1
|
Zhao A, Zhang G, Wei H, Yan X, Gan J, Jiang X. Heat shock proteins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic implications. Exp Neurol 2025; 390:115284. [PMID: 40318821 DOI: 10.1016/j.expneurol.2025.115284] [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: 03/03/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) remains a significant challenge in ischemic stroke treatment. Heat shock proteins (HSPs), a cadre of molecular chaperones, have emerged as pivotal regulators in this pathological cascade. This review synthesizes the latest research on HSPs in CIRI from 2013 to 2024 focusing on their multifaceted roles and therapeutic potential. We explore the diverse cellular functions of HSPs, including regulation of oxidative stress, apoptosis, necroptosis, ferroptosis, autophagy, neuroinflammation, and blood-brain barrier integrity. Key HSPs, such as HSP90, HSP70, HSP32, HSP60, HSP47, and small HSPs, are investigated for their specific mechanisms of action in CIRI. Potential therapeutic strategies targeting HSPs, including HSP inhibitors, traditional Chinese medicine components, and gene therapy, are discussed. This review provides a comprehensive understanding of HSPs in CIRI and offers insights into the development of innovative neuroprotective treatments.
Collapse
Affiliation(s)
- Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
2
|
Wilkinson T, Fletcher E, Ring A, Bradley C, Papoutsi E, Miserlis D, Smith RS, Bohannon WT, Pipinos II, Koutakis P. Deciphering the Role of Ferroptosis in the Pathogenesis of Peripheral Artery Disease Myopathy. BIOLOGY 2025; 14:537. [PMID: 40427725 PMCID: PMC12108827 DOI: 10.3390/biology14050537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
This study investigates ferroptosis in the context of peripheral artery disease (PAD), a vascular disease characterized by atherosclerosis of the lower extremities. Muscle atrophy and increased oxidative stress are hallmarks of PAD and correlate with worse clinical outcomes. Given ferroptosis' association with oxidative stress, we explored its role in PAD myopathy by examining gene and protein markers related to metabolic pathways implicated in ferroptosis using both human PAD patients and cultured myotubes. Intermittent claudication (IC) PAD patients, critical limb ischemia (CLI) PAD patients, and non-PAD controls were recruited for this study. Calf muscle biopsies were analyzed for gene expression using qPCR, and protein levels were determined by Western blotting. Cultured myotubes treated with the ferroptosis inducer erastin provided an in vitro comparison. Results demonstrated upregulation of ferroptosis markers such as lipid peroxidation and PTGS2 gene expression in the muscle of CLI PAD patients compared to controls. Increased expression of ferroptosis-related genes HMOX1, ACSL4, ELAVL1, and Beclin-1 was also observed. Protein analysis showed trends consistent with gene expression in some ferroptosis markers. The increase in ferroptosis markers in CLI PAD patients, particularly in iron metabolism and autophagy pathways, suggests ferroptosis contributes to PAD myopathy.
Collapse
Affiliation(s)
| | - Emma Fletcher
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
| | - Andrew Ring
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | | | - Evlampia Papoutsi
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
| | - Dimitrios Miserlis
- Department of Surgery and Perioperative Care, Dell Medical School, University of Texas, Austin, TX 78712, USA
| | - Robert S. Smith
- Department of Surgery, Baylor Scott and White Medical Center, Temple, TX 76508, USA
| | - William T. Bohannon
- Department of Surgery, Baylor Scott and White Medical Center, Temple, TX 76508, USA
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, Waco, TX 76798, USA
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
3
|
Arif HM, Fu M, Wang R. Hydrogen Sulfide (H 2S) Metabolism, Iron Overload, and Apoptosis-Autophagy Equilibrium in Vascular Smooth Muscle Cells. Antioxidants (Basel) 2025; 14:560. [PMID: 40427442 PMCID: PMC12108171 DOI: 10.3390/antiox14050560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Iron overload contributes to proliferative vascular diseases, yet its interplay with hydrogen sulfide (H2S) in vascular smooth muscle cell (VSMC) proliferation remains poorly understood. This study elucidates H2S's role in mitigating iron-overload-induced oxidative stress and cellular damage. Using aortic VSMCs from wildtype (WT) and cystathionine γ-lyase-knockout (CSE-KO) mice treated with ferric ammonium citrate (FAC) at concentrations equivalent to serum levels of iron and citrate, we demonstrate that FAC triggers the integrated stress response (ISR) in WT cells, upregulating CSE to enhance H2S production. The ISR mediator ATF4 activates caspases but simultaneously induces CSE, which inhibits caspase activity and promotes autophagy via AMPK signaling. In CSE-KO cells, iron overload leads to diminished Ferritin upregulation, unchecked Caspase activation, and impaired autophagy compared to WT cells. Exogenous H2S restored iron homeostasis by enhancing Ferritin expression, activating NRF2 antioxidant pathways, and restoring apoptosis-autophagy equilibrium in both WT and KO cells. These findings establish H2S as a critical regulator of iron-induced VSMC dysfunction, highlighting its therapeutic potential in managing vascular pathologies linked to iron dysregulation.
Collapse
Affiliation(s)
- Hassan Mustafa Arif
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (H.M.A.); (M.F.)
| | - Ming Fu
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (H.M.A.); (M.F.)
- College of Basic Medicine, Shandong Second Medical University, Weifang 261053, China
| | - Rui Wang
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (H.M.A.); (M.F.)
- College of Basic Medicine, Shandong Second Medical University, Weifang 261053, China
| |
Collapse
|
4
|
Lindsay RT, Rhodes CJ. Reactive Oxygen Species (ROS) in Metabolic Disease-Don't Shoot the Metabolic Messenger. Int J Mol Sci 2025; 26:2622. [PMID: 40141264 PMCID: PMC11942130 DOI: 10.3390/ijms26062622] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Reactive oxygen species (ROS) are widely considered key to pathogenesis in chronic metabolic disease. Consequently, much attention is rightly focused on minimising oxidative damage. However, for ROS production to be most effectively modulated, it is crucial to first appreciate that ROS do not solely function as pathological mediators. There are >90 gene products specifically evolved to generate, handle, and tightly buffer the cellular concentration of ROS. Therefore, it is likely that ROS plays a role as integral homeostatic signalling components and only become toxic in extremis. This review explores these commonly overlooked normal physiological functions, including how ROS are generated in response to environmental or hormonal stimuli, the mechanisms by which the signals are propagated and regulated, and how the cell effectively brings the signal to an end after an appropriate duration. In the course of this, several specific and better-characterised signalling mechanisms that rely upon ROS are explored, and the threshold at which ROS cross from beneficial signalling molecules to pathology mediators is discussed.
Collapse
Affiliation(s)
- Ross T. Lindsay
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Christopher J. Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| |
Collapse
|
5
|
Liu XM, Li Z, Wang XY, Ding BW, Wang JQ, Qiao X, Feng YK, Hao JH, Xu JY. Self-assembled HO-1i-Pt(IV) nanomedicine targeting p38/MAPK and MDR pathways for cancer chemo-immunotherapy. J Control Release 2025; 379:797-813. [PMID: 39848589 DOI: 10.1016/j.jconrel.2025.01.050] [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: 09/05/2024] [Revised: 01/08/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
Platinum(II)-based antitumor drugs are widely used in clinics but limited by severe side effects and resistance. Multi-target Platinum(IV) complexes are emerging as ideal alternatives. Heme oxygenase-1 (HO-1) works as a rate-limiting step in heme degradation and is overexpressed in malignant tumors. Herein, HO-1i-based Platinum(IV) prodrugs are prepared and candidate complex 15 is further developed into self-assembled nanoparticles (15-NPs). 15 and 15-NPs significantly increase cytotoxicity, particularly in HepG2 (74.77- and 96.14-fold increases) and A549cisR (38.6- and 47.24-fold increases), while reducing toxicity towards normal cells compared to cisplatin. In vitro experiments show 15 and 15-NPs activated multiple pathways, including p38/MAPK- and MDR-related proteins, achieving multi-target synergistic chemosensitization and anti-resistance, further verified by RNA-sequencing analysis. In vivo tests demonstrate that 15 and 15-NPs efficiently inhibit tumor growth and systemic toxicity, especially 15-NPs with optimal tumor-inhibition rate and survival (80% and 100%), superior to cisplatin (40% and 50%), attributing to its extra endocytosis, EPR effect, and precisely tumor-targeted release besides the advantage of a free HO-1i-Pt(IV) prodrug. Additionally, 15 and 15-NPs distinctly regulate T-cell and macrophage functions, thereby exhibiting a chemoimmuno-combined action. This study highlights that multi-functional Platinum(IV) prodrug target-delivered to tumors via carrier-free nanoparticles may represent an effective modality for improving cancer therapy.
Collapse
Affiliation(s)
- Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiao-Ya Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Bo-Wen Ding
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jia-Qian Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yu-Kuan Feng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Ji-Hui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
6
|
Luo J, Bian C, Liu M, Fang Y, Jin L, Yu R, Huang H. Research on gene editing and immunosuppressants in kidney xenotransplantation. Transpl Immunol 2025; 89:102184. [PMID: 39900229 DOI: 10.1016/j.trim.2025.102184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 01/18/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
Gene-edited pig organ transplantation can solve the serious shortage of human donor organs. Currently, xenotransplantation is rapidly developing and has made significant breakthroughs. The use of GTKO (Gal knockout) pigs is an important step forward. The subsequent knockout of three genes combined with the transfer of immune-related genes effectively prolonged the survival time of non-human primate (NHP) transplantation in xenotransplantation. Due to the success of allogeneic kidney transplantation on NHP, this gene editing protocol was recently applied to clinical patients. Two patients underwent allogeneic kidney transplantation and survived for 51 days and 47 days. Exceeding the hyperacute rejection period proves that appropriate gene editing strategies and the combination of immunosuppressive agents contribute to the success of xenotransplantation. To further enhance the feasibility of pig kidney xenograft, this article mainly explores the effects of the NHP xenograft gene editing scheme and immunosuppressants on prolonging transplant survival time.
Collapse
Affiliation(s)
- JiaJiao Luo
- Department of Organ Transplantation Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - CongWen Bian
- Department of Organ Transplantation Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Min Liu
- Department of Organ Transplantation Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuan Fang
- Department of Organ Transplantation Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Jin
- Department of Organ Transplantation Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Rui Yu
- Department of Organ Transplantation Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - HanFei Huang
- Department of Organ Transplantation Department, First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
7
|
Shi Y, Hong R, Fan Z, Huan R, Gao Y, Ma M, Liu T, Pan C. Chronic environmental exposure to polystyrene microplastics increases the risk of nonalcoholic fatty liver disease. Toxicology 2025; 511:154067. [PMID: 39864238 DOI: 10.1016/j.tox.2025.154067] [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: 12/03/2024] [Revised: 01/18/2025] [Accepted: 01/24/2025] [Indexed: 01/28/2025]
Abstract
Microplastics (MPs), as the crucial environmental pollutants, can be easily transported into the human body and accumulate in the liver. However, current studies mainly focus on acute exposure to MPs, investigations on long-term interactions with MPs alone remain limited. Thereby, we examined noxious properties of MPs and selected the most common polystyrene (PS) MPs as the research object, including unmodified PS MPs (PS-MPs) and positive-charged PS MPs (PS-NH2) at 10 mg/L employing oral drinking water methods in mice for six consecutive months in vivo. In vitro, we treated the human hepatocyte cells with MPs at 25 μg/mL to explore involved mechanisms. The results revealed that six-month MPs exposure led to nonalcoholic fatty liver disease (NAFLD) including impaired liver functions, extensive lipid depositions accompanied by abnormal levels of metabolic genes and PS-NH2 MPs exerted a stronger effect than PS-MPs. Concurrently, mice treated with MPs revealed the accumulation of senescent hepatocytes, leading to increased secretions of senescent phenotypes in the liver. We also discovered that MPs initiated the HO-1/Nrf2 axis consequently inducing ferroptosis in vivo and in vitro, as shown by massive iron deposition, extensive lipid peroxidation along with significant protein expressions in ferroptosis-related markers. Additionally, targeting the HO-1/Nrf2 pathway to further alleviate ferroptosis with corresponding inhibitors could efficiently alleviate cell senescence. Therefore, our study reveals new evidence of the relationship between chronic exposure to MPs and NAFLD and furthers the understanding of how plastic pollution affects human health.
Collapse
Affiliation(s)
- Yujie Shi
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Runyang Hong
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Zhencheng Fan
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Ran Huan
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Yajie Gao
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Min Ma
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China; Department of Obstetrics and Gynecology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province 225001, China; Jiangsu Key Laboratory of Non coding RNA Basic and Clinical Transformation, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Tingting Liu
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China
| | - Chun Pan
- Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu Province 225009, China; Jiangsu Key Laboratory of Non coding RNA Basic and Clinical Transformation, Yangzhou University, Yangzhou, Jiangsu Province 225009, China.
| |
Collapse
|
8
|
Shu Y, Wu X, Zhang D, Jiang S, Ma W. Exploring the Mechanisms of Iron Overload-Induced Liver Injury in Rats Based on Transcriptomics and Proteomics. BIOLOGY 2025; 14:81. [PMID: 39857310 PMCID: PMC11761193 DOI: 10.3390/biology14010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Iron is a trace element that is indispensable for the growth and development of animals. Excessive iron supplementation may lead to iron overload and elevated reactive oxygen species (ROS) production in animals, causing cellular damage. Nevertheless, the precise mechanism by which iron overload causes cell injury remains to be fully elucidated. In this study, 16 male SD rats aged 6 to 7 weeks were randomly assigned to either a control group (CON) or an iron overload group (IO). Rats in the iron overload group received 150 mg/kg iron dextran injections every three days for a duration of four weeks. The results indicated that iron treatment with iron dextran significantly increased the scores of steatosis (p < 0.05) and inflammation (p < 0.05) in the NAS score. The integrated transcriptomic and proteomic analysis suggests that HO-1 and Lnc286.2 are potentially significant in iron overload-induced liver injury in rats. In vitro experiments utilizing ferric ammonium citrate (FAC) were conducted to establish an iron overload model in rat liver-derived BRL-3A cells. The result found that FAC treatment can significantly increase the BRL-3A cell's Fe2+ content (p < 0.05), ROS (p < 0.01), lipid ROS (p < 0.01) levels, and the expression of the HO-1 gene and protein (p < 0.01), aligning with proteomic and transcriptomic findings. HO-1 inhibition can significantly decrease BRL-3A cell vitality (p < 0.01) and promote ROS (p < 0.05) and lipid ROS (p < 0.01), thus aggravating FAC-induced BRL-3A cell iron overload damage. Using the agonist of HO-1 agonist cobalt protoporphyrin (CoPP) to induce HO-1 overexpression can significantly alleviate the decrease in FAC-induced BRL-3A cell viability (p < 0.01), ROS (p < 0.01), and lipid ROS (p < 0.01). In addition, siLnc286.2 treatment can increase HO-1 expression, alleviate the decline of FAC-induced BRL-3A cell activity, and increase lipid ROS (p < 0.05) content. In conclusion, the findings of this study suggest that by suppressing the expression of Lnc286.2, we can enhance the expression of HO-1, which in turn alleviates lipid peroxidation in cells and increases their antioxidant capacity, thereby exerting a protective effect against liver cell injury induced by iron overload.
Collapse
Affiliation(s)
- Yujia Shu
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (X.W.); (D.Z.); (S.J.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuanfu Wu
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (X.W.); (D.Z.); (S.J.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Dongxu Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (X.W.); (D.Z.); (S.J.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuxia Jiang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (X.W.); (D.Z.); (S.J.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Wenqiang Ma
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (X.W.); (D.Z.); (S.J.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
9
|
Arosio P, Cairo G, Bou-Abdallah F. A Brief History of Ferritin, an Ancient and Versatile Protein. Int J Mol Sci 2024; 26:206. [PMID: 39796064 PMCID: PMC11719527 DOI: 10.3390/ijms26010206] [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: 11/24/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Ferritin, a highly conserved iron storage protein, is among the earliest proteins that have been purified, named, and characterized due to its unique properties that continue to captivate researchers. Ferritin is composed of 24 subunits that form an almost spherical shell delimiting a cavity where thousands of iron atoms can be stored in a nontoxic ferric form, thereby preventing cytosolic iron from catalyzing oxidative stress. Mitochondrial and extracellular ferritin have also been described and characterized, with the latter being associated with several signaling functions. In addition, serum ferritin serves as a reliable indicator of both iron stores and inflammatory conditions. First identified and purified through crystallization in 1937, ferritin has since drawn significant attention for its critical role in iron metabolism and regulation. Its unique structural features have recently been exploited for many diverse biological and technological applications. To date, more than 40,000 publications have explored this remarkable protein. Here, we present a historical overview, tracing its journey from discovery to current applications and highlighting the evolution of biochemical techniques developed for its structure-function characterization over the past eight decades.
Collapse
Affiliation(s)
- Paolo Arosio
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
| | - Fadi Bou-Abdallah
- Department of Chemistry, State University of New York at Potsdam, Potsdam, NY 13676, USA;
| |
Collapse
|
10
|
Zhang M, Chen X, Zhang Y. Mechanisms of Vitamins Inhibiting Ferroptosis. Antioxidants (Basel) 2024; 13:1571. [PMID: 39765898 PMCID: PMC11673384 DOI: 10.3390/antiox13121571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis is an iron-dependent form of cell death, which is characterized by the uncontrolled and overwhelming peroxidation of cell membrane lipids. Ferroptosis has been implicated in the progression of various pathologies, including steatotic liver, heart failure, neurodegenerative diseases, and diabetes. Targeted inhibition of ferroptosis provides a promising strategy to treat ferroptosis-related diseases. Multivitamins, including vitamins A, B, C, D, E, and K, have shown a good ability to inhibit ferroptosis. For example, vitamin A significantly upregulated the expression of several key ferroptotic gatekeepers genes through nuclear retinoic acid receptors and retinoic X receptors (RAR/RXR). Vitamin B6 could compensate for the impaired glutathione (GSH) levels and restore Glutathione peroxidase 4 (GPX4) expression in cells, ultimately inhibiting ferroptosis. Vitamin D could up-regulate the expression of several anti-ferroptosis proteins by activating vitamin D receptors. Vitamin E and hydroquinone vitamin K (VKH2) can directly inhibit the propagation of lipid peroxidation, thereby inhibiting ferroptosis. In this review, we summarize the currently understood mechanisms by which vitamins inhibit ferroptosis to provide reference information for future research on the development of ferroptosis inhibitors.
Collapse
Affiliation(s)
- Meng Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (M.Z.); (X.C.)
| | - Xin Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (M.Z.); (X.C.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yumei Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (M.Z.); (X.C.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
11
|
Rybnikářová A, Buchal R, Pláteník J. Ferritin with methylglyoxal produces reactive oxygen species but remains functional. Free Radic Res 2024; 58:702-722. [PMID: 39508662 DOI: 10.1080/10715762.2024.2417281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Iron is necessary for life, but the simultaneous iron-catalyzed formation of reactive oxygen species (ROS) is involved in pathogenesis of many diseases. One of them is diabetes mellitus, a widespread disease with severe long-term complications, including neuropathy, retinopathy, and nephropathy. Much evidence points to methylglyoxal, a potent glycating agent, as the key mediator of diabetic complications. In diabetes, there is also a peculiar dysregulation of iron homeostasis, leading to an expansion of redox-active iron. This in vitro study focuses on the interaction of methylglyoxal with ferritin, which is the main cellular protein for iron storage. Methylglyoxal effectively liberates iron from horse spleen ferritin, as well as synthetic iron cores; in both cases, it is partially mediated by superoxide. The interaction of methylglyoxal with ferritin increases the production of hydrogen peroxide, much above the generation of peroxide by methylglyoxal alone, in an iron-dependent manner. Glycation with methylglyoxal results in structural changes in ferritin. All of these findings can be demonstrated with pathophysiologically relevant (submillimolar) methylglyoxal concentrations. However, the rate of iron release by ascorbate, the ferroxidase activity, or the diameter of gated pores even in intensely glycated ferritin is not altered. In conclusion, although the functional features of ferritin resist alterations due to glycation, the interaction of methylglyoxal with ferritin liberates iron and markedly increases ROS production, both of which could enhance oxidative stress in vivo. Our findings may have implications for the pathogenesis of long-term diabetic complications, as well as for the use of ferritin as a nanocarrier in chemotherapy.
Collapse
Affiliation(s)
- Adriana Rybnikářová
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Praha 2, Czech Republic
| | - Richard Buchal
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Praha 2, Czech Republic
| | - Jan Pláteník
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, Praha 2, Czech Republic
| |
Collapse
|
12
|
Zhu Z, Wu J, Wen Y, Wu X, Bao H, Wang M, Kang K. Advances in the Effects of Heat Stress on Ovarian Granulosa Cells: Unveiling Novel Ferroptosis Pathways. Vet Sci 2024; 11:464. [PMID: 39453056 PMCID: PMC11511475 DOI: 10.3390/vetsci11100464] [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: 08/12/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Heat stress has been one of the key research areas for researchers due to the wide-ranging effects and complex mechanisms of action of its stress product reactive oxygen species (ROS). The aim of this paper is to comprehensively review and summarize the effects of heat stress on ovarian granulosa cells and their mechanism of action. We systematically reviewed the effects of heat stress on ovarian granulosa cells, including intracellular steroid hormone changes, oxidative stress, apoptosis, and mitochondrial function. Meanwhile, this paper discusses in detail several major mechanisms by which heat stress induces apoptosis in ovarian granulosa cells, such as through the activation of apoptosis-related genes, induction of endoplasmic reticulum stress, and the mitochondrial pathway. In addition, we analyzed the mechanism of ferroptosis in ovarian granulosa cells under heat stress conditions, summarized the potential association between heat stress and ferroptosis in light of the existing literature, and explored the key factors in the mechanism of action of heat stress, such as the signaling pathways of Nrf2/Keap1, HSPs, and JNK, and analyzed their possible roles in the process of ferroptosis. Finally, this paper provides an outlook on the future research direction, describing the possible interaction between heat stress and ferroptosis, with a view to providing a theoretical basis for further understanding and revealing the complex mechanism of ferroptosis occurrence in ovarian granulosa cells under heat stress.
Collapse
Affiliation(s)
- Zhen Zhu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Z.Z.); (J.W.)
| | - Jiang Wu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Z.Z.); (J.W.)
| | - Yuguo Wen
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Z.Z.); (J.W.)
| | - Xiaocheng Wu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Z.Z.); (J.W.)
| | - Huimingda Bao
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Z.Z.); (J.W.)
| | - Min Wang
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Z.Z.); (J.W.)
| | - Kai Kang
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
13
|
Gáll T, Pethő D, Erdélyi K, Egri V, Balla JG, Nagy A, Nagy A, Póliska S, Gram M, Gábriel R, Nagy P, Balla J, Balla G. Heme: A link between hemorrhage and retinopathy of prematurity progression. Redox Biol 2024; 76:103316. [PMID: 39260060 PMCID: PMC11415884 DOI: 10.1016/j.redox.2024.103316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
Neovascularization is implicated in the pathology of retinopathy of prematurity (ROP), diabetic retinopathy (DR), and age-related macular degeneration (AMD), which are the leading causes of blindness worldwide. In our work, we analyzed how heme released during hemorrhage affects hypoxic response and neovascularization. Our retrospective clinical analysis demonstrated, that hemorrhage was associated with more severe retinal neovascularization in ROP patients. Our heme-stimulated human retinal pigment epithelial (ARPE-19) cell studies demonstrated increased expression of positive regulators of angiogenesis, including vascular endothelial growth factor-A (VEGFA), a key player of ROP, DR and AMD, and highlighted the activation of the PI3K/AKT/mTOR/VEGFA pathway involved in angiogenesis in response to heme. Furthermore, heme decreased oxidative phosphorylation in the mitochondria, augmented glycolysis, facilitated HIF-1α nuclear translocation, and increased VEGFA/GLUT1/PDK1 expression suggesting HIF-1α-driven hypoxic response in ARPE-19 cells without effecting the metabolism of reactive oxygen species. Inhibitors of HIF-1α, PI3K and suppression of mTOR pathway by clinically promising drug, rapamycin, mitigated heme-provoked cellular response. Our data proved that oxidatively modified forms of hemoglobin can be sources of heme to induce VEGFA during retinal hemorrhage. We propose that hemorrhage is involved in the pathology of ROP, DR, and AMD.
Collapse
Affiliation(s)
- Tamás Gáll
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Dávid Pethő
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary; HUN-REN-UD Vascular Biology and Myocardium Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, Debrecen, H-4032, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Katalin Erdélyi
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest H-1122, Hungary
| | - Virág Egri
- Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Jázon György Balla
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Annamária Nagy
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary; HUN-REN-UD Vascular Biology and Myocardium Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, Debrecen, H-4032, Hungary
| | - Annamária Nagy
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Magnus Gram
- Pediatrics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Department of Neonatology, Skåne University Hospital, Lund, Sweden; Biofilms - Research Center for Biointerfaces, Department of Biomedical Science, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Róbert Gábriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, H-7624, Hungary; János Szentágothai Research Centre, University of Pécs, Pécs, H-7624, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest H-1122, Hungary; Chemistry Institute, University of Debrecen, Debrecen, H-4032, Hungary; Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology, University of Veterinary Medicine; Budapest, Hungary
| | - József Balla
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary; HUN-REN-UD Vascular Biology and Myocardium Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, Debrecen, H-4032, Hungary
| | - György Balla
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary; Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary.
| |
Collapse
|
14
|
Alsharabasy AM, Lagarias PI, Papavasileiou KD, Afantitis A, Farràs P, Glynn S, Pandit A. Examining Hemin and its Derivatives: Induction of Heme-Oxygenase-1 Activity and Oxidative Stress in Breast Cancer Cells through Collaborative Experimental Analysis and Molecular Dynamics Simulations. J Med Chem 2024; 67:15411-15427. [PMID: 39159487 PMCID: PMC11403666 DOI: 10.1021/acs.jmedchem.4c00989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Hemin triggers intracellular reactive oxygen species (ROS) accumulation and enhances heme oxygenase-1 (HOX-1) activity, indicating its potential as an anticancer agent, though precise control of its intracellular levels is crucial. The study explores the impact of hemin and its derivatives, hemin-tyrosine, and hemin-styrene (H-Styr) conjugates on migration, HOX-1 expression, specific apoptosis markers, mitochondrial functions, and ROS generation in breast cancer cells. Molecular docking and dynamics simulations were used to understand the interactions among HOX-1, heme, and the compounds. Hemin outperforms its derivatives in inducing HOX-1 expression, exhibiting pro-oxidative effects and reducing cell migration. Molecular simulations show that heme binds favorably to HOX-1, followed by the other compounds, primarily through van der Waals and electrostatic forces. However, only van der Waals forces determine the H-Styr complexation. These interactions, influenced by metalloporphyrin characteristics, provide insights into HOX-1 regulation and ROS generation, potentially guiding the development of breast cancer therapies targeting oxidative stress.
Collapse
Affiliation(s)
- Amir M Alsharabasy
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | | | - Konstantinos D Papavasileiou
- Department of ChemoInformatics, Novamechanics Ltd., Nicosia 1070, Cyprus
- Department of Chemoinformatics, Novamechanics MIKE, Piraeus 18545, Greece
- Division of Data Driven Innovation, Entelos Institute, Larnaca 6059, Cyprus
| | - Antreas Afantitis
- Department of ChemoInformatics, Novamechanics Ltd., Nicosia 1070, Cyprus
- Department of Chemoinformatics, Novamechanics MIKE, Piraeus 18545, Greece
- Division of Data Driven Innovation, Entelos Institute, Larnaca 6059, Cyprus
| | - Pau Farràs
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- School of Biological and Chemical Sciences, Ryan Institute, University of Galway, Galway H91 TK33, Ireland
| | - Sharon Glynn
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway H91 YR71, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
15
|
Cui Y, Liao Y, Chen Y, Zhao X, Zhang Y, Wang H, Li L, Zhang X, Chen K, Jia M, Tian J, Ruan X, Shi Y, Yang P, Chen J. Low expression of selenoprotein S induces oxidative damage in cartilages. J Trace Elem Med Biol 2024; 85:127492. [PMID: 38964025 DOI: 10.1016/j.jtemb.2024.127492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
Low levels of the indispensable trace element selenium (Se) can cause oxidative stress and disrupt environmental homeostasis in humans and animals. Selenoprotein S (Selenos), of which Se is a key component, is a member of the selenoprotein family involved in various biological processes. This study aimed to investigate whether low-level SELENOS gene expression can induce oxidative stress and decrease the antioxidative capacity of chondrocytes. Compared with control cells, SELENOS-knockdown ATDC5 cells showed substantially higher dihydroethidium, reactive oxygen species and malondialdehyde levels, and lower superoxide dismutase (SOD) expression. Knockout of the gene in C57BL/6 mice increased the 8-hydroxy-2-deoxyguanosine level considerably and decreased SOD expression in cartilages relative to the levels in wild-type mice. The results showed that the increased nuclear factor erythroid 2-related factor 2/heme oxygenase-1 signaling mediated by low-level SELENOS expression was involved in oxidative damage. The proliferative zone of the cartilage growth plate of SELENOS-knockout mice was shortened, suggesting cartilage differentiation dysfunction. In conclusion, this study confirmed that low-level Selenos expression plays a role in oxidative stress in cartilages.
Collapse
Affiliation(s)
- Yixin Cui
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yucheng Liao
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yonghui Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Xu Zhao
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yi Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Hui Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Lian Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Xinhe Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Kunpan Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Mingzhao Jia
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Jing Tian
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Xingran Ruan
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yawen Shi
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Pinglin Yang
- Department of Orthopaedic Surgery, Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, People's Republic of China.
| | - Jinghong Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| |
Collapse
|
16
|
Tan AF, Sakam SSB, Piera K, Rajahram GS, William T, Barber BE, Anstey NM, Grigg MJ, Kho S. Neutrophil activation, acute lung injury and disease severity in Plasmodium knowlesi malaria. PLoS Negl Trop Dis 2024; 18:e0012424. [PMID: 39150978 DOI: 10.1371/journal.pntd.0012424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/28/2024] [Accepted: 08/02/2024] [Indexed: 08/18/2024] Open
Abstract
The risk of severe malaria from the zoonotic parasite Plasmodium knowlesi approximates that from P. falciparum. In severe falciparum malaria, neutrophil activation contributes to inflammatory pathogenesis, including acute lung injury (ALI). The role of neutrophil activation in the pathogenesis of severe knowlesi malaria has not been examined. We evaluated 213 patients with P. knowlesi mono-infection (138 non-severe, 75 severe) and 49 Plasmodium-negative controls from Malaysia. Markers of neutrophil activation (soluble neutrophil elastase [NE], citrullinated histone [CitH3] and circulating neutrophil extracellular traps [NETs]) were quantified in peripheral blood by microscopy and immunoassays. Findings were correlated with malaria severity, ALI clinical criteria, biomarkers of parasite biomass, haemolysis, and endothelial activation. Neutrophil activation increased with disease severity, with median levels higher in severe than non-severe malaria and controls for NE (380[IQR:210-930]ng/mL, 236[139-448]ng/mL, 218[134-307]ng/mL, respectively) and CitH3 (8.72[IQR:3.0-23.1]ng/mL, 4.29[1.46-9.49]ng/mL, 1.53[0.6-2.59]ng/mL, respectively)[all p<0.01]. NETs were higher in severe malaria compared to controls (126/μL[IQR:49-323] vs 51[20-75]/μL, p<0.001). In non-severe malaria, neutrophil activation fell significantly upon discharge from hospital (p<0.03). In severe disease, NETs, NE, and CitH3 were correlated with parasitaemia, cell-free haemoglobin and angiopoietin-2 (all Pearson's r>0.24, p<0.05). Plasma NE and angiopoietin-2 were higher in knowlesi patients with ALI than those without (p<0.008); neutrophilia was associated with an increased risk of ALI (aOR 3.27, p<0.01). In conclusion, neutrophil activation is increased in ALI and in proportion to disease severity in knowlesi malaria, is associated with endothelial activation, and may contribute to disease pathogenesis. Trials of adjunctive therapies to regulate neutrophil activation are warranted in severe knowlesi malaria.
Collapse
Affiliation(s)
- Angelica F Tan
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Infectious Diseases Society Kota Kinabalu Sabah - Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sitti Saimah Binti Sakam
- Infectious Diseases Society Kota Kinabalu Sabah - Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
| | - Kim Piera
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
| | - Giri S Rajahram
- Infectious Diseases Society Kota Kinabalu Sabah - Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
- Clinical Research Centre, Queen Elizabeth Hospital, Kota Kinabalu, Malaysia
- Queen Elizabeth Hospital II, Ministry of Health Malaysia, Kota Kinabalu, Malaysia
- School of Medicine and Health Sciences, Monash University Malaysia, Kuala Lumpur, Malaysia
| | - Timothy William
- Infectious Diseases Society Kota Kinabalu Sabah - Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
- Clinical Research Centre, Queen Elizabeth Hospital, Kota Kinabalu, Malaysia
| | - Bridget E Barber
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Infectious Diseases Society Kota Kinabalu Sabah - Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
| | - Matthew J Grigg
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Infectious Diseases Society Kota Kinabalu Sabah - Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
| | - Steven Kho
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
| |
Collapse
|
17
|
Zhang J, Fan M, Tang J, Lin X, Liu G, Wen C, Xu X. Possibility and challenge of plant-derived ferritin cages encapsulated polyphenols in the precise nutrition field. Int J Biol Macromol 2024; 275:133579. [PMID: 38964678 DOI: 10.1016/j.ijbiomac.2024.133579] [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: 05/10/2024] [Revised: 06/24/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Polyphenols have attracted extensive attention due to their rich functional activities, such as antioxidant, anti-inflammatory and anti-tumor. However, the low solubility and poor stability limit their bioavailability and functional activities. Plant-derived ferritin cages have a unique hollow cage structure that can embed polyphenols to improve their unfavorable properties. Therefore, it is essential to adequately elaborate and summarize plant-derived ferritin cages to maximize their potential benefits in nutritional interventions. This review focuses on the fundamental properties of plant-derived ferritin cages, including the preparation process, purification technology, identification methods, and structural and functional properties. The relevant research on ferritin cages in polyphenol delivery has been summarized, including the delivery of water/lipid soluble polyphenols, modification of ferritin cages, and the interaction between polyphenols and ferritin cages. The research progress, shortcomings and prospects of plant-derived ferritin cages in precise nutrition are introduced. In addition, the relevant research on ferritin in immune response and protein engineering is also discussed to provide the theoretical basis for applying plant-derived ferritin cages in many frontier fields.
Collapse
Affiliation(s)
- Jixian Zhang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China; Guangling College, Yangzhou University, Yangzhou 225000, China
| | - Meidi Fan
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jialuo Tang
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Xinying Lin
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Guoyan Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Chaoting Wen
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China.
| | - Xin Xu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China.
| |
Collapse
|
18
|
Bauer M, Ermolaeva M, Singer M, Wetzker R, Soares MP. Hormesis as an adaptive response to infection. Trends Mol Med 2024; 30:633-641. [PMID: 38744580 DOI: 10.1016/j.molmed.2024.04.012] [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: 02/08/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
Hormesis is a phenomenon whereby low-level stress can improve cellular, organ, or organismal fitness in response to a subsequent similar or other stress insult. Whereas hormesis is thought to contribute to the fitness benefits arising from symbiotic host-microbe interactions, the putative benefits of hormesis in host-pathogen interactions have yet to be explored. Hormetic responses have nonetheless been reported in experimental models of infection, a common feature of which is regulation of host mitochondrial function. We propose that these mitohormetic responses could be harnessed therapeutically to limit the severity of infectious diseases.
Collapse
Affiliation(s)
- Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| | - Maria Ermolaeva
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, UK
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Miguel P Soares
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany; Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
19
|
Sadeghi M, Moslehi A, Kheiry H, Kiani FK, Zarei A, Khodakarami A, Karpisheh V, Masjedi A, Rahnama B, Hojjat-Farsangi M, Raeisi M, Yousefi M, Movasaghpour Akbari AA, Jadidi-Niaragh F. The sensitivity of acute myeloid leukemia cells to cytarabine is increased by suppressing the expression of Heme oxygenase-1 and hypoxia-inducible factor 1-alpha. Cancer Cell Int 2024; 24:217. [PMID: 38918761 PMCID: PMC11197338 DOI: 10.1186/s12935-024-03393-3] [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: 01/27/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML), a malignancy Often resistant to common chemotherapy regimens (Cytarabine (Ara-c) + Daunorubicin (DNR)), is accompanied by frequent relapses. Many factors are involved in causing chemoresistance. Heme Oxygenase-1 (HO-1) and Hypoxia-Inducible Factor 1-alpha (HIF-1α) are two of the most well-known genes, reported to be overexpressed in AML and promote resistance against chemotherapy according to several studies. The main chemotherapy agent used for AML treatment is Ara-c. We hypothesized that simultaneous targeting of HO-1 and HIF-1α could sensitize AML cells to Ara-c. METHOD In this study, we used our recently developed, Trans-Activator of Transcription (TAT) - Chitosan-Carboxymethyl Dextran (CCMD) - Poly Ethylene Glycol (PEG) - Nanoparticles (NPs), to deliver Ara-c along with siRNA molecules against the HO-1 and HIF-1α genes to AML primary cells (ex vivo) and cell lines including THP-1, KG-1, and HL-60 (in vitro). Subsequently, the effect of the single or combinational treatment on the growth, proliferation, apoptosis, and Reactive Oxygen Species (ROS) formation was evaluated. RESULTS The designed NPs had a high potential in transfecting cells with siRNAs and drug. The results demonstrated that treatment of cells with Ara-c elevated the generation of ROS in the cells while decreasing the proliferation potential. Following the silencing of HO-1, the rate of apoptosis and ROS generation in response to Ara-c increased significantly. While proliferation and growth inhibition were considerably evident in HIF-1α-siRNA-transfected-AML cells compared to cells treated with free Ara-c. We found that the co-inhibition of genes could further sensitize AML cells to Ara-c treatment. CONCLUSIONS As far as we are aware, this study is the first to simultaneously inhibit the HO-1 and HIF-1α genes in AML using NPs. It can be concluded that HO-1 causes chemoresistance by protecting cells from ROS damage. Whereas, HIF-1α mostly exerts prolific and direct anti-apoptotic effects. These findings imply that simultaneous inhibition of HO-1 and HIF-1α can overcome Ara-c resistance and help improve the prognosis of AML patients.
Collapse
Affiliation(s)
- Mohammad Sadeghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asma Moslehi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadiseh Kheiry
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asieh Zarei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Khodakarami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Masjedi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Badrossadat Rahnama
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mortaza Raeisi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Deng B, He X, Wang Z, Kang J, Zhang G, Li L, Kang X. HSP70 protects PC12 cells against TBHP-induced apoptosis and oxidative stress by activating the Nrf2/HO-1 signaling pathway. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00924-0. [PMID: 38807023 DOI: 10.1007/s11626-024-00924-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
HSP70 exhibits neuroprotective, antioxidant, and anti-apoptotic properties, which are crucial in preventing spinal cord injury (SCI) induced by oxidative stress and apoptosis. In this study, we assessed the potential protective effects and underlying mechanisms of HSP70 on tert-butyl hydroperoxide (TBHP)-damaged PC12 cells in an in vitro model of SCI. To establish the model, PC12 cells were subjected to oxidative damage induced by TBHP, followed by overexpression of HSP70. Cell viability was assessed using the CCK8 kit, intracellular reactive oxygen species level was evaluated using a commercial kit, cell apoptosis was detected using the Annexin V-APC/7-ADD Apoptosis Detection Kit, and the oxidative stress level was determined using SOD and MDA assay kits. Western blot analysis was used to detect the expression levels of Bax, cleaved caspase-3, and Bcl-2 proteins. Furthermore, immunofluorescence staining and Western bolt were used to detect the expression levels of proteins associated with the Nrf2/HO-1 signaling pathway. We found that HSP70 overexpression reduced apoptosis and oxidative stress in TBHP-induced PC12 cells. Furthermore, it activated the Nrf2/HO-1 signaling pathway. In addition, the Nrf2 inhibitor ML385 attenuated the protective effects of HSP70 on TBHP-induced PC12 cells. In conclusion, HSP70 can partially alleviate TBHP-induced apoptosis and oxidative stress in PC12 cells by promoting the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Bo Deng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xuegang He
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zhaoheng Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Jihe Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
21
|
Meza-Buendia AK, Aparicio-Trejo OE, Díaz F, Pedraza-Chaverri J, Álvarez-Delgado C, Rosas C. Climate change consequences on the systemic heart of female Octopus maya: oxidative phosphorylation assessment and the antioxidant system. Biol Open 2024; 13:bio060103. [PMID: 38752595 PMCID: PMC11155352 DOI: 10.1242/bio.060103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 04/05/2024] [Indexed: 06/09/2024] Open
Abstract
There is evidence that indicates that temperature modulates the reproduction of the tropical species Octopus maya, through the over- or under-expression of many genes in the brain. If the oxygen supply to the brain depends on the circulatory system, how temperature affects different tissues will begin in the heart, responsible for pumping the oxygen to tissues. The present study examines the impact of heat stress on the mitochondrial function of the systemic heart of adult O. maya. The mitochondrial metabolism and antioxidant defense system were measured in the systemic heart tissue of female organisms acclimated to different temperatures (24, 26, and 30°C). The results show that acclimation temperature affects respiratory State 3 and State 4o (oligomycin-induced) with higher values observed in females acclimated at 26°C. The antioxidant defense system is also affected by acclimation temperature with significant differences observed in superoxide dismutase, glutathione S-transferase activities, and glutathione levels. The results suggest that high temperatures (30°C) could exert physical limitations on the circulatory system through the heart pumping, affecting nutrient and oxygen transport to other tissues, including the brain, which exerts control over the reproductive system. The role of the cardiovascular system in supporting aerobic metabolism in octopus females is discussed.
Collapse
Affiliation(s)
- Ana Karen Meza-Buendia
- Departamento de Biotecnología Marina, Centro de Investigación Científica y de Educación Superior de Ensenada, 22860Ensenada, Baja California, México
| | - Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología “Ignacio Chávez”, 14080 Ciudad de México, México
| | - Fernando Díaz
- Departamento de Biotecnología Marina, Centro de Investigación Científica y de Educación Superior de Ensenada, 22860Ensenada, Baja California, México
| | - José Pedraza-Chaverri
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Carolina Álvarez-Delgado
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, 22860 Ensenada, Baja California, México
| | - Carlos Rosas
- Laboratorio de Ecofisiología Aplicada, Unidad Multidisciplinaria de Docencia e Investigación, de Sisal, Facultad de Ciencias, Universidad Nacional Autónoma de México, 97356 Puerto de Abrigo, Sisal, Yucatán, México
| |
Collapse
|
22
|
Hou DY, Lu JJ, Zhang X, Abudukeyoumu A, Li MQ, Zhu XY, Xie F. Heme metabolism and HO-1 in the pathogenesis and potential intervention of endometriosis. Am J Reprod Immunol 2024; 91:e13855. [PMID: 38745499 DOI: 10.1111/aji.13855] [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: 08/13/2023] [Revised: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Endometriosis (EM) is one of the diseases related to retrograded menstruation and hemoglobin. Heme, released from hemoglobin, is degraded by heme oxygenase-1 (HO-1). In EM lesions, heme metabolites regulate processes such as inflammation, redox balance, autophagy, dysmenorrhea, malignancy, and invasion, where macrophages (Mø) play a fundamental role in their interactions. Regulation occurs at molecular, cellular, and pathological levels. Numerous studies suggest that heme is an indispensable component in EM and may contribute to its pathogenesis. The regulatory role of heme in EM encompasses cytokines, signaling pathways, and kinases that mediate cellular responses to external stimuli. HO-1, a catalytic enzyme in the catabolic phase of heme, mitigates heme's cytotoxicity in EM due to its antioxidant, anti-inflammatory, and anti-proliferative properties. Certain compounds may intervene in EM by targeting heme metabolism, guiding the development of appropriate treatments for all stages of endometriosis.
Collapse
Affiliation(s)
- Ding-Yu Hou
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Jia-Jing Lu
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Xing Zhang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Ayitila Abudukeyoumu
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Jiading District, Shanghai, People's Republic of China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Xiao-Yong Zhu
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
| | - Feng Xie
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, People's Republic of China
- Medical Center of Diagnosis and Treatment for Cervical and Intrauterine Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
23
|
Ghoneum M, Badr El-Din NK, Alaa El-Dein M. Anti-radiation effect of MRN-100: a hydro-ferrate fluid, in vivo. JOURNAL OF RADIATION RESEARCH 2024; 65:145-158. [PMID: 38247158 PMCID: PMC10959437 DOI: 10.1093/jrr/rrad095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/01/2023] [Indexed: 01/23/2024]
Abstract
Ionizing radiation (IR) severely harms many organs, especially the hematopoietic tissue, mandating the development of protective nutraceuticals. MRN-100, a hydro-ferrate fluid, has been shown to protect γ-radiated fish against hematopoietic tissue damage and lethality. The current study aimed to examine MRN-100's protective effect against irradiated mice and explore the mechanisms underlying its effect. Mice received a single acute, sub-lethal, 5 Gy, whole body dose of X-ray IR. MRN-100 treatment was administered daily for 2-weeks pre-irradiation until 1-week post-irradiation. Spleen and blood were analysed for oxidative stress, hematological, histological and biochemical parameters. Radiation exposure markedly decreased complete blood count (CBC) parameters including hemoglobin, hematocrit, red blood cells, platelets, white blood cells and lymphocytes, and significantly increased neutrophils. In contrast, MRN-100 supplementation to irradiated mice ameliorated all CBC parameters and protected against DNA damage in both splenic cells and serum. It also had an antioxidant effect, increasing the levels of glutathione, superoxide dismutase, catalase and total antioxidant capacity, which were otherwise decreased by irradiation. MRN-100 intake reduced the oxidative stress biomarker levels of nitric oxide, protein carbonyl, malondialdehyde, reactive oxygen species and 8-hydroxydeoxyguanosine, a marker specific to DNA damage. Furthermore, MRN-100 enhanced serum iron and reversed the radiation-induced elevations of liver enzymes. Finally, MRN-100 protected splenic tissue from irradiation as observed by histology. We conclude that MRN-100 consumption may protect against oxidative stress generated by radiation exposure, suggesting that it may be employed as an adjuvant treatment to prevent radiation's severe damage to important organs.
Collapse
Affiliation(s)
- Mamdooh Ghoneum
- Department of Surgery, Charles Drew University of Medicine and Science, 1621 East 120th Street, Los Angeles, California 90059, USA
- Department of Surgery, University of California Los Angeles, 405 Hilgard Ave, Los Angeles, CA 90095, USA
| | - Nariman K Badr El-Din
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Mai Alaa El-Dein
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
24
|
Wang F, Deng G, Liang N, Hu P, Liu K, Liu T, Li Y, Yuan M, Liu L, Xie J, Qiao L, Liu F, Zhang J. Serum ferritin level is an effective prognostic factor for lung cancer immunotherapy. Cancer Biol Ther 2023; 24:2285367. [PMID: 38031846 PMCID: PMC10783829 DOI: 10.1080/15384047.2023.2285367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023] Open
Abstract
Immunotherapy of lung cancer has achieved promising clinical results. However, it is urgent to develop predictive biomarkers for effective immunotherapy. While ferroptosis plays a critical role in immunotherapy efficacy, ferritin is an important regulatory factor. We, therefore, hypothesize that basal serum ferritin levels before immunotherapy and their corresponding changes during immunotherapy can be useful predictors of immunotherapy response in patients with lung cancer. We measured serum ferritin levels in 107 patients with lung cancer before and during immune checkpoint blockade treatments and studied the correlation between ferritin levels, response rate, and survival. Moreover, the correlation between basal ferritin and PD-L1 expression, tumor stages and pathological types was also analyzed. Patients with lower basal serum ferritin levels before immunotherapy had longer progression-free survival (PFS) (median 7 vs 4 months, P = .023) and higher disease control rate (DCR) (X2 = 4.837, P = .028), those with downregulated serum ferritin levels during immunotherapy correlated with longer PFS (median 9.5 vs 4 months, P < .001) and higher DCR (X2 = 6.475, P = .011). However, the "integrated factor", which was calculated as the combination of lower basal serum ferritin levels before immunotherapy and downregulated serum ferritin levels during immunotherapy, correlated with prolonged PFS (P < .001). Multivariate analyses revealed that the basal serum ferritin levels before immunotherapy and the corresponding changes during immunotherapy were both strong independent prognostic factors (hazard ratio (HR) = 1.60, P = .041; HR = 2.65, P = .001). These findings suggest that serum ferritin levels can be used as a prognostic biomarker for lung cancer in predicting immunotherapy efficacy.
Collapse
Affiliation(s)
- Fei Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Oncology, Zaozhuang Shizhong District People’s Hospital, Zaozhuang, China
| | - Guodong Deng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Ning Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Pingping Hu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Kuo Liu
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Tong Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Yang Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Meng Yuan
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Weifang Medical University, Jinan, China
| | - Li Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Jian Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Lili Qiao
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Fengjun Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiandong Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
25
|
Xiao Q, Sun CC, Tang CF. Heme oxygenase-1: A potential therapeutic target for improving skeletal muscle atrophy. Exp Gerontol 2023; 184:112335. [PMID: 37984695 DOI: 10.1016/j.exger.2023.112335] [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: 10/07/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Skeletal muscle atrophy is a common muscle disease that is directly caused by an imbalance in protein synthesis and degradation. At the histological level, it is mainly characterized by a reduction in muscle mass and fiber cross-sectional area (CSA). Patients with skeletal muscle atrophy present with reduced motor ability, easy fatigue, and poor life quality. Heme oxygenase-1 (HO-1) is an inducible enzyme that catalyzes the degradation of heme and has attracted much attention for its anti-oxidation effects. In addition, there is growing evidence that HO-1 plays an important role in anti-inflammatory, anti-apoptosis, pro-angiogenesis, and maintaining skeletal muscle homeostasis, making it a potential therapeutic target for improving skeletal muscle atrophy. Here, we review the pathogenesis of skeletal muscle atrophy, the biology of HO-1 and its regulation, and the biological function of HO-1 in skeletal muscle homeostasis, with a specific focus on the role of HO-1 in skeletal muscle atrophy, aiming to observe the therapeutic potential of HO-1 for skeletal muscle atrophy.
Collapse
Affiliation(s)
- Qin Xiao
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan 410012, China; School of Physical Education, Hunan First Normal University, Changsha, Hunan 410205, China
| | - Chen-Chen Sun
- School of Physical Education, Hunan First Normal University, Changsha, Hunan 410205, China.
| | - Chang-Fa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan 410012, China.
| |
Collapse
|
26
|
Fang G, Li X, Yang F, Huang T, Qiu C, Peng K, Yang Y, Lan C. Galangin attenuates doxorubicin-induced cardiotoxicity via activating nuclear factor erythroid 2-related factor 2/heme oxygenase 1 signaling pathway to suppress oxidative stress and inflammation. Phytother Res 2023; 37:5854-5870. [PMID: 37655750 DOI: 10.1002/ptr.7991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023]
Abstract
Doxorubicin (DOX) has aroused contradiction between its potent anti-tumor capacity and severe cardiotoxicity. Galangin (Gal) possesses antioxidant, anti-inflammatory, and antiapoptotic activities. We aimed to explore the role and underlying mechanisms of Gal on DOX-induced cardiotoxicity. Mice were intraperitoneally injected with DOX (3 mg/kg, every 2 days for 2 weeks) to generate cardiotoxicity model and Gal (15 mg/kg, 2 weeks) was co-administered via gavage daily. Nuclear factor erythroid 2-related factor 2 (Nrf2) specific inhibitor, ML385, was employed to explore the underlying mechanisms. Compared to DOX-insulted mice, Gal effectively improved cardiac dysfunction and ameliorated myocardial damage. DOX-induced increase of reactive oxygen species, malondialdehyde, and NADPH oxidase activity and downregulation of superoxide dismutase (SOD) activity were blunted by Gal. Gal also markedly blocked increase of IL-1β, IL-6, and TNF-α in DOX-insulted heart. Mechanistically, Gal reversed DOX-induced downregulation of Nrf2, HO-1, and promoted nuclear translocation of Nrf2. ML385 markedly blunted the cardioprotective effects of Gal, as well as inhibitive effects on oxidative stress and inflammation. Gal ameliorates DOX-induced cardiotoxicity by suppressing oxidative stress and inflammation via activating Nrf2/HO-1 signaling pathway. Gal may serve as a promising cardioprotective agent for DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Guangyao Fang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Xiuchuan Li
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| | - Fengyuan Yang
- Department of Nephrology, General Hospital of Western Theater Command, Chengdu, China
| | - Ting Huang
- Department of Medical Oncology, People's Hospital of Luotian County, Huanggang, China
| | - Chenming Qiu
- Department of Burn and Plastic Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Ke Peng
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| | - Yongjian Yang
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Cong Lan
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
27
|
Perera DN, Palliyaguruge CL, Eapasinghe DD, Liyanage DM, Seneviratne RACH, Demini SMD, Jayasinghe JASM, Faizan M, Rajagopalan U, Galhena BP, Hays H, Senathilake K, Tennekoon KH, Samarakoon SR. Factors affecting iron absorption and the role of fortification in enhancing iron levels. NUTR BULL 2023; 48:442-457. [PMID: 37965925 DOI: 10.1111/nbu.12643] [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: 05/17/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 11/16/2023]
Abstract
Iron is an important micronutrient required for a number of biological processes including oxygen transport, cellular respiration, the synthesis of nucleic acids and the activity of key enzymes. The World Health Organization has recognised iron deficiency as the most common nutritional deficiency globally and as a major determinant of anaemia. Iron deficiency anaemia affects 40% of all children between the ages of 6 and 59 months, 37% of mothers who are pregnant and 30% of women between the ages of 15 and 49 years worldwide. Dietary iron exists in two main forms known as haem iron and non-haem iron. Haem iron is obtained from animal sources such as meat and shows higher bioavailability than non-haem iron, which can be obtained from both plant and animal sources. Different components in food can enhance or inhibit iron absorption from the diet. Components such as meat proteins and organic acids increase iron absorption, while phytate, calcium and polyphenols reduce iron absorption. Iron levels in the body are tightly regulated since both iron overload and iron deficiency can exert harmful effects on human health. Iron is stored mainly as haemoglobin and as iron bound to proteins such as ferritin and hemosiderin. Iron deficiency affects individuals at increased risk due to factors such as age, pregnancy, menstruation and various diseases. Different solutions for iron deficiency are applied at individual and community levels. Iron supplements and intravenous iron can be used to treat individuals with iron deficiency, while various types of iron-fortified foods and biofortified crops can be employed for larger communities. Foods such as rice, flour and biscuits have been used to prepare fortified iron products. However, it is important to ensure the fortification process does not exert significant negative effects on organoleptic properties and the shelf life of the food product.
Collapse
Affiliation(s)
- Dipun Nirmal Perera
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | | | - Dasuni Dilkini Eapasinghe
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | - Dilmi Maleesha Liyanage
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | - R A C Haily Seneviratne
- Department of Food Sciences Technology, Faculty of Livestock Fisheries and Nutrition, Wayamba University of Sri Lanka, Kuliyapitiya, Sri Lanka
| | - S M D Demini
- Lanka ORIX Leasing Company (LOLC) Advanced Technologies (Pvt) Ltd, Ethul Kotte, Sri Lanka
| | - J A S M Jayasinghe
- Lanka ORIX Leasing Company (LOLC) Advanced Technologies (Pvt) Ltd, Ethul Kotte, Sri Lanka
| | - Mishal Faizan
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | | | - B Prasanna Galhena
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | - Hasi Hays
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | - Kanishka Senathilake
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | - Kamani H Tennekoon
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | - Sameera R Samarakoon
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
28
|
Liu R, Yang J, Li Y, Xie J, Wang J. Heme oxygenase-1: The roles of both good and evil in neurodegenerative diseases. J Neurochem 2023; 167:347-361. [PMID: 37746863 DOI: 10.1111/jnc.15969] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/26/2023]
Abstract
Heme oxygenase-1 (HO-1) is the only way for cells to decompose heme. It can cleave heme to produce carbon monoxide (CO), ferrous iron (Fe2+ ), and biliverdin (BV). BV is reduced to bilirubin (BR) by biliverdin reductase(BVR). In previous studies, HO-1 was considered to have protective effects because of its anti-inflammatory, anti-apoptosis, and antiproliferation functions. However, emerging experimental studies have found that the metabolites derived from HO-1 can cause increase iin intracellular oxidative stress, mitochondrial damage, iron death, and autophagy. Because of its particularity, it is very meaningful to understand its exact mechanism. In this review, we summarized the protective and toxic effects of HO-1, its potential mechanism, its role in neurodegenerative diseases and related drug research. This knowledge may be beneficial to the development of new therapies for neurodegenerative diseases and is crucial to the development of new therapeutic strategies and biomarkers.
Collapse
Affiliation(s)
- Rong Liu
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Jiahua Yang
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Yinghui Li
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Jun Wang
- School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Rahimi K, Shirvani N, Sanaie P, Javadi A, Khademi M. The effects of alpha-pinene on the Nrf2-HO1 signaling pathway in gastric damage in rats. Mol Biol Rep 2023; 50:8615-8622. [PMID: 37648947 DOI: 10.1007/s11033-023-08765-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Alpha-pinene (α-pinene) is a monoterpene with gastroprotective activity. We evaluated the gastroprotective effect of α-pinene in the gastric damage model with ethanol. METHODS We evaluated the macroscopic evaluation of the stomach cavity, alteration in pH, mRNA expression of nuclear factor erythroid 2- related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), total antioxidant capacity (TAC) levels, and histopathological changes. RESULTS Pretreatment with α-pinene (10, 50 and 100 mg/kg i.p.) before oral administration of ethanol reduced gastric mucosal damage by increasing the percentage of ulcer inhibition. Alpha-pinene also increased gastric pH similar to omeprazole. In addition, the histopathological examination showed that in the groups pretreated with α-pinene 50 and 100 mg/kg, and omeprazole20 mg/kg, the lesions were less than the control group. Moreover, α- pinene 10, 50, 100, and omeprazole 20 mg/kg upregulated the NRF2 and HO1. CONCLUSIONS Our results show that pretreatment with α-pinene is effective in reducing ethanol-induced gastric damage through regulation of Nrf2/HO-1.
Collapse
Affiliation(s)
- Kaveh Rahimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Negin Shirvani
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Parham Sanaie
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Arian Javadi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mahsa Khademi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
30
|
Lazzarato L, Bianchi L, Andolfo A, Granata A, Lombardi M, Sinelli M, Rolando B, Carini M, Corsini A, Fruttero R, Arnaboldi L. Proteomics Studies Suggest That Nitric Oxide Donor Furoxans Inhibit In Vitro Vascular Smooth Muscle Cell Proliferation by Nitric Oxide-Independent Mechanisms. Molecules 2023; 28:5724. [PMID: 37570694 PMCID: PMC10420201 DOI: 10.3390/molecules28155724] [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: 05/30/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Physiologically, smooth muscle cells (SMC) and nitric oxide (NO) produced by endothelial cells strictly cooperate to maintain vasal homeostasis. In atherosclerosis, where this equilibrium is altered, molecules providing exogenous NO and able to inhibit SMC proliferation may represent valuable antiatherosclerotic agents. Searching for dual antiproliferative and NO-donor molecules, we found that furoxans significantly decreased SMC proliferation in vitro, albeit with different potencies. We therefore assessed whether this property is dependent on their thiol-induced ring opening. Indeed, while furazans (analogues unable to release NO) are not effective, furoxans' inhibitory potency parallels with the electron-attractor capacity of the group in 3 of the ring, making this effect tunable. To demonstrate whether their specific block on G1-S phase could be NO-dependent, we supplemented SMCs with furoxans and inhibitors of GMP- and/or of the polyamine pathway, which regulate NO-induced SMC proliferation, but they failed in preventing the antiproliferative effect. To find the real mechanism of this property, our proteomics studies revealed that eleven cellular proteins (with SUMO1 being central) and networks involved in cell homeostasis/proliferation are modulated by furoxans, probably by interaction with adducts generated after degradation. Altogether, thanks to their dual effect and pharmacological flexibility, furoxans may be evaluated in the future as antiatherosclerotic molecules.
Collapse
Affiliation(s)
- Loretta Lazzarato
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy; (L.L.); (B.R.); (R.F.)
| | - Laura Bianchi
- Functional Proteomics Laboratory, Department of Life Sciences, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy;
| | - Annapaola Andolfo
- Proteomics and Metabolomics Facility (ProMeFa), Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy;
| | - Agnese Granata
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Matteo Lombardi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Matteo Sinelli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Barbara Rolando
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy; (L.L.); (B.R.); (R.F.)
| | - Marina Carini
- Department of Pharmaceutical Sciences “Pietro Pratesi”, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy;
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| | - Roberta Fruttero
- Department of Drug Science and Technology, Università degli Studi di Torino, Via Pietro Giuria 9, 10125 Torino, Italy; (L.L.); (B.R.); (R.F.)
| | - Lorenzo Arnaboldi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (A.G.); (M.L.); (M.S.); (A.C.)
| |
Collapse
|
31
|
Yuan X, Li L, Zhang Y, Ai R, Li D, Dou Y, Hou M, Zhao D, Zhao S, Nan Y. Heme oxygenase 1 alleviates nonalcoholic steatohepatitis by suppressing hepatic ferroptosis. Lipids Health Dis 2023; 22:99. [PMID: 37422643 DOI: 10.1186/s12944-023-01855-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/22/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Heme oxygenase 1 (HO-1) has an influential but insufficiently investigated effect on ferroptosis, which is a novel form of programmed cell death and may play an effect on nonalcoholic steatohepatitis (NASH). However, the understanding of the mechanism is limited. Herein, our study aimed to explore the mechanism and role of HO-1 in NASH ferroptosis. METHODS Hepatocyte conditional HO-1 knockout (HO-1HEPKO) C57BL/6J mice were established and fed a high-fat diet (HFD). Additionally, wild-type mice were fed either a normal diet or a HFD. Hepatic steatosis, inflammation, fibrosis, lipid peroxidation, and iron overload were assessed. AML12 and HepG2 cells were used to investigate the underlying mechanisms in vitro. Finally, liver sections from NASH patients were used to clinically validate the histopathology of ferroptosis. RESULTS In mice, HFD caused lipid accumulation, inflammation, fibrosis, and lipid peroxidation, which were aggravated by HO-1HEPKO. In line with the in vivo results, HO-1 knockdown upregulated reactive oxygen species accumulation, lipid peroxidation, and iron overload in AML12 and HepG2 cells. Additionally, HO-1 knockdown reduced the GSH and SOD levels, which was in contrast to HO-1 overexpression in vitro. Furthermore, the present study revealed that the NF-κB signaling pathway was associated with ferroptosis in NASH models. Likewise, these findings were consistent with the liver histopathology results of NASH patients. CONCLUSION The current study showed that HO-1 could alleviate NASH progression by mediating ferroptosis.
Collapse
Affiliation(s)
- Xiwei Yuan
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Lu Li
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Ying Zhang
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Rong Ai
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Dongdong Li
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Yao Dou
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Mengmeng Hou
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Dandan Zhao
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Suxian Zhao
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Hebei Provincial Key Laboratory of liver fibrosis in chronic liver diseases, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
32
|
Tola AJ, Missihoun TD. Iron Availability Influences Protein Carbonylation in Arabidopsis thaliana Plants. Int J Mol Sci 2023; 24:ijms24119732. [PMID: 37298684 DOI: 10.3390/ijms24119732] [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: 04/28/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Protein carbonylation is an irreversible form of post-translational modification triggered by reactive oxygen species in animal and plant cells. It occurs either through the metal-catalyzed oxidation of Lys, Arg, Pro, and Thr side chains or the addition of α, β-unsaturated aldehydes and ketones to the side chains of Cys, Lys, and His. Recent genetic studies concerning plants pointed to an implication of protein carbonylation in gene regulation through phytohormones. However, for protein carbonylation to stand out as a signal transduction mechanism, such as phosphorylation and ubiquitination, it must be controlled in time and space by a still unknown trigger. In this study, we tested the hypothesis that the profile and extent of protein carbonylation are influenced by iron homeostasis in vivo. For this, we compared the profile and the contents of the carbonylated proteins in the Arabidopsis thaliana wild-type and mutant-deficient in three ferritin genes under normal and stress conditions. Additionally, we examined the proteins specifically carbonylated in wild-type seedlings exposed to iron-deficient conditions. Our results indicated that proteins were differentially carbonylated between the wild type and the triple ferritin mutant Fer1-3-4 in the leaves, stems, and flowers under normal growth conditions. The profile of the carbonylated proteins was also different between the wild type and the ferritin triple mutant exposed to heat stress, thus pointing to the influence of iron on the carbonylation of proteins. Consistent with this, the exposure of the seedlings to iron deficiency and iron excess greatly influenced the carbonylation of certain proteins involved in intracellular signal transduction, translation, and iron deficiency response. Overall, the study underlined the importance of iron homeostasis in the occurrence of protein carbonylation in vivo.
Collapse
Affiliation(s)
- Adesola J Tola
- Groupe de Recherche en Biologie Végétale (GRBV), Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G9A 5H7, Canada
| | - Tagnon D Missihoun
- Groupe de Recherche en Biologie Végétale (GRBV), Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G9A 5H7, Canada
| |
Collapse
|
33
|
Falco L, Tessitore V, Ciccarelli G, Malvezzi M, D’Andrea A, Imbalzano E, Golino P, Russo V. Antioxidant Properties of Oral Antithrombotic Therapies in Atherosclerotic Disease and Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1185. [PMID: 37371915 PMCID: PMC10294911 DOI: 10.3390/antiox12061185] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
The thrombosis-related diseases are one of the leading causes of illness and death in the general population, and despite significant improvements in long-term survival due to remarkable advances in pharmacologic therapy, they continue to pose a tremendous burden on healthcare systems. The oxidative stress plays a role of pivotal importance in thrombosis pathophysiology. The anticoagulant and antiplatelet drugs commonly used in the management of thrombosis-related diseases show several pleiotropic effects, beyond the antithrombotic effects. The present review aims to describe the current evidence about the antioxidant effects of the oral antithrombotic therapies in patients with atherosclerotic disease and atrial fibrillation.
Collapse
Affiliation(s)
- Luigi Falco
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, Italy; (L.F.); (V.T.); (G.C.); (M.M.); (P.G.)
| | - Viviana Tessitore
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, Italy; (L.F.); (V.T.); (G.C.); (M.M.); (P.G.)
| | - Giovanni Ciccarelli
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, Italy; (L.F.); (V.T.); (G.C.); (M.M.); (P.G.)
| | - Marco Malvezzi
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, Italy; (L.F.); (V.T.); (G.C.); (M.M.); (P.G.)
| | | | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Paolo Golino
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, Italy; (L.F.); (V.T.); (G.C.); (M.M.); (P.G.)
| | - Vincenzo Russo
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, Italy; (L.F.); (V.T.); (G.C.); (M.M.); (P.G.)
| |
Collapse
|
34
|
Tian M, Hu T, Ying J, Cui H, Huangfu N. Increased selenium and decreased iron levels in relation to risk of coronary artery disease in patients with diabetes. Front Nutr 2023; 10:1103330. [PMID: 37275636 PMCID: PMC10233138 DOI: 10.3389/fnut.2023.1103330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/03/2023] [Indexed: 06/07/2023] Open
Abstract
Background Observational studies have reported inconsistent associations between micronutrient levels and the risk of coronary artery disease (CAD) in diabetic patients. We aim to explore the causal association between genetically predicted concentrations of micronutrients (phosphorus, magnesium, selenium, iron, zinc, and copper) and CAD in patients with diabetes. Methods Single nucleotide polymorphisms (SNPs) connected to serum micronutrient levels were extracted from the corresponding published genome-wide association studies (GWASs). Summary-level statistics for CAD in diabetic patients were obtained from a GWAS of 15,666 patients with diabetes. The primary analysis was carried out with the inverse variance weighted approach, and sensitivity analyses using other statistical methods were further employed to assess the robustness of the results. Results Genetically predicted selenium level was causally associated with a higher risk of CAD in diabetic patients (odds ratio [OR]: 1.25; 95% confidence interval [CI]: 1.10-1.42; p = 5.01 × 10-4). While, genetically predicted iron concentrations in patients with diabetes were inversely associated with the risk of CAD (OR: 0.82; 95% CI: 0.75-0.90; p = 2.16 × 10-5). The association pattern kept robust in most sensitivity analyses. Nominally significant associations were observed for magnesium and copper with the risk of CAD in patients with diabetes. No consistent evidence was found for the causal associations between phosphorus and zinc levels, and the risk of CAD in patients with diabetes. Conclusion We provide consistent evidence for the causal effect of increased selenium and decreased iron levels on CAD in patients with diabetes, highlighting the necessity of micronutrient monitoring and application in these patients.
Collapse
Affiliation(s)
- Mengyun Tian
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, School of Medicine, Ningbo University, Ningbo, China
| | - Teng Hu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, School of Medicine, Ningbo University, Ningbo, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Ningbo, China
| | - Jiajun Ying
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
| | - Hanbin Cui
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, School of Medicine, Ningbo University, Ningbo, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Ningbo, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
| | - Ning Huangfu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, School of Medicine, Ningbo University, Ningbo, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Ningbo, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
| |
Collapse
|
35
|
Voltarelli VA, Alves de Souza RW, Miyauchi K, Hauser CJ, Otterbein LE. Heme: The Lord of the Iron Ring. Antioxidants (Basel) 2023; 12:antiox12051074. [PMID: 37237940 DOI: 10.3390/antiox12051074] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Heme is an iron-protoporphyrin complex with an essential physiologic function for all cells, especially for those in which heme is a key prosthetic group of proteins such as hemoglobin, myoglobin, and cytochromes of the mitochondria. However, it is also known that heme can participate in pro-oxidant and pro-inflammatory responses, leading to cytotoxicity in various tissues and organs such as the kidney, brain, heart, liver, and in immune cells. Indeed, heme, released as a result of tissue damage, can stimulate local and remote inflammatory reactions. These can initiate innate immune responses that, if left uncontrolled, can compound primary injuries and promote organ failure. In contrast, a cadre of heme receptors are arrayed on the plasma membrane that is designed either for heme import into the cell, or for the purpose of activating specific signaling pathways. Thus, free heme can serve either as a deleterious molecule, or one that can traffic and initiate highly specific cellular responses that are teleologically important for survival. Herein, we review heme metabolism and signaling pathways, including heme synthesis, degradation, and scavenging. We will focus on trauma and inflammatory diseases, including traumatic brain injury, trauma-related sepsis, cancer, and cardiovascular diseases where current work suggests that heme may be most important.
Collapse
Affiliation(s)
- Vanessa Azevedo Voltarelli
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rodrigo W Alves de Souza
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kenji Miyauchi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Carl J Hauser
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Leo Edmond Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
36
|
Sadeghi M, Fathi M, Gholizadeh Navashenaq J, Mohammadi H, Yousefi M, Hojjat-Farsangi M, Namdar A, Movasaghpour Akbari AA, Jadidi-Niaragh F. The prognostic and therapeutic potential of HO-1 in leukemia and MDS. Cell Commun Signal 2023; 21:57. [PMID: 36915102 PMCID: PMC10009952 DOI: 10.1186/s12964-023-01074-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 02/11/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Heme oxygenase-1 (HO-1), a heme-degrading enzyme, is proven to have anti-apoptotic effects in several malignancies. In addition, HO-1 is reported to cause chemoresistance and increase cell survival. Growing evidence indicates that HO-1 contributes to the course of hematological malignancies as well. Here, the expression pattern, prognostic value, and the effect of HO-1 targeting in HMs are discussed. MAIN BODY According to the recent literature, it was discovered that HO-1 is overexpressed in myelodysplastic syndromes (MDS), chronic myeloid leukemia (CML), acute myeloblastic leukemia (AML), and acute lymphoblastic leukemia (ALL) cells and is associated with high-risk disease. Furthermore, in addition to HO-1 expression by leukemic and MDS cells, CML, AML, and ALL leukemic stem cells express this protein as well, making it a potential target for eliminating minimal residual disease (MRD). Moreover, it was concluded that HO-1 induces tumor progression and prevents apoptosis through various pathways. CONCLUSION HO-1 has great potential in determining the prognosis of leukemia and MDS patients. HO-1 induces resistance to several chemotherapeutic agents as well as tyrosine kinase inhibitors and following its inhibition, chemo-sensitivity increases. Moreover, the exact role of HO-1 in Chronic Lymphocytic Leukemia (CLL) is yet unknown. While findings illustrate that MDS and other leukemic patients could benefit from HO-1 targeting. Future studies can help broaden our knowledge regarding the role of HO-1 in MDS and leukemia. Video abstract.
Collapse
Affiliation(s)
- Mohammad Sadeghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Fathi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Afshin Namdar
- Department of Immunology, University of Toronto, Toronto, Canada
| | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
37
|
Łoboda A, Dulak J. Nuclear Factor Erythroid 2-Related Factor 2 and Its Targets in Skeletal Muscle Repair and Regeneration. Antioxid Redox Signal 2023; 38:619-642. [PMID: 36597355 DOI: 10.1089/ars.2022.0208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Significance: Skeletal muscles have a robust regenerative capacity in response to acute and chronic injuries. Muscle repair and redox homeostasis are intimately linked; increased generation of reactive oxygen species leads to cellular dysfunction and contributes to muscle wasting and progression of muscle diseases. In exemplary muscle disease, Duchenne muscular dystrophy (DMD), caused by mutations in the DMD gene that encodes the muscle structural protein dystrophin, the regeneration machinery is severely compromised, while oxidative stress contributes to the progression of the disease. The nuclear factor erythroid 2-related factor 2 (NRF2) and its target genes, including heme oxygenase-1 (HO-1), provide protective mechanisms against oxidative insults. Recent Advances: Relevant advances have been evolving in recent years in understanding the mechanisms by which NRF2 regulates processes that contribute to effective muscle regeneration. To this end, pathways related to muscle satellite cell differentiation, oxidative stress, mitochondrial metabolism, inflammation, fibrosis, and angiogenesis have been studied. The regulatory role of NRF2 in skeletal muscle ferroptosis has been also suggested. Animal studies have shown that NRF2 pathway activation can stop or reverse skeletal muscle pathology, especially when endogenous stress defence mechanisms are imbalanced. Critical Issues: Despite the growing recognition of NRF2 as a factor that regulates various aspects of muscle regeneration, the mechanistic impact on muscle pathology in various models of muscle injury remains imprecise. Future Directions: Further studies are necessary to fully uncover the role of NRF2 in muscle regeneration, both in physiological and pathological conditions, and to investigate the possibilities for development of new therapeutic modalities. Antioxid. Redox Signal. 38, 619-642.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| |
Collapse
|
38
|
Chen S, Liu Y, Zhong Z, Wei C, Liu Y, Zhu X. Peritoneal immune microenvironment of endometriosis: Role and therapeutic perspectives. Front Immunol 2023; 14:1134663. [PMID: 36865552 PMCID: PMC9971222 DOI: 10.3389/fimmu.2023.1134663] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023] Open
Abstract
Endometriosis, an estrogen-dependent chronic inflammatory disease characterized by the growth of endometrium-like tissues outside the uterine cavity, affects 10% of reproductive-age women. Although the pathogenesis of endometriosis is uncertain, it is widely accepted that retrograde menstruation results in ectopic endometrial tissue implantation. Given that not all women with retrograde menstruation develop endometriosis, immune factors have been hypothesized to affect the pathogenesis of endometriosis. In this review, we demonstrate that the peritoneal immune microenvironment, including innate immunity and adaptive immunity, plays a central role in the pathogenesis of endometriosis. Current evidence supports the fact that immune cells, such as macrophages, natural killer (NK) cells, dendritic cells (DCs), neutrophils, T cells, and B cells, as well as cytokines and inflammatory mediators, contribute to the vascularization and fibrogenesis of endometriotic lesions, accelerating the implantation and development of ectopic endometrial lesions. Endocrine system dysfunction influences the immune microenvironment through overexpressed estrogen and progesterone resistance. In light of the limitations of hormonal therapy, we describe the prospects for potential diagnostic biomarkers and nonhormonal therapy based on the regulation of the immune microenvironment. Further studies are warranted to explore the available diagnostic biomarkers and immunological therapeutic strategies for endometriosis.
Collapse
Affiliation(s)
- Siman Chen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Yukai Liu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Zhiqi Zhong
- Xinglin College, Nantong University, Nantong, Jiangsu, China
| | - Chunyan Wei
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Yuyin Liu
- Department of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyong Zhu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China,Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China,*Correspondence: Xiaoyong Zhu,
| |
Collapse
|
39
|
The Influence of Lactoferrin in Plasma and Peritoneal Fluid on Iron Metabolism in Women with Endometriosis. Int J Mol Sci 2023; 24:ijms24021619. [PMID: 36675136 PMCID: PMC9863839 DOI: 10.3390/ijms24021619] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
The aim of this study was to investigate the relationship between lactoferrin and iron and its binding proteins in women with endometriosis by simultaneously measuring these parameters in plasma and peritoneal fluid. Ninety women were evaluated, of whom 57 were confirmed as having endometriosis. Lactoferrin was measured by ELISA, transferrin, ferritin and iron on a Cobas 8000 analyser. Lactoferrin and transferrin in peritoneal fluid were lower compared to plasma, in contrast to ferritin and iron. In plasma, lactoferrin showeds associations with iron and transferrin in endometriosis and with ferritin in the group without endometriosis. Lactoferrin in peritoneal fluid correlated with lactoferrin, iron and transferrin of plasma in patients without endometriosis. The ratio of lactoferrin concentration in peritoneal fluid to plasma differentiated stage I versus IV of endometriosis and was negatively correlated with the iron ratio in patients without endometriosis. The ferritin ratio differentiated women with and without endometriosis. The very high ferritin ratios, especially in advanced stages of endometriosis, suggest the protective involvement of this protein in peritoneal fluid and the loss of this role by lactoferrin. The results demonstrate the validity of assessing iron metabolism in women with endometriosis, which may be useful as a marker of the disease and its progression.
Collapse
|
40
|
Jo SY, Cho HJ, Kim TM. Fenoldopam Mesylate Enhances the Survival of Mesenchymal Stem Cells Under Oxidative Stress and Increases the Therapeutic Function in Acute Kidney Injury. Cell Transplant 2023; 32:9636897221147920. [PMID: 36594258 PMCID: PMC9830573 DOI: 10.1177/09636897221147920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have gained interest as an alternative therapeutic option for renal diseases, including acute kidney injury (AKI). However, their use is often limited owing to low survival rates in vivo. Fenoldopam mesylate (FD) is a selective dopamine D1 receptor agonist with antioxidative and anti-apoptotic roles. Herein, we investigated whether FD can enhance the survival of MSCs undergoing oxidative stress in vitro. In addition, the therapeutic effect of MSCs and FD-treated MSCs (FD-MSCs) was compared in a mouse model of AKI induced by cisplatin. The survival of MSCs under oxidative stress was augmented by FD treatment. FD induced the phosphorylation of cAMP response element-binding protein and AKT, contributing to enhanced growth compared with untreated MSCs. The expression of nuclear factor erythroid-2-related factor 2 (NRF2) and heme oxygenase-1 was increased by FD treatment, and nuclear translocation of NRF2 was found exclusively in FD-MSCs. FD downregulated BAX expression, increased the mitochondrial membrane potential, reduced reactive oxygen species generation, and decreased the apoptotic death of MSCs induced by oxidative stress. Moreover, renal function and tubular injury were improved in FD-MSCs compared with non-treated MSCs. Furthermore, tubular injury, apoptosis, and macrophage infiltration, as well as the serum level of tumor necrosis factor-α were reduced, while tubular cell proliferation was markedly increased in FD-MSCs compared with MSCs. Our study demonstrated that FD increases the survivability of MSCs in an oxidative environment, and its use may be effective in preparing robust therapeutic MSCs.
Collapse
Affiliation(s)
- Seo Yeon Jo
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, South Korea
| | - Hye Jin Cho
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, South Korea,Institutes of Green Bio Science & Technology, Seoul National University, Pyeongchang, South Korea,Tae Min Kim, Graduate School of International Agricultural Technology and Institutes of Green Bio Science & Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang 25354, Gangwon-do, South Korea.
| |
Collapse
|
41
|
Saliu AO, Akanji AM, Idowu AO. Luffa cylindrica (Linn. M. J. Roem) Reduces Oxidative Stress In Vivo in Plasmodium berghei-Infected Albino Mice. IBNOSINA JOURNAL OF MEDICINE AND BIOMEDICAL SCIENCES 2022. [DOI: 10.1055/s-0042-1758033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Background: Malaria is endemic in sub-Saharan Africa, and oxidative stress has been implicated in malaria disease. Luffa cylindrica is an ethnomedicinal plant used to treat various diseases, including malaria. The oxidative stress-reducing potential of L. cylindrica in malaria-disease state of Plasmodium berghei NK-65 parasite-infected mice was carried out in vivo.
Methods: Mice were infected with P. berghei NK-65, and the effect of administration of methanolic leaves extract (100, 200, and 400 mg/kg b.w) of L. cylindrica on percentage parasitemia in blood smear, antioxidant enzymes (catalase CAT, superoxide dismutase SOD, glutathione-s-transferase GST), non-enzymatic antioxidant (reduced glutathione GSH) and malondialdehyde concentration in tissues (plasma, liver, kidneys, and spleen) of mice was investigated and compared to chloroquine and artesunate as reference antimalarial drugs. Phytochemical constituents of the extract were determined by standard methods.
Results: Saponins, tannins, terpenes, phenolics, flavonoids, alkaloids, and glycosides were the phytochemical constituents identified in the extract. The extract at three doses (100, 200, and 400 mg/kg b.w.) investigated caused a significant reduction (p < 0.05) of parasite growth with over 90% reduction in parasitemia level in mice infected with the parasite. The extract also ameliorated oxidative stress in mice by significantly (p < 0.05) increasing the activities of CAT, SOD, and GST in the studied tissues of mice. The level of malondialdehyde, a marker of oxidative stress in mice, was also significantly (p < 0.05) reduced by the extract. The results were comparable with chloroquine- and artesunate-treated groups.
Conclusion: The study concludes that L. cylindrica is an effective therapy for treating malaria and for the management of its oxidative stress-related complications due to its antioxidant properties.
Collapse
Affiliation(s)
- Aduke Oluremi Saliu
- Department of Environmental Health Science, Faculty of Health Sciences, National Open University of Nigeria, Abuja, Nigeria
| | - Adewumi Musbau Akanji
- Department of Biochemistry, Faculty of Life Sciences, University of Ilorin, Kwara State, Nigeria
| | - Ayodeji Oluwafemi Idowu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Federal University of Health Sciences, Benue State, Nigeria
| |
Collapse
|
42
|
Nath KA, Singh RD, Croatt AJ, Adams CM. Heme Proteins and Kidney Injury: Beyond Rhabdomyolysis. KIDNEY360 2022; 3:1969-1979. [PMID: 36514409 PMCID: PMC9717624 DOI: 10.34067/kid.0005442022] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022]
Abstract
Heme proteins, the stuff of life, represent an ingenious biologic strategy that capitalizes on the biochemical versatility of heme, and yet is one that avoids the inherent risks to cellular vitality posed by unfettered and promiscuously reactive heme. Heme proteins, however, may be a double-edged sword because they can damage the kidney in certain settings. Although such injury is often viewed mainly within the context of rhabdomyolysis and the nephrotoxicity of myoglobin, an increasing literature now attests to the fact that involvement of heme proteins in renal injury ranges well beyond the confines of this single disease (and its analog, hemolysis); indeed, through the release of the defining heme motif, destabilization of intracellular heme proteins may be a common pathway for acute kidney injury, in general, and irrespective of the underlying insult. This brief review outlines current understanding regarding processes underlying such heme protein-induced acute kidney injury (AKI) and chronic kidney disease (CKD). Topics covered include, among others, the basis for renal injury after the exposure of the kidney to and its incorporation of myoglobin and hemoglobin; auto-oxidation of myoglobin and hemoglobin; destabilization of heme proteins and the release of heme; heme/iron/oxidant pathways of renal injury; generation of reactive oxygen species and reactive nitrogen species by NOX, iNOS, and myeloperoxidase; and the role of circulating cell-free hemoglobin in AKI and CKD. Also covered are the characteristics of the kidney that render this organ uniquely vulnerable to injury after myolysis and hemolysis, and pathobiologic effects emanating from free, labile heme. Mechanisms that defend against the toxicity of heme proteins are discussed, and the review concludes by outlining the therapeutic strategies that have arisen from current understanding of mechanisms of renal injury caused by heme proteins and how such mechanisms may be interrupted.
Collapse
Affiliation(s)
- Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher M. Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic Rochester, Minnesota
| |
Collapse
|
43
|
Zhang Y, Yang S, Qiu Z, Huang L, Huang L, Liang Y, Liu X, Wang M, Zhou B. Pyrogallol enhances therapeutic effect of human umbilical cord mesenchymal stem cells against LPS-mediated inflammation and lung injury via activation of Nrf2/HO-1 signaling. Free Radic Biol Med 2022; 191:66-81. [PMID: 36028178 DOI: 10.1016/j.freeradbiomed.2022.08.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/31/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022]
Abstract
The main challenges in clinical applications of mesenchymal stem cells (MSCs) are attributed to their heterogeneity. It is believed that preconditioning of MSCs with active compounds may enhance the expression of potentially therapeutic molecules and thus achieve stable and effective therapeutic outcomes. In the present study, we investigated the mechanism by which pyrogallol increased the therapeutic efficacy of human umbilical cord mesenchymal stem cells (hUCMSCs) against LPS-induced acute lung injury (ALI). hUCMSCs with pyrogallol treatment increased expression of HO-1 at both mRNA and protein levels, accompanied by Kelch-Like ECH-Associated Protein 1 (Keap1) degradation, and upregulation of the Nrf2 protein levels as well as nuclear translocation of Nrf2. Moreover, the modulation of Keap1 and Nrf2 as well as HO-1 upregulation by pyrogallol was reversed by pretreatment with N-acetylcysteine (NAC) and a P38 kinase inhibitor (SB203580). Whereas, NAC pretreatment abrogated pyrogallol-mediated activation of P38 kinase, indicating that pyrogallol-derived ROS led to P38 kinase activation, thus promoting Nrf2/HO-1 signaling. Additionally, we found that the induction of p62 by the pyrogallol-mediated ROS/P38/Nrf2 axis interacted with Keap1 and resulted in autophagic degradation of Keap1, which created a positive feedback loop to further release of Nrf2. Furthermore, the increased expression of HO-1 in pyrogallol-pretreated hUCMSCs led to enhanced inhibitory effects on LPS-mediated TLR4/P-P65 signaling in BEAS-2B cells, resulting in increasing suppression of LPS-indued expression of a series of pro-inflammatory mediators. Compared to untreated hUCMSCs, Sprague-Dawley (SD) rats with pyrogallol-primed hUCMSCs transplantation showed enhanced improvements in LPS-mediated lung pathological alterations, the increased lung index (lung/body ratio), apoptosis of epithelial cells, the activation of TLR4/NF-κB signaling as well as the release of pro-inflammatory mediators. Together, these results suggested that hUCMSCs with pyrogallol pretreatment enhanced the therapeutic efficacy of hUCMSCs, which may provide a promising therapeutic strategy to maximize the therapeutic efficacy of hUCMSC-based therapy for treating LPS-associated ALI.
Collapse
Affiliation(s)
- Yuehan Zhang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Sushan Yang
- Department of Clinical Laboratory, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Zhenhua Qiu
- Department of Clinical Laboratory, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Linyan Huang
- Department of Hematopathology, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Yueyun Liang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Xuanyu Liu
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Maosheng Wang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China.
| | - Beixian Zhou
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China; Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 510315, Guangzhou, China.
| |
Collapse
|
44
|
The Role of Concomitant Nrf2 Targeting and Stem Cell Therapy in Cerebrovascular Disease. Antioxidants (Basel) 2022; 11:antiox11081447. [PMID: 35892653 PMCID: PMC9332234 DOI: 10.3390/antiox11081447] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the reality that a death from cerebrovascular accident occurs every 3.5 min in the United States, there are few therapeutic options which are typically limited to a narrow window of opportunity in time for damage mitigation and recovery. Novel therapies have targeted pathological processes secondary to the initial insult, such as oxidative damage and peripheral inflammation. One of the greatest challenges to therapy is the frequently permanent damage within the CNS, attributed to a lack of sufficient neurogenesis. Thus, recent use of cell-based therapies for stroke have shown promising results. Unfortunately, stroke-induced inflammatory and oxidative damage limit the therapeutic potential of these stem cells. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been implicated in endogenous antioxidant and anti-inflammatory activity, thus presenting an attractive target for novel therapeutics to enhance stem cell therapy and promote neurogenesis. This review assesses the current literature on the concomitant use of stem cell therapy and Nrf2 targeting via pharmaceutical and natural agents, highlighting the need to elucidate both upstream and downstream pathways in optimizing Nrf2 treatments in the setting of cerebrovascular disease.
Collapse
|
45
|
Differential Sensitivity of Two Leukemia Cell Lines towards Two Major Gas Plasma Products Hydrogen Peroxide and Hypochlorous Acid. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12157429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Oxidative stress has major implications for health and disease. At the same time, the term collectively describes the reactions to different types of reactive oxygen species (ROS) and oxidants, including hydrogen peroxide (H2O2) and hypochlorous acid (HOCl). However, how both compare in terms of cytotoxicity and mechanism of action is less known. Using two leukemia cell lines, Jurkat and THP-1, as model systems at similar cell concentrations, we found an 8-fold greater sensitivity of the former over the latter for H2O2 exposure. Unexpectantly, this was not the case with HOCl exposure. Jurkat cells were 2-fold more resistant to HOCl-induced cytotoxicity than THP-1 cells. In each cell type, the relatively more toxic oxidant also induced activation of caspases 3 and 7 at earlier time points, as time-lapse fluorescence microscopy revealed. The effects observed did not markedly correlate with changes in intracellular GSH and GSSG levels. In addition, siRNA-mediated knockdown of the Nrf2 target HMOX-1 encoding for HO-1 protein and the growth and survival factor IL-8 revealed Jurkat cells to become more sensitive to HOCl, while HO-1 and IL-8 siRNA-mediated knockdown in THP-1 cells produced greater sensitivity towards H2O2. siRNA-mediated knockdown of catalase increased oxidant sensitivity only negligibly. Collectively, the data suggest striking HOCl-resistance of Jurkat and H2O2 resistance of THP-1 cells, showing similar protective roles of HO-1 and IL-8, while caspase activation kinetics differ.
Collapse
|
46
|
Ferrer JLM, Garcia RL. Antioxidant Systems, lncRNAs, and Tunneling Nanotubes in Cell Death Rescue from Cigarette Smoke Exposure. Cells 2022; 11:2277. [PMID: 35892574 PMCID: PMC9330437 DOI: 10.3390/cells11152277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 12/10/2022] Open
Abstract
Cigarette smoke is a rich source of carcinogens and reactive oxygen species (ROS) that can damage macromolecules including DNA. Repair systems can restore DNA integrity. Depending on the duration or intensity of stress signals, cells may utilize various survival and adaptive mechanisms. ROS levels are kept in check through redundant detoxification processes controlled largely by antioxidant systems. This review covers and expands on the mechanisms available to cigarette smoke-exposed cancer cells for restoring the redox balance. These include multiple layers of transcriptional control, each of which is posited to be activated upon reaching a particular stress threshold, among them the NRF2 pathway, the AP-1 and NF-kB pathways, and, finally, TP53, which triggers apoptosis if extreme toxicity is reached. The review also discusses long noncoding RNAs, which have been implicated recently in regulating oxidative stress-with roles in ROS detoxification, the inflammatory response, oxidative stress-induced apoptosis, and mitochondrial oxidative phosphorylation. Lastly, the emerging roles of tunneling nanotubes in providing additional mechanisms for metabolic rescue and the regulation of redox imbalance are considered, further highlighting the expanded redox reset arsenal available to cells.
Collapse
Affiliation(s)
| | - Reynaldo L. Garcia
- Disease Molecular Biology and Epigenetics Laboratory, National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City 1101, Philippines;
| |
Collapse
|
47
|
Shahwan M, Alhumaydhi FA, Sharaf SE, Alghamdi BS, Baeesa S, Tayeb HO, Ashraf GM, Shamsi A. Computational insight into the binding of bryostatin 1 with ferritin: implication of natural compounds in Alzheimer's disease therapeutics. J Biomol Struct Dyn 2022:1-11. [PMID: 35787781 DOI: 10.1080/07391102.2022.2092552] [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: 10/17/2022]
Abstract
Neuronal damage in iron-sensitive brain regions occurs as a result of iron dyshomeostasis. Increased iron levels and iron-related pathogenic triggers are associated with neurodegenerative diseases, including Alzheimer's disease (AD). Ferritin is a key player involved in iron homeostasis. Major pathological hallmarks of AD are amyloid plaques, neurofibrillary tangles (NFTs) and synaptic loss that lead to cognitive dysfunction and memory loss. Natural compounds persist in being the most excellent molecules in the area of drug discovery because of their different range of therapeutic applications. Bryostatins are naturally occurring macrocyclic lactones that can be implicated in AD therapeutics. Among them, Bryostatin 1 regulates protein kinase C, a crucial player in AD pathophysiology, thus highlighting the importance of bryostatin 1 in AD management. Thus, this study explores the binding mechanism of Bryotstain 1 with ferritin. In this work, the molecular docking calculations revealed that bryostatin 1 has an appreciable binding potential towards ferritin by forming stable hydrogen bonds (H-bonds). Molecular dynamics simulation studies deciphered the binding mechanism and conformational dynamics of ferrritin-bryostatin 1 system. The analyses of root mean square deviation, root mean square fluctuations, Rg, solvent accessible surface area, H-bonds and principal component analysis revealed the stability of the ferritin-bryostatin 1 docked complex throughout the trajectory of 100 ns. Moreover, the free energy landscape analysis advocated that the ferritin-bryostatin 1 complex stabilized to the global minimum. Altogether, the present work delineated the binding of bryostatin 1 with ferritin that can be implicated in the management of AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Moyad Shahwan
- College of Pharmacy & Health sciences, Ajman University, Ajman, United Arab Emirates.,Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Sharaf E Sharaf
- Pharmaceutical Chemistry Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Clinical Research Administration, Executive Administration of Research and Innovation, King Abdullah Medical City in Holy Capital, Makkah, Saudi Arabia
| | - Badrah S Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saleh Baeesa
- Division of Neurosurgery, College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Haythum O Tayeb
- The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,Division of Neurology, Department of Internal Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.,Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| |
Collapse
|
48
|
Xiong NX, Huang JF, Li KX, Li SY, Zhao JH, Wang R, Ou J, Fan LF, Luo SW, Liu SJ. Comparative analysis on the immunoregulatory roles of ferritin M in hybrid fish (Carassius cuvieri ♀ × Carassius auratus red var ♂) and its parental species after bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2022; 126:197-210. [PMID: 35609760 DOI: 10.1016/j.fsi.2022.05.039] [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: 05/01/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Ferritin M is involved in the regulation of fish immunity. In this study, open reading frame (ORF) sequences of ferritin M from hybrid fish and its parental species were 534 bp. Tissue-specific analysis indicated that the highest level of ferritin M from red crucian carp was observed in kidney, while peaked expressions of ferritin M from white crucian carp and hybrid carp were observed in gill. Elevated levels of ferritin M from hybrid carp and its parental species were detected in immune-related tissues following Aeromonas hydrophila infection or in cultured fish cell lines after lipopolysaccharide (LPS) challenge. Ferritin M overexpression could attenuate NF-κB and TNFα promoter activity in their respective fish cells. Purified ferritin M fusion proteins elicited in vitro binding activity to A. hydrophila and Edwardsiella tarda, lowered bacterial dissemination to tissues and alleviated inflammatory response. Furthermore, treatment with ferritin M fusion proteins could mitigate bacteria-induced liver damage and rescue antioxidant activity. These results suggested that ferritin M in hybrid fish showed a similar immune defense against bacteria infection in comparison with those of its parental species.
Collapse
Affiliation(s)
- Ning-Xia Xiong
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Jin-Fang Huang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Ke-Xin Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Shi-Yun Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Jia-Hui Zhao
- Foreign Studies College, Hunan Normal University, Changsha, 410081, PR China
| | - Rou Wang
- Foreign Studies College, Hunan Normal University, Changsha, 410081, PR China
| | - Jie Ou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China
| | - Lan-Fen Fan
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, PR China
| | - Sheng-Wei Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China.
| | - Shao-Jun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, PR China.
| |
Collapse
|
49
|
Essential Protective Role of Catalytically Active Antibodies (Abzymes) with Redox Antioxidant Functions in Animals and Humans. Int J Mol Sci 2022; 23:ijms23073898. [PMID: 35409256 PMCID: PMC8999700 DOI: 10.3390/ijms23073898] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 12/13/2022] Open
Abstract
During the life of aerobic organisms, the oxygen resulting from numerous reactions is converted into reactive oxygen species (ROS). Many ROS are dangerous due to their high reactivity; they are strong oxidants, and react with various cell components, leading to their damage. To protect against ROS overproduction, enzymatic and non-enzymatic systems are evolved in aerobic cells. Several known non-enzymatic antioxidants have a relatively low specific antioxidant activity. Superoxide dismutases, catalase, glutathione peroxidase, glutathione S-transferase, thioredoxin, and the peroxiredoxin families are the most important enzyme antioxidants. Artificial antibodies catalyzing redox reactions using different approaches have been created. During the past several decades, it has been shown that the blood and various biological fluids of humans and animals contain natural antibodies that catalyze different redox reactions, such as classical enzymes. This review, for the first time, summarizes data on existing non-enzymatic antioxidants, canonical enzymes, and artificial or natural antibodies (abzymes) with redox functions. Comparing abzymes with superoxide dismutase, catalase, peroxide-dependent peroxidase, and H2O2-independent oxidoreductase activities with the same activities as classical enzymes was carried out. The features of abzymes with the redox activities are described, including their exceptional diversity in the optimal pH values, dependency and independence on various metal ions, and the reaction rate constants for healthy donors and patients with different autoimmune diseases. The entire body of evidence indicates that abzymes with redox antioxidant activities existing in the blood for a long time compared to enzymes are an essential part of the protection system of humans and animals from oxidative stress.
Collapse
|
50
|
Heme Oxygenase-1: An Anti-Inflammatory Effector in Cardiovascular, Lung, and Related Metabolic Disorders. Antioxidants (Basel) 2022; 11:antiox11030555. [PMID: 35326205 PMCID: PMC8944973 DOI: 10.3390/antiox11030555] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/24/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
The heme oxygenase (HO) enzyme system catabolizes heme to carbon monoxide (CO), ferrous iron, and biliverdin-IXα (BV), which is reduced to bilirubin-IXα (BR) by biliverdin reductase (BVR). HO activity is represented by two distinct isozymes, the inducible form, HO-1, and a constitutive form, HO-2, encoded by distinct genes (HMOX1, HMOX2, respectively). HO-1 responds to transcriptional activation in response to a wide variety of chemical and physical stimuli, including its natural substrate heme, oxidants, and phytochemical antioxidants. The expression of HO-1 is regulated by NF-E2-related factor-2 and counter-regulated by Bach-1, in a heme-sensitive manner. Additionally, HMOX1 promoter polymorphisms have been associated with human disease. The induction of HO-1 can confer protection in inflammatory conditions through removal of heme, a pro-oxidant and potential catalyst of lipid peroxidation, whereas iron released from HO activity may trigger ferritin synthesis or ferroptosis. The production of heme-derived reaction products (i.e., BV, BR) may contribute to HO-dependent cytoprotection via antioxidant and immunomodulatory effects. Additionally, BVR and BR have newly recognized roles in lipid regulation. CO may alter mitochondrial function leading to modulation of downstream signaling pathways that culminate in anti-apoptotic, anti-inflammatory, anti-proliferative and immunomodulatory effects. This review will present evidence for beneficial effects of HO-1 and its reaction products in human diseases, including cardiovascular disease (CVD), metabolic conditions, including diabetes and obesity, as well as acute and chronic diseases of the liver, kidney, or lung. Strategies targeting the HO-1 pathway, including genetic or chemical modulation of HO-1 expression, or application of BR, CO gas, or CO donor compounds show therapeutic potential in inflammatory conditions, including organ ischemia/reperfusion injury. Evidence from human studies indicate that HO-1 expression may represent a biomarker of oxidative stress in various clinical conditions, while increases in serum BR levels have been correlated inversely to risk of CVD and metabolic disease. Ongoing human clinical trials investigate the potential of CO as a therapeutic in human disease.
Collapse
|