intestine. By contrast, administration of a purified mutant form of Prx6 (Prx6C47S) without peroxidase activity had no protective effect. CONCLUSION The application of exogenous Prx6 enables normalization of the antioxidant status of the small intestine and reduction of cell destruction upon I/R-induced organ damage.
Collapse
Affiliation(s)
- A E Gordeeva
- Institute of Cell Biophysics, Russian Academy of Sciences, 3, Institutskaya, Pushchino, Moscow Region, Russia, 142290.
| | - A A Temnov
- N.V. Sklifosovsky Research Institute for Emergency Medicine of Moscow Healthcare Department, Moscow, Russia.
| | - A A Charnagalov
- Department of Structural and Computational Biology, Max. F. Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030, Vienna, Austria.
| | - M G Sharapov
- Institute of Cell Biophysics, Russian Academy of Sciences, 3, Institutskaya, Pushchino, Moscow Region, Russia, 142290.
| | - E E Fesenko
- Institute of Cell Biophysics, Russian Academy of Sciences, 3, Institutskaya, Pushchino, Moscow Region, Russia, 142290.
| | - V I Novoselov
- Institute of Cell Biophysics, Russian Academy of Sciences, 3, Institutskaya, Pushchino, Moscow Region, Russia, 142290.
| |
Collapse
|
24
|
Li X, Ye F, Li L, Chang W, Wu X, Chen J. The role of HO-1 in protection against lead-induced neurotoxicity. Neurotoxicology 2015; 52:1-11. [PMID: 26542248 DOI: 10.1016/j.neuro.2015.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/05/2015] [Accepted: 10/28/2015] [Indexed: 12/01/2022]
Abstract
Lead is a pervasive and persistent environmental pollutant that exerts deleterious effects on all living organisms and continues to threaten public health on a global scale. Heme oxygenase-1 (HO-1) is a stress-inducible enzyme that mediates antioxidative and cytoprotective effects to maintain cellular redox homeostasis and protect cells from oxidative stress. This study was designed to explore the role of HO-1 in protection against lead neurotoxicity and the signaling pathways involved. Lead acetate (PbAc) exposure resulted in increased HO-1 expression in primary rat hippocampal neurons and SH-SY5Y cells. PbAc-induced intracellular reactive oxygen species (ROS) also increased, and cell viability decreased in SH-SY5Y cells. We further demonstrated that HO-1 could be induced by PbAc through the P38, ERK1/2, and PI3K/AKT signaling pathways in a ROS-dependent manner and through the JNK pathway in a ROS-independent manner. Further investigation revealed that HO-1 overexpression significantly restrained cell apoptosis and ROS production induced by PbAc in SH-SY5Y cells. Moreover, HO-1 knockdown aggravated PbAc-induced cell apoptosis and ROS production. Our results indicated that HO-1 was a novel protective factor that could efficiently inhibit PbAc-induced oxidative stress and cell death in the nervous system, thereby providing the potential therapeutic strategies for the prevention and treatment of lead-related diseases.
Collapse
Affiliation(s)
- Xiaoyi Li
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Fang Ye
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Ministry of Education Key Lab for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Lili Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Ministry of Education Key Lab for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Wei Chang
- Department of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, PR China
| | - Xiongwen Wu
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Jun Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Ministry of Education Key Lab for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
25
|
Wang J, He GZ, Wang YK, Zhu QK, Chen W, Guo T. TLR4-HMGB1-, MyD88- and TRIF-dependent signaling in mouse intestinal ischemia/reperfusion injury. World J Gastroenterol 2015; 21:8314-8325. [PMID: 26217083 PMCID: PMC4507101 DOI: 10.3748/wjg.v21.i27.8314] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/21/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To characterize high-mobility group protein 1-toll-like receptor 4 (HMGB1-TLR4) and downstream signaling pathways in intestinal ischemia/reperfusion (I/R) injury.
METHODS: Forty specific-pathogen-free male C57BL/6 mice were randomly divided into five groups (n = 8 per group): sham, control, anti-HMGB1, anti-myeloid differentiation gene 88 (MyD88), and anti-translocating-chain-associating membrane protein (TRIF) antibody groups. Vehicle with the control IgG antibody, anti-HMGB1, anti-MyD88, or anti-TRIF antibodies (all 1 mg/kg, 0.025%) were injected via the caudal vein 30 min prior to ischemia. After anesthetization, the abdominal wall was opened and the superior mesenteric artery was exposed, followed by 60 min mesenteric ischemia and then 60 min reperfusion. For the sham group, the abdominal wall was opened for 120 min without I/R. Levels of serum nuclear factor (NF)-κB p65, interleukin (IL)-6, and tumor necrosis factor (TNF)-α were measured, along with myeloperoxidase activity in the lung and liver. In addition,morphologic changes that occurred in the lung and intestinal tissues were evaluated. Levels of mRNA transcripts encoding HMGB1 and NF-κB were measured by real-time quantitative PCR, and levels of HMGB1 and NF-κB protein were measured by Western blot. Results were analyzed using one-way analysis of variance.
RESULTS: Blocking HMGB1, MyD88, and TRIF expression by injecting anti-HMGB1, anti-MyD88, or anti-TRIF antibodies prior to ischemia reduced the levels of inflammatory cytokines in serum; NF-κB p65: 104.64 ± 11.89, 228.53 ± 24.85, 145.00 ± 33.63, 191.12 ± 13.22, and 183.73 ± 10.81 (P < 0.05); IL-6: 50.02 ± 6.33, 104.91 ± 31.18, 62.28 ± 6.73, 85.90 ± 17.37, and 78.14 ± 7.38 (P < 0.05); TNF-α, 43.79 ± 4.18, 70.81 ± 6.97, 52.76 ± 5.71, 63.19 ± 5.47, and 59.70 ± 4.63 (P < 0.05) for the sham, control, anti-HMGB1, anti-MyD88, and anti-TRIF groups, respectively (all in pg/mL).Antibodies also alleviated tissue injury in the lung and small intestine compared with the control group in the mouse intestinal I/R model. The administration of anti-HMGB1, anti-MyD88, and anti-TRIF antibodies markedly reduced damage caused by I/R, for which anti-HMGB1 antibody had the most obvious effect.
CONCLUSION: HMGB1 and its downstream signaling pathway play important roles in the mouse intestinal I/R injury, and the effect of the TRIF-dependent pathway is slightly greater.
Collapse
|
26
|
Organ-Protective Effects of Red Wine Extract, Resveratrol, in Oxidative Stress-Mediated Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:568634. [PMID: 26161238 PMCID: PMC4487914 DOI: 10.1155/2015/568634] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/09/2014] [Indexed: 12/25/2022]
Abstract
Resveratrol, a polyphenol extracted from red wine, possesses potential antioxidative and anti-inflammatory effects, including the reduction of free radicals and proinflammatory mediators overproduction, the alteration of the expression of adhesion molecules, and the inhibition of neutrophil function. A growing body of evidence indicates that resveratrol plays an important role in reducing organ damage following ischemia- and hemorrhage-induced reperfusion injury. Such protective phenomenon is reported to be implicated in decreasing the formation and reaction of reactive oxygen species and pro-nflammatory cytokines, as well as the mediation of a variety of intracellular signaling pathways, including the nitric oxide synthase, nicotinamide adenine dinucleotide phosphate oxidase, deacetylase sirtuin 1, mitogen-activated protein kinase, peroxisome proliferator-activated receptor-gamma coactivator 1 alpha, hemeoxygenase-1, and estrogen receptor-related pathways. Reperfusion injury is a complex pathophysiological process that involves multiple factors and pathways. The resveratrol is an effective reactive oxygen species scavenger that exhibits an antioxidative property. In this review, the organ-protective effects of resveratrol in oxidative stress-related reperfusion injury will be discussed.
Collapse
|
27
|
Ginsenoside Rb1 Treatment Attenuates Pulmonary Inflammatory Cytokine Release and Tissue Injury following Intestinal Ischemia Reperfusion Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:843721. [PMID: 26161243 PMCID: PMC4487341 DOI: 10.1155/2015/843721] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/22/2014] [Accepted: 12/27/2014] [Indexed: 12/19/2022]
Abstract
Objective. Intestinal ischemia reperfusion (II/R) injury plays a critical role in remote organ dysfunction, such as lung injury, which is associated with nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway. In the present study, we tested whether ginsenoside Rb1 attenuated II/R induced lung injury by Nrf2/HO-1 pathway. Methods. II/R injury was induced in male C57BL/6J mice by 45 min of superior mesenteric artery (SMA) occlusion followed by 2 hours of reperfusion. Ginsenoside Rb1 was administrated prior to reperfusion with or without ATRA (all-transretinoic acid, the inhibitor of Nrf2/ARE signaling pathway) administration before II/R. Results. II/R induced lung histological injury, which is accompanied with increased levels of malondialdehyde (MDA), interleukin- (IL-) 6, and tumor necrosis factor- (TNF-) α but decreased levels of superoxide dismutase (SOD) and IL-10 in the lung tissues. Ginsenoside Rb1 reduced lung histological injury and the levels of TNF-α and MDA, as well as wet/dry weight ratio. Interestingly, the increased Nrf2 and HO-1 expression induced by II/R in the lung tissues was promoted by ginsenoside Rb1 treatment. All these changes could be inhibited or prevented by ATRA. Conclusion. Ginsenoside Rb1 is capable of ameliorating II/R induced lung injuries by activating Nrf2/HO-1 pathway.
Collapse
|
28
|
Zhang X, He F, Yang J, Chen ZS. Protective effects of epigallocatechin-3-gallate on intestinal ischemia reperfusion injury through enhanced activation of PI3K/Akt pathway in rats. ACTA ACUST UNITED AC 2015; 35:378-383. [PMID: 26072077 DOI: 10.1007/s11596-015-1441-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/17/2014] [Indexed: 12/18/2022]
Abstract
Inflammation plays a critical role in intestinal ischemia reperfusion injury (IRI). Epigallocatechin-3-gallate (EGCG) has been demonstrated to possess anti-inflammatory effect. This study examined the effect of EGCG on intestinal IRI and explored the possible mechanisms. Male Wistar rats were randomly divided into three groups: sham-operated group (Sham), IRI control group (IRI) and IRI-EGCG group (EGCG). Rats in IRI-EGCG group were administered dissolved EGCG in drinking water (0.4 mg/mL) for 14 days prior to IRI induction. A rat model of intestinal IRI was established by ligating the superior mesenteric artery (SMA) for 30 min, followed by reperfusion for 1 h. Intestinal histology, pro-inflammatory cytokines and mediators were examined and the effect of EGCG on PI3K/Akt signalling was assessed. EGCG significantly alleviated the pathological changes of the intestine and suppressed the IRI-induced up-regulation of TNF-α, IL-1 and IL-6 mRNA and protein expression in the serum and intestine. The mechanism might be that EGCG enhanced the activation of PI3K/Akt signalling pathway. In conclusion, the administration of EGCG can significantly mitigate the acute intestinal IRI in rats by enhancing the activation of PI3K/Akt signalling pathway to suppress inflammatory response and might be a promising alternative for the prevention or treatment of intestinal IRI in the clinical practice.
Collapse
Affiliation(s)
- Xuan Zhang
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Institute of Organ Transplantation, Wuhan, 430030, China
| | - Fan He
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Institute of Organ Transplantation, Wuhan, 430030, China.,Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Yang
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Institute of Organ Transplantation, Wuhan, 430030, China
| | - Zhi-Shui Chen
- Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Institute of Organ Transplantation, Wuhan, 430030, China.
| |
Collapse
|
29
|
Ketamine reduces LPS-induced HMGB1 via activation of the Nrf2/HO-1 pathway and NF-κB suppression. J Trauma Acute Care Surg 2015; 78:784-92. [PMID: 25807407 DOI: 10.1097/ta.0000000000000588] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Ketamine, as an anesthetic agent, has an anti-inflammatory effect. In the present study, we investigated whether ketamine inhibits release of high mobility group box 1 (HMGB1), a late-phase cytokine of sepsis, in lipopolysaccharide (LPS)-stimulated macrophages through heme oxygenase-1 (HO-1) induction. METHODS Macrophages were preincubated with various concentrations of ketamine and then treated with LPS (1 μg/mL). The cell culture supernatants were collected to measure inflammatory mediators (HMGB1, nitric oxide, tumor necrosis factor-α, and interleukin 1β) by enzyme-linked immunosorbent assay. Moreover, HO-1 protein expression, the phosphorylation and degradation of IκB-α, and the nuclear translocation of nuclear factor E2-related factor 2 and nuclear factor κB (NF-κB) p65 were tested by Western blot analysis. In addition, to further identify the role of HO-1 in this process, tin protoporphyrin (SnPP), an HO-1 inhibitor, was used. RESULTS Ketamine treatment dose-dependently attenuated the increased levels of proinflammatory mediators (HMGB1, nitric oxide, tumor necrosis factor α, and interleukin 1β) and increased the HO-1 protein expression in LPS-activated macrophages. Furthermore, ketamine suppressed the phosphorylation and degradation of IκB-α as well as the LPS-stimulated nuclear translocation of NF-κB p65 in macrophages. In addition, the present study also demonstrated that ketamine induced HO-1 expression through the nuclear translocation of nuclear factor E2-related factor 2 in macrophages. The effects of ketamine on LPS-induced proinflammatory cytokines production were partially reversed by the HO inhibitor tin protoporphyrin (SnPP). CONCLUSION Ketamine inhibits the release of HMGB1 in LPS-stimulated macrophages, and this effect is at least partly mediated by the activation of the Nrf2/HO-1 pathway and NF-κB suppression.
Collapse
|
30
|
Sun D, Li C, Gao J, Li S, Wang H. Effects of selenium deficiency on principal indexes of chicken kidney function. Biol Trace Elem Res 2015; 164:58-63. [PMID: 25476001 DOI: 10.1007/s12011-014-0196-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 11/25/2014] [Indexed: 11/29/2022]
Abstract
Selenium (Se) deficiency leads to many pathological changes in animals. However, there have been very few reports regarding chicken tissue injury in the kidney caused by Se deficiency. In this study, a chicken Se-deficient disease model has been constructed, and two renal function indexes [including creatinine (CREA) and uric acid (URIC)], seven renal antioxidative function indexes [including glutathione peroxidase (GPx), anti-hydroxyl radical (AHR), catalase (CAT), hydrogen peroxide (H2O2), nitrogen monoxide (NO), glutathione (GSH), and malonyldialdehyde (MDA)], and two organ/tissue injury-related indexes [including inducible nitric oxide synthase (iNOS) and inducible heme oxygenase (HO)-1] were detected and analyzed to investigate the effects of Se deficiency on chicken kidney tissue. The results showed that Se deficiency caused a significant increase in CREA and URIC levels and a decrease in renal antioxidative capacity. Meanwhile, Se deficiency upregulated the expression of organ/tissue injury-related genes, such as the messenger RNA (mRNA) of HO-1 and iNOS as well as their protein expression levels, in the chicken kidney tissue. These data suggest that Se deficiency in birds triggers renal function regression and oxidative stress in the kidney tissue.
Collapse
Affiliation(s)
- Dongbo Sun
- College of Veterinary Medicine, Northeast Agricultural University, No. 59 Mucai Street, Xiangfang District, 150030, Harbin, People's Republic of China
| | | | | | | | | |
Collapse
|
31
|
Hebbel RP. Ischemia-reperfusion injury in sickle cell anemia: relationship to acute chest syndrome, endothelial dysfunction, arterial vasculopathy, and inflammatory pain. Hematol Oncol Clin North Am 2014; 28:181-98. [PMID: 24589261 DOI: 10.1016/j.hoc.2013.11.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ischemia-reperfusion (I/R) physiology, also called reperfusion injury, instigates vascular and tissue injury in human disease states. This review describes why sickle cell anemia should be conceptualized in this fashion and how I/R physiology explains the genesis of characteristic aspects of vascular pathobiology and clinical disease in sickle cell anemia. The nature of I/R and its relevance to sickle cell anemia are discussed, with an emphasis on the acute chest syndrome, endothelial dysfunction with aberrant vasoregulation, circle of Willis vasculopathy, and inflammatory pain. Viewing sickle disease from this perspective elucidates defining pathophysiology and identifies a host of novel potential therapeutic targets.
Collapse
Affiliation(s)
- Robert P Hebbel
- Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota Medical School, 420 Delaware Street South East, Mayo Mail Code 480, Minneapolis, MN 55455, USA.
| |
Collapse
|
32
|
Babu D, Motterlini R, Lefebvre RA. CO and CO-releasing molecules (CO-RMs) in acute gastrointestinal inflammation. Br J Pharmacol 2014; 172:1557-73. [PMID: 24641722 DOI: 10.1111/bph.12632] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/30/2014] [Accepted: 02/05/2014] [Indexed: 12/13/2022] Open
Abstract
Carbon monoxide (CO) is enzymatically generated in mammalian cells alongside the liberation of iron and the production of biliverdin and bilirubin. This occurs during the degradation of haem by haem oxygenase (HO) enzymes, a class of ubiquitous proteins consisting of constitutive and inducible isoforms. The constitutive HO2 is present in the gastrointestinal tract in neurons and interstitial cells of Cajal and CO released from these cells might contribute to intestinal inhibitory neurotransmission and/or to the control of intestinal smooth muscle cell membrane potential. On the other hand, increased expression of the inducible HO1 is now recognized as a beneficial response to oxidative stress and inflammation. Among the products of haem metabolism, CO appears to contribute primarily to the antioxidant and anti-inflammatory effects of the HO1 pathway explaining the studies conducted to exploit CO as a possible therapeutic agent. This article reviews the effects and, as far as known today, the mechanism(s) of action of CO administered either as CO gas or via CO-releasing molecules in acute gastrointestinal inflammation. We provide here a comprehensive overview on the effect of CO in experimental in vivo models of post-operative ileus, intestinal injury during sepsis and necrotizing enterocolitis. In addition, we will analyse the in vitro data obtained so far on the effect of CO on intestinal epithelial cell lines exposed to cytokines, considering the important role of the intestinal mucosa in the pathology of gastrointestinal inflammation.
Collapse
Affiliation(s)
- D Babu
- Heymans Institute of Pharmacology, Ghent University, Gent, Belgium
| | | | | |
Collapse
|
33
|
Fang T, Li J, Cao Z, Chen M, Shen W, Huang L. Heme oxygenase-1 is involved in sodium hydrosulfide-induced lateral root formation in tomato seedlings. PLANT CELL REPORTS 2014; 33:969-78. [PMID: 24556961 DOI: 10.1007/s00299-014-1577-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 01/23/2014] [Accepted: 01/28/2014] [Indexed: 05/09/2023]
Abstract
By using pharmacological and molecular approaches, we discovered the involvement of HO-1 in NaHS-induced lateral root formation in tomato seedlings. Heme oxygenase-1 (HO-1) and hydrogen sulfide (H2S) regulate various responses to abiotic stress and root development, but their involvement in the simultaneous regulation of plant lateral root (LR) formation is poorly understood. In this report, we observed that the exogenously applied H2S donor sodium hydrosulfide (NaHS) and the HO-1 inducer hemin induce LR formation in tomato seedlings by triggering intracellular signaling events involving the induction of tomato HO-1 (SlHO-1), and the modulation of cell cycle regulatory genes, including the up-regulation of SlCDKA;1 and SlCYCA2;1, and simultaneous down-regulation of SlKRP2. The response of NaHS in the induction of LR formation was impaired by the potent inhibition of HO-1, which was further blocked when 50 % saturation of carbon monoxide (CO) aqueous solution, one of the catalytic by-products of HO-1, was added. Further molecular evidence revealed that the NaHS-modulated gene expression of cell cycle regulatory genes was sensitive to the inhibition of HO-1 and reversed by cotreatment with CO. The impairment of LR density and length as well as lateral root primordia number, the decreased tomato HO-1 gene expression and HO activity caused by an H2S scavenger hypotaurine were partially rescued by the addition of NaHS, hemin and CO (in particular). Together, these results revealed that at least in our experimental conditions, HO-1 might be involved in NaHS-induced tomato LR formation. Additionally, the use of NaHS and hemin compounds in crop root organogenesis should be explored.
Collapse
Affiliation(s)
- Tao Fang
- College of Life Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | | | | | | | | | | |
Collapse
|
34
|
Wang Y, Wei S, Wang J, Fang Q, Chai Q. Phenethyl isothiocyanate inhibits growth of human chronic myeloid leukemia K562 cells via reactive oxygen species generation and caspases. Mol Med Rep 2014; 10:543-9. [PMID: 24788892 DOI: 10.3892/mmr.2014.2167] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 03/19/2014] [Indexed: 11/06/2022] Open
Abstract
Phenethyl isothiocyanate (PEITC), a potential cancer chemopreventive constituent of cruciferous vegetables, including watercress, has been reported to inhibit cancer cell growth by arresting the cell cycle and inducing apoptosis in various human cancer cell models. However, the role of PEITC in the inhibition of human chronic myeloid leukemia (CML) K562 cell growth and its underlying mechanisms have yet to be elucidated. In the present study, PEITC was found to induce cell death through the induction of reactive oxygen species (ROS) stress and oxidative damage. Heme oxygenase‑1 (HO‑1), which participates in the development of numerous tumors and the sensitivity of these tumors to chemotherapeutic drugs, plays a protective role by modulating oxidative injury. Therefore, the present study assessed the inhibitory effect of PEITC on K562 cells and whether HO‑1 facilitated cell apoptosis and ROS generation. PEITC was found to suppress cell growth and cause apoptosis by promoting Fas and Fas ligand expression, increasing ROS generation and by the successive release of cytochrome c as well as the activation of caspase‑9 and caspase‑3. PEITC was also combined with the HO‑1 inhibitor zinc protoporphyrin IX and the inducer hemin to assess whether HO‑1 determines cell survival and ROS generation. The results of the present study suggest that PEITC may be a potential anti‑tumor compound for CML therapy, and that HO‑1 has a critical function in PEITC‑induced apoptosis and ROS generation.
Collapse
Affiliation(s)
- Yating Wang
- Department of Hematology, First Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| | - Sixi Wei
- Department of Hematology, First Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| | - Jishi Wang
- Department of Hematology, First Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| | - Qin Fang
- Department of Pharmacy, First Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| | - Qixiang Chai
- Department of Hematology, First Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
35
|
Heme, an essential nutrient from dietary proteins, critically impacts diverse physiological and pathological processes. Nutrients 2014; 6:1080-102. [PMID: 24633395 PMCID: PMC3967179 DOI: 10.3390/nu6031080] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 02/14/2014] [Accepted: 02/19/2014] [Indexed: 12/11/2022] Open
Abstract
Heme constitutes 95% of functional iron in the human body, as well as two-thirds of the average person’s iron intake in developed countries. Hence, a wide range of epidemiological studies have focused on examining the association of dietary heme intake, mainly from red meat, with the risks of common diseases. High heme intake is associated with increased risk of several cancers, including colorectal cancer, pancreatic cancer and lung cancer. Likewise, the evidence for increased risks of type-2 diabetes and coronary heart disease associated with high heme intake is compelling. Furthermore, recent comparative metabolic and molecular studies of lung cancer cells showed that cancer cells require increased intracellular heme biosynthesis and uptake to meet the increased demand for oxygen-utilizing hemoproteins. Increased levels of hemoproteins in turn lead to intensified oxygen consumption and cellular energy generation, thereby fueling cancer cell progression. Together, both epidemiological and molecular studies support the idea that heme positively impacts cancer progression. However, it is also worth noting that heme deficiency can cause serious diseases in humans, such as anemia, porphyrias, and Alzheimer’s disease. This review attempts to summarize the latest literature in understanding the role of dietary heme intake and heme function in diverse diseases.
Collapse
|