1
|
Zuo R, Li R, Sun Z, Liu Y. Selenomethionine combined with allicin delays reactive oxidative stress-induced apoptosis, inflammation, endoplasmic reticulum stress, and barrier damage in IPEC-J2 cells via the GPX4 signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 295:118144. [PMID: 40215688 DOI: 10.1016/j.ecoenv.2025.118144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/11/2025] [Accepted: 04/01/2025] [Indexed: 04/21/2025]
Abstract
To increase livestock productivity and improve economic efficiency, farms tend to focus on the growth rate of livestock and poultry. This strategy can result in a reduced resistance to reactive oxidative stress (ROS) and heat stress. Selenomethionine (SeMet) and allicin have antioxidant properties, but their excessive intake can lead to toxicity. Co-administration improves antioxidant protection and reduces side-effects but also reduces the cost of administration. We undertook a study to elucidate the antioxidant effect of glutathione peroxidase (GPX) 4 in SeMet and allicin. The synergistic antioxidant effect, attenuation of endoplasmic reticulum stress (ERS), enhancement of expression of tight-junction proteins, and inhibition of apoptosis, ferroptosis, inflammatory responses of SeMet and allicin were attenuated significantly after inhibition of GPX4 according to western blotting (P < 0.05). These results indicated that activation of the GPX4 pathway was the key to the synergistic maintenance of barrier function, attenuation of ERS, as well as inhibition of apoptosis, ferroptosis, and inflammatory responses by SeMet and allicin. SeMet and allicin could protect the intestinal barrier from oxidative damage by synergistically activating the GPX4 pathway, increasing antioxidant capacity, and improving growth performance. In conclusion, SeMet and allicin could be used as a new drug combination to alleviate diseases associated with intestinal ROS and aid in the development of new antioxidant feed additives.
Collapse
Affiliation(s)
- Runan Zuo
- Animal-derived food safety innovation team, Pharmacology and Toxicology Laboratory, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, PR China; Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China
| | - Ruichao Li
- Animal-derived food safety innovation team, Pharmacology and Toxicology Laboratory, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, PR China
| | - Zeyuan Sun
- Animal-derived food safety innovation team, Pharmacology and Toxicology Laboratory, College of Veterinary Medicine, Anhui Agricultural University, Hefei 230036, PR China
| | - Yongshi Liu
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, PR China.
| |
Collapse
|
2
|
Ruhee RT, Ma S, Suzuki K. Effects of Sulforaphane Treatment on Skeletal Muscle from Exhaustive Exercise-Induced Inflammation and Oxidative Stress Through the Nrf2/HO-1 Signaling Pathway. Antioxidants (Basel) 2025; 14:210. [PMID: 40002396 PMCID: PMC11851896 DOI: 10.3390/antiox14020210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/31/2024] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Skeletal muscle is primarily involved in exercise performance and health promotion. Sulforaphane (SFN) is a naturally occurring isothiocyanate that indirectly activates the transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2), thus inducing the expression of Nrf2 target genes, including antioxidant enzymes. This study aimed to identify the effects of a single dose of SFN administration on exhaustive exercise-induced inflammation and oxidative stress in skeletal muscle tissue and elucidate the underlying mechanisms. Thirty-six mice were divided into four groups: control, SFN, exercise (Ex), and SFN + Ex. The SFN group and SFN + Ex group received SFN orally (50 mg/kg body weight) 2 h before the running test. Exercise significantly reduced plasma glucose levels, while the SFN-treated group exhibited a smaller reduction. Acute exhaustive exercise increased the expression of pro-inflammatory cytokines in muscle tissue, while the SFN + Ex group exhibited significantly reduced expression of pro-inflammatory cytokines. The gene expression of Nrf2 and its target enzymes, including heme oxygenase (HO)-1, superoxide dismutase (SOD)-1, catalase (CAT), and glutathione peroxidase (GPx)-1, was measured in the gastrocnemius and soleus muscle tissue. Compared with the Ex group, the SFN + Ex group showed upregulated expression of all these parameters, including Nrf2. SFN treatment reduced acute exhaustive exercise-induced oxidative stress and inflammation via activation of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Ruheea Taskin Ruhee
- Japan Society for the Promotion of Sciences, Chiyoda Ku 102-0083, Tokyo, Japan
| | - Sihui Ma
- Faculty of Human Sciences, Waseda University, Tokorozawa 359-1192, Japan;
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| |
Collapse
|
3
|
Lv W, Hu S, Yang F, Lin D, Zou H, Zhang W, Yang Q, Li L, Chen X, Wu Y. Heme oxygenase-1: potential therapeutic targets for periodontitis. PeerJ 2024; 12:e18237. [PMID: 39430558 PMCID: PMC11488498 DOI: 10.7717/peerj.18237] [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: 06/17/2024] [Accepted: 09/15/2024] [Indexed: 10/22/2024] Open
Abstract
Periodontitis is one of the most prevalent inflammatory disease worldwide, which affects 11% of the global population and is a major cause of tooth loss. Recently, oxidative stress (OS) has been found to be the pivital pathophysiological mechanism of periodontitis, and overactivated OS will lead to inflammation, apoptosis, pyroptosis and alveolar bone resorption. Interestingly, heme oxygenase-1 (HO-1), a rate-limiting enzyme in heme degradation, can exert antioxidant activites through its products-carbon monoxide (CO), Fe2+, biliverdin and bilirubin in the inflammatory microenvironment, thus exhibiting anti-inflammatory, anti-apoptotic, anti-pyroptosis and bone homeostasis-regulating properties. In this review, particular focus is given to the role of HO-1 in periodontitis, including the spatial-temporal expression in periodental tissues and pathophysiological mechanisms of HO-1 in periodontitis, as well as the current therapeutic applications of HO-1 targeted drugs for periodontitis. This review aims to elucidate the potential applications of various HO-1 targeted drug therapy in the management of periodontitis, investigate the influence of diverse functional groups on HO-1 and periodontitis, and pave the way for the development of a new generation of therapeutics that will benefit patients suffering from periodontitis.
Collapse
Affiliation(s)
- Weiwei Lv
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shichen Hu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Fei Yang
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Dong Lin
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Haodong Zou
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Wanyan Zhang
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qin Yang
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lihua Li
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaowen Chen
- School of Medical Imaging, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yan Wu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Stomatology, North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
4
|
Wei SM, Huang YM. Effect of sulforaphane on testicular ischemia-reperfusion injury induced by testicular torsion-detorsion in rats. Sci Rep 2024; 14:23420. [PMID: 39379457 PMCID: PMC11461801 DOI: 10.1038/s41598-024-74756-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
Testicular ischemia-reperfusion induces enhanced concentration of reactive oxygen species. The increased reactive oxygen species harm cellular lipids, nucleic acids, proteins, and carbohydrates, and ultimately cause testicular injury. Sulforaphane, a kind of natural dietary isothiocyanate, exists predominantly in some cruciferous vegetables, like broccoli and cabbage. It can protect tissues from oxidative stress-induced damage. Herein, we analyzed the effectiveness of sulforaphane in treating ischemia-reperfusion injury occurring after testicular torsion-detorsion. Male rats (n = 60) were grouped as follows: sham-operated group, unilateral testicular ischemia-reperfusion group, and unilateral testicular ischemia-reperfusion group receiving sulforaphane treatment at 5 mg/kg. No testicular torsion-detorsion was performed in the sham group. Unilateral testicular ischemia-reperfusion model was created by detorsion after 2 h of left testicular torsion. In the sulforaphane-treated group, intraperitoneal sulforaphane (5 mg/kg) was administered at left testicular detorsion. Biochemical assay, Western blot, and hematoxylin and eosin staining were used to evaluate testicular malondialdehyde content (an important marker of reactive oxygen species), protein levels of superoxide dismutase and catalase (intracellular antioxidant defense mechanism), and testicular reproductive function, respectively. In testicular tissues, malondialdehyde content was significantly promoted, while protein levels of superoxide dismutase and catalase, and testicular reproductive function were significantly reduced in ipsilateral testes by testicular ischemia-reperfusion. Nevertheless, sulforaphane administration partially reversed the effect of testicular ischemia-reperfusion on these indexes. It can be concluded that sulforaphane elevates protein levels of superoxide dismutase and catalase, and suppresses reactive oxygen species content, thereby preventing ischemia-reperfusion injury in testis.
Collapse
Affiliation(s)
- Si-Ming Wei
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou City, 310015, Zhejiang Province, China.
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou City, 310053, Zhejiang Province, China.
| | - Yu-Min Huang
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou City, 310058, Zhejiang Province, China
| |
Collapse
|
5
|
Wang B, Zhang C, Ma J, Wang Y, Zhang L, Yang X, Jia T, Zhang K, Zhang Q. Protective Role of Sulforaphane against Physiological Toxicity of Triphenyltin in Common Carp ( Cyprinus carpio haematopterus). Antioxidants (Basel) 2024; 13:1173. [PMID: 39456427 PMCID: PMC11504319 DOI: 10.3390/antiox13101173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
This experiment mainly explored the protective role of sulforaphane (SFN) against physiological toxicity of triphenyltin (TPT) in Cyprinus carpio haematopterus. In total, 320 Fish (56.90 ± 0.40 g) were randomly divided into four groups with four replicates each. The control group was fed the basal diet, the TPT group (TPT) was exposed to 10 ng L-1 TPT on the basis of the control group, the SFN + TPT group (TPT + SFN) was fed a diet supplemented with 10 mg kg-1 SFN on the TPT group, and the SFN group (SFN) was fed a diet supplemented with 10 mg kg-1 SFN. After 56 days of breeding trials, the results showed that TPT exposure resulted in a remarkable decrease (p < 0.05) in final weight, weight gain rate (WGR), specific growth rate (SGR), and condition factor (CF), but an increase (p < 0.05) in feed conversion ratio (FCR) and hepatosomatic index (HSI) of fish. TPT treatment decreased (p < 0.05) the amounts of hematocrit (Hct) and hemoglobin (Hb), plasma complement component 3 (C3) and C4 contents, alternative complement pathway (ACH50), acid phosphatase (ACP) and lysozyme (LZM) activities, liver glutathione (GSH) content, catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPX) activities, interleukin 10 (IL-10), and SOD mRNA expressions, but increased (p < 0.05) plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, liver malonaldehyde (MDA) content, tumor Cyclooxygenase 2 (COX2), and necrosis factor α (TNFα), IL-1β, and MDA mRNA expressions. A histological analysis of the liver showed that a higher occurrence rates of the hepatocyte hypertrophy, nuclear disappearance and hepatocyte vacuolization was observed in the hepatocytes of fish exposed to TPT, and it was accompanied by the dilation of hepatic sinusoids. In addition, the toxicity induced by TPT was significantly improved in the groups that were treated with SFN, and SFN was able to improve growth performance and immunity, alleviate TPT-induced changes in inflammatory factors, ameliorate oxidative stress, and increase the activity of antioxidant enzymes (p < 0.05). The addition of SFN also alleviated liver damage caused by TPT and protected the structural integrity of the liver. Overall, these findings suggest that TPT inhibited the growth, immunity, and antioxidant capacity of Cyprinus carpio haematopteru. Dietary SFN could be beneficial for growth promotion, immunity, antioxidant capacity, and protection of liver structural integrity. Therefore, SFN is a prospective feed supplement for ameliorating the damage caused to fish by TPT contamination.
Collapse
Affiliation(s)
- Bingke Wang
- Henan Academy of Fishery Sciences, Zhengzhou 450044, China; (B.W.)
- Henan Fishery Engineering Technology Research Center, Zhengzhou 450044, China
| | - Chunnuan Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
| | - Jianshuang Ma
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China
| | - Yanhui Wang
- Henan Academy of Fishery Sciences, Zhengzhou 450044, China; (B.W.)
- Henan Fishery Engineering Technology Research Center, Zhengzhou 450044, China
| | - Ling Zhang
- Henan Academy of Fishery Sciences, Zhengzhou 450044, China; (B.W.)
- Henan Fishery Engineering Technology Research Center, Zhengzhou 450044, China
| | - Xingli Yang
- Henan Academy of Fishery Sciences, Zhengzhou 450044, China; (B.W.)
- Henan Fishery Engineering Technology Research Center, Zhengzhou 450044, China
| | - Tao Jia
- Henan Academy of Fishery Sciences, Zhengzhou 450044, China; (B.W.)
- Henan Fishery Engineering Technology Research Center, Zhengzhou 450044, China
| | - Kaisong Zhang
- Henan Academy of Fishery Sciences, Zhengzhou 450044, China; (B.W.)
- Henan Fishery Engineering Technology Research Center, Zhengzhou 450044, China
| | - Qin Zhang
- Henan Academy of Fishery Sciences, Zhengzhou 450044, China; (B.W.)
- Henan Fishery Engineering Technology Research Center, Zhengzhou 450044, China
| |
Collapse
|
6
|
Habtemariam S. Anti-Inflammatory Therapeutic Mechanisms of Isothiocyanates: Insights from Sulforaphane. Biomedicines 2024; 12:1169. [PMID: 38927376 PMCID: PMC11200786 DOI: 10.3390/biomedicines12061169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Isothiocyanates (ITCs) belong to a group of natural products that possess a highly reactive electrophilic -N=C=S functional group. They are stored in plants as precursor molecules, glucosinolates, which are processed by the tyrosinase enzyme upon plant tissue damage to release ITCs, along with other products. Isolated from broccoli, sulforaphane is by far the most studied antioxidant ITC, acting primarily through the induction of a transcription factor, the nuclear factor erythroid 2-related factor 2 (Nrf2), which upregulates downstream antioxidant genes/proteins. Paradoxically, sulforaphane, as a pro-oxidant compound, can also increase the levels of reactive oxygen species, a mechanism which is attributed to its anticancer effect. Beyond highlighting the common pro-oxidant and antioxidant effects of sulforaphane, the present paper was designed to assess the diverse anti-inflammatory mechanisms reported to date using a variety of in vitro and in vivo experimental models. Sulforaphane downregulates the expression of pro-inflammatory cytokines, chemokines, adhesion molecules, cycloxyhenase-2, and inducible nitric oxide synthase. The signalling pathways of nuclear factor κB, activator protein 1, sirtuins 1, silent information regulator sirtuin 1 and 3, and microRNAs are among those affected by sulforaphane. These anti-inflammatory actions are sometimes due to direct action via interaction with the sulfhydryl structural moiety of cysteine residues in enzymes/proteins. The following are among the topics discussed in this paper: paradoxical signalling pathways such as the immunosuppressant or immunostimulant mechanisms; crosstalk between the oxidative and inflammatory pathways; and effects dependent on health and disease states.
Collapse
Affiliation(s)
- Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| |
Collapse
|
7
|
Tie H, Kuang G, Gong X, Zhang L, Zhao Z, Wu S, Huang W, Chen X, Yuan Y, Li Z, Li H, Zhang L, Wan J, Wang B. LXA4 protected mice from renal ischemia/reperfusion injury by promoting IRG1/Nrf2 and IRAK-M-TRAF6 signal pathways. Clin Immunol 2024; 261:110167. [PMID: 38453127 DOI: 10.1016/j.clim.2024.110167] [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: 09/24/2023] [Revised: 01/26/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
Excessive inflammatory response and increased oxidative stress play an essential role in the pathophysiology of ischemia/reperfusion (I/R)-induced acute kidney injury (IRI-AKI). Emerging evidence suggests that lipoxin A4 (LXA4), as an endogenous negative regulator in inflammation, can ameliorate several I/R injuries. However, the mechanisms and effects of LXA4 on IRI-AKI remain unknown. In this study, A bilateral renal I/R mouse model was used to evaluate the role of LXA4 in wild-type, IRG1 knockout, and IRAK-M knockout mice. Our results showed that LXA4, as well as 5-LOX and ALXR, were quickly induced, and subsequently decreased by renal I/R. LXA4 pretreatment improved renal I/R-induced renal function impairment and renal damage and inhibited inflammatory responses and oxidative stresses in mice kidneys. Notably, LXA4 inhibited I/R-induced the activation of TLR4 signal pathway including decreased phosphorylation of TAK1, p36, and p65, but did not affect TLR4 and p-IRAK-1. The analysis of transcriptomic sequencing data and immunoblotting suggested that innate immune signal molecules interleukin-1 receptor-associated kinase-M (IRAK-M) and immunoresponsive gene 1 (IRG1) might be the key targets of LXA4. Further, the knockout of IRG1 or IRAK-M abolished the beneficial effects of LXA4 on IRI-AKI. In addition, IRG1 deficiency reversed the up-regulation of IRAK-M by LXA4, while IRAK-M knockout had no impact on the IRG1 expression, indicating that IRAK-M is a downstream molecule of IRG1. Mechanistically, we found that LXA4-promoted IRG1-itaconate not only enhanced Nrf2 activation and increased HO-1 and NQO1, but also upregulated IRAK-M, which interacted with TRAF6 by competing with IRAK-1, resulting in deactivation of TLR4 downstream signal in IRI-AKI. These data suggested that LXA4 protected against IRI-AKI via promoting IRG1/Itaconate-Nrf2 and IRAK-M-TRAF6 signaling pathways, providing the rationale for a novel strategy for preventing and treating IRI-AKI.
Collapse
Affiliation(s)
- Hongtao Tie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Ge Kuang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Lidan Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zizuo Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengwang Wu
- Department of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wenya Huang
- Yiling Women and Children's Hospital of Yichang City, Hubei, China
| | - Xiahong Chen
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yinglin Yuan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenhan Li
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University; Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China..
| | - Bin Wang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
8
|
Hasan GM, Anwar S, Shamsi A, Sohal SS, Hassan MI. The neuroprotective potential of phytochemicals in traumatic brain injury: mechanistic insights and pharmacological implications. Front Pharmacol 2024; 14:1330098. [PMID: 38239205 PMCID: PMC10794744 DOI: 10.3389/fphar.2023.1330098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024] Open
Abstract
Traumatic brain injury (TBI) leads to brain damage, comprising both immediate primary damage and a subsequent cascade of secondary injury mechanisms. The primary injury results in localized brain damage, while the secondary damage initiates inflammatory responses, followed by the disruption of the blood-brain barrier, infiltration of peripheral blood cells, brain edema, and the release of various immune mediators, including chemotactic factors and interleukins. TBI disrupts molecular signaling, cell structures, and functions. In addition to physical tissue damage, such as axonal injuries, contusions, and haemorrhages, TBI interferes with brain functioning, impacting cognition, decision-making, memory, attention, and speech capabilities. Despite a deep understanding of the pathophysiology of TBI, an intensive effort to evaluate the underlying mechanisms with effective therapeutic interventions is imperative to manage the repercussions of TBI. Studies have commenced to explore the potential of employing natural compounds as therapeutic interventions for TBI. These compounds are characterized by their low toxicity and limited interactions with conventional drugs. Moreover, many natural compounds demonstrate the capacity to target various aspects of the secondary injury process. While our understanding of the pathophysiology of TBI, there is an urgent need for effective therapeutic interventions to mitigate its consequences. Here, we aimed to summarize the mechanism of action and the role of phytochemicals against TBI progression. This review discusses the therapeutic implications of various phytonutrients and addresses primary and secondary consequences of TBI. In addition, we highlighted the roles of emerging phytochemicals as promising candidates for therapeutic intervention of TBI. The review highlights the neuroprotective roles of phytochemicals against TBI and the mechanistic approach. Furthermore, our efforts focused on the underlying mechanisms, providing a better understanding of the therapeutic potential of phytochemicals in TBI therapeutics.
Collapse
Affiliation(s)
- Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
9
|
Hassanein EHM, Ibrahim IM, Abd-Alhameed EK, Sharawi ZW, Jaber FA, Althagafy HS. Nrf2/HO-1 as a therapeutic target in renal fibrosis. Life Sci 2023; 334:122209. [PMID: 37890696 DOI: 10.1016/j.lfs.2023.122209] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
Chronic kidney disease (CKD) is one of the most prevalent chronic diseases and affects between 10 and 14 % of the world's population. The World Health Organization estimates that by 2040, the disease will be fifth in prevalence. End-stage CKD is characterized by renal fibrosis, which can eventually lead to kidney failure and death. Renal fibrosis develops due to multiple injuries and involves oxidative stress and inflammation. In the human body, nuclear factor erythroid 2-related factor 2 (Nrf2) plays an important role in the expression of antioxidant, anti-inflammatory, and cytoprotective genes, which prevents oxidative stress and inflammation damage. Heme oxygenase (HO-1) is an inducible homolog influenced by heme products and after exposure to cellular stress inducers such as oxidants, inflammatory chemokines/cytokines, and tissue damage as an outcome or downstream of Nrf2 activation. HO-1 is known for its antioxidative properties, which play an important role in regulating oxidative stress. In renal diseases-induced tissue fibrosis and xenobiotics-induced renal fibrosis, Nrf2/HO-1 has been targeted with promising results. This review summarizes these studies and highlights the interesting bioactive compounds that may assist in attenuating renal fibrosis mediated by HO-1 activation. In conclusion, Nrf2/HO-1 signal activation could have a renoprotective effect strategy against CKD caused by oxidative stress, inflammation, and consequent renal fibrosis.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt.
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Zeina W Sharawi
- Biological Sciences Department, Faculty of Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Fatima A Jaber
- Department of Biology, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Saudi Arabia
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
10
|
Yan L, Yan Y. Therapeutic potential of sulforaphane in liver diseases: a review. Front Pharmacol 2023; 14:1256029. [PMID: 37705537 PMCID: PMC10495681 DOI: 10.3389/fphar.2023.1256029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023] Open
Abstract
The burden of liver diseases such as metabolic-associated fatty liver diseases and hepatocellular carcinoma has increased rapidly worldwide over the past decades. However, pharmacological therapies for these liver diseases are insufficient. Sulforaphane (SFN), an isothiocyanate that is mainly found in cruciferous vegetables, has been found to have a broad spectrum of activities like antioxidation, anti-inflammation, anti-diabetic, and anticancer effects. Recently, a growing number of studies have reported that SFN could significantly ameliorate hepatic steatosis and prevent the development of fatty liver, improve insulin sensitivity, attenuate oxidative damage and liver injury, induce apoptosis, and inhibit the proliferation of hepatoma cells through multiple signaling pathways. Moreover, many clinical studies have demonstrated that SFN is harmless to the human body and well-tolerated by individuals. This emerging evidence suggests SFN to be a promising drug candidate in the treatment of liver diseases. Nevertheless, limitations exist in the development of SFN as a hepatoprotective drug due to its special properties, including instability, water insolubility, and high inter-individual variation of bioavailability when used from broccoli sprout extracts. Herein, we comprehensively review the recent progress of SFN in the treatment of common liver diseases and the underlying mechanisms, with the aim to provide a better understanding of the therapeutic potential of SFN in liver diseases.
Collapse
Affiliation(s)
- Liang Yan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | |
Collapse
|
11
|
Sulforaphane alleviated vascular remodeling in hypoxic pulmonary hypertension via inhibiting inflammation and oxidative stress. J Nutr Biochem 2023; 111:109182. [PMID: 36220525 DOI: 10.1016/j.jnutbio.2022.109182] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/05/2022]
Abstract
Hypoxic pulmonary hypertension (HPH) is a cardiopulmonary disease featured by pulmonary vascular remodeling, which is due to abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) and dysfunction of endothelial cells (ECs). Sulforaphane (SFN) is a natural isothiocyanate extracted from cruciferous vegetables with promising anti-inflammatory and anti-oxidative activities. This study aimed to explore the effect and mechanism of SFN on HPH. Male mice were exposed to persistent chronic hypoxia for 4 weeks to induce HPH. The results demonstrated that SFN repressed the increased right ventricular systolic pressure (RVSP) and attenuated the right ventricular hypertrophy and pulmonary arteries remodeling in HPH mice. In particular, after SFN treatment, the CD68 positive cells in lung sections were reduced; TNF-α and IL-6 levels in lungs and serum declined; activation of NF-κB in PASMCs was inhibited in response to hypoxia. Besides, SFN enhanced the superoxide dismutase (SOD) activity in serum, SOD2 expression, total glutathione levels, and GSH/GSSG ratio in PASMCs, along with a decrease in malondialdehyde (MDA) contents in serum and ROS production in PASMCs after hypoxia exposure. Notably, SFN, as an Nrf2 activator, reversed the reduction in Nrf2 expression in hypoxic PASMCs. In vitro, SFN treatment inhibited hyperproliferation and promoted apoptosis of PASMCs under hypoxia conditions. SFN also prevented the apoptosis of pulmonary microvascular ECs caused by hypoxia. Therefore, these data suggested that SFN could significantly restrain the inflammation and oxidative stress, thereby inhibiting PASMCs proliferation, promoting PASMCs apoptosis, and reversing hypoxia injury in ECs to improve pulmonary vascular remodeling.
Collapse
|
12
|
Cong P, Tong C, Mao S, Shi X, Liu Y, Shi L, Jin H, Liu Y, Hou M. Proteomic global proteins analysis in blast lung injury reveals the altered characteristics of crucial proteins in response to oxidative stress, oxidation-reduction process and lipid metabolic process. Exp Lung Res 2022; 48:275-290. [PMID: 36346360 DOI: 10.1080/01902148.2022.2143596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background: Blast lung injury (BLI) is the most common fatal blast injury induced by overpressure wave in the events of terrorist attack, gas and underground explosion. Our previous work revealed the characteristics of inflammationrelated key proteins involved in BLI, including those regulating inflammatory response, leukocyte transendothelial migration, phagocytosis, and immune process. However, the molecular characteristics of oxidative-related proteins in BLI ar still lacking. Methods: In this study, protein expression profiling of the blast lungs obtained by tandem mass tag (TMT) spectrometry quantitative proteomics were re-analyzed to identify the characteristics of oxidative-related key proteins. Forty-eight male C57BL/6 mice were randomly divided into six groups: control, 12 h, 24 h, 48 h, 72 h and 1 w after blast exposure. The differential protein expression was identified by bioinformatics analysis and verified by western blotting. Results: The results demonstrated that thoracic blast exposure induced reactive oxygen species generation and lipid peroxidation in the lungs. Analysis of global proteins and oxidative-related proteomes showed that 62, 59, 73, 69, 27 proteins (accounted for 204 distinct proteins) were identified to be associated with oxidative stress at 12 h, 24 h, 48 h, 72 h, and 1 week after blast exposure, respectively. These 204 distinct proteins were mainly enriched in response to oxidative stress, oxidation-reduction process and lipid metabolic process. We also validated these results by western blotting. Conclusions: These findings provided new perspectives on blast-induced oxidative injury in lung, which may potentially benefit the development of future treatment of BLI.
Collapse
Affiliation(s)
- Peifang Cong
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning Province, China
| | - Changci Tong
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning Province, China
| | - Shun Mao
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning Province, China
| | - Xiuyun Shi
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, Liaoning Province, China
| | - Ying Liu
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, Liaoning Province, China
| | - Lin Shi
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning Province, China
| | - Hongxu Jin
- Emergency Medicine Department of General Hospital of Northern theatre command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, Liaoning Province, China
| | - Yunen Liu
- Shuren International College, Shenyang Medical College, Shenyang, Liaoning Province, China
| | - Mingxiao Hou
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning Province, China.,Shuren International College, Shenyang Medical College, Shenyang, Liaoning Province, China.,The Second Affiliated Hospital of Shenyang Medical College, The Veterans General Hospital of Liaoning Province, Shenyang, Liaoning Province, China
| |
Collapse
|
13
|
Hui B, Shu Y, Yang D, Wang Z, Zhang L, Lei N, Yang Z. Sinomenine pretreatment alleviates hepatic ischemia/reperfusion injury through activating Nrf-2/HO-1 pathway. Immun Inflamm Dis 2022; 10:e700. [PMID: 36169257 PMCID: PMC9517062 DOI: 10.1002/iid3.700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Ischemia-reperfusion (IR) injury is induced by an interrupted blood flow and succeeding blood restoration, which is common in the operation of liver transplantation. Serious IR injury is a major reason leading to transplant failure. Hepatic IR is featured by excessive inflammatory response, oxidative stress, and apoptosis. Sinomenine (SIN) is derived from the herb Sinomeniumacutum and shows properties of anti-inflammation and antiapoptosis in multiple IR-induced organ injuries. However, the effect of SIN in hepatic IR has not been investigated. METHODS This study aims to investigate impacts of SIN on hepatic IR and the involved signaling pathway. An in vivo rat model of syngeneic orthotopic liver transplantation was constructed to induce the hepatic IR injury. RESULTS Results showed that SIN pretreatment provided a significant prevention against IR-induced hepatic injury as manifested by the downregulated activities of serum alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase, the alleviatedoxidative stress as shown by increased activities of serum superoxide dismutase and glutathione peroxidase, and decreased serum level of malondialdehyde, the suppressed inflammatory responses as shown by downregulated serum tumor necrosis factor-α, interleukin (IL)-6, IL-8 levels, and upregulated IL-10 level, as well as attenuated apoptosis as shown by decreased protein expression of cleaved caspase-3 and -9. In line with these results, SIN pretreatment also alleviatedthe hepatic histopathological changes in IR rats and induced Nrf-2/HO-1 activation. The use of brusatol, a selective inhibitor for Nrf-2, effectively reversed SIN-induced above effects. CONCLUSIONS Altogether, our results demonstrate that SIN might be a useful therapeutic drug for preventing hepatic IR-induced injury during clinical liver transplantation.
Collapse
Affiliation(s)
- Bo Hui
- Department of General Surgery Unit‐4The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Yantao Shu
- Department of General Surgery Unit‐4The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Dandan Yang
- Department of General Surgery Unit‐4The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Zhidong Wang
- Department of General Surgery Unit‐4The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Li Zhang
- Department of General Surgery Unit‐4The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Nina Lei
- Department of General Surgery Unit‐4The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Zhengan Yang
- Department of General Surgery Unit‐4The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
14
|
Dana AH, Alejandro SP. Role of sulforaphane in endoplasmic reticulum homeostasis through regulation of the antioxidant response. Life Sci 2022; 299:120554. [PMID: 35452639 DOI: 10.1016/j.lfs.2022.120554] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 02/09/2023]
Abstract
Nowadays, the nutraceutical agent sulforaphane (SFN) shows great versatility in turning on different cellular responses. Mainly, this isothiocyanate acts as a master regulator of cellular homeostasis due to its antioxidant response and cytoplasmic, mitochondrial, and endoplasmic reticulum (ER) protein modulation. Even more, SFN acts as an effective strategy to counteract oxidative stress, apoptosis, and ER stress, among others as seen in different injury models. Particularly, ER stress is buffered by the unfolded protein response (UPR) activation, which is the first instance in orchestrating the recovery of ER function. Interestingly, different studies highlight a close interrelationship between ER stress and oxidative stress, two events driven by the accumulation of reactive oxygen species (ROS). This response inevitably perpetuates itself and acts as a vicious cycle that triggers the development of different pathologies, such as cardiovascular diseases, neurodegenerative diseases, and others. Accordingly, it is vital to target ER stress and oxidative stress to increase the effectiveness of clinical therapies used to treat these diseases. Therefore, our study is focused on the role of SFN in preserving cellular homeostasis balance by regulating the ER stress response through the Nrf2-modulated antioxidant pathway.
Collapse
Affiliation(s)
- Arana-Hidalgo Dana
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Silva-Palacios Alejandro
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|
15
|
Du P, Zhang X, Luo K, Li Y, Fu C, Xiao J, Xiao Q. Curculigoside mitigates hepatic ischemia/reperfusion-induced oxidative stress, inflammation, and apoptosis via activation of the Nrf-2/HO-1 pathway. Hum Exp Toxicol 2022; 41:9603271221087146. [PMID: 35331031 DOI: 10.1177/09603271221087146] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Curculigoside has been shown to decrease oxidative stress and inflammatory reactions in many disorders, but its effects during hepatic ischemia-reperfusion injury (IRI) remain unknown. This research aims to determine the protective role and the potential mechanism of action of curculigoside in hepatic IRI. Here, a well-established rat model of partial warm IRI was constructed; serum ALT/AST and H&E staining were employed to assay the extent of liver injury; the superoxide dismutase, malondialdehyde, IL-6, and TNF-α contents were determined using the corresponding kits; the apoptosis index was evaluated by TUNEL staining; and the expression of Nrf-2, HO-1, and apoptosis-associated proteins was detected by qRT-PCR and Western blotting. The results showed that curculigoside pretreatment effectively mitigated hepatic IRI, as demonstrated by decreases in the levels of serum aminotransferases, hepatocellular necrosis and apoptosis, oxidative stress markers, infiltration of inflammatory cells, and secretion of proinflammatory cytokines. Mechanistically, the expression of Nrf-2 and HO-1 was greatly suppressed by hepatic IRI and reactivated by curculigoside. Furthermore, cotreatment with ML-385, an inhibitor of Nrf-2, counteracted the protective effect of curculigoside against hepatic IRI. The results of our study show that curculigoside plays a protective role in hepatic IRI by inhibiting oxidative stress, inflammation, and apoptosis and that its effects may be associated with activation of the Nrf-2/HO-1 pathway.
Collapse
Affiliation(s)
- Peng Du
- Department of General Surgery, 117970The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xingjian Zhang
- Department of General Surgery, 117970The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kaifeng Luo
- Department of General Surgery, 117970The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yong Li
- Department of General Surgery, 117970The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chengchao Fu
- Department of General Surgery, 117970The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiansheng Xiao
- Department of General Surgery, 117970The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qi Xiao
- Department of General Surgery, 117970The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|