1
|
Anash M, Maparu K, Singh S. Unraveling cell death mechanisms in traumatic brain injury: dynamic roles of ferroptosis and necroptosis. Mol Biol Rep 2025; 52:381. [PMID: 40208458 DOI: 10.1007/s11033-025-10489-0] [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: 01/10/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Traumatic brain injury (TBI) remains a major cause of mortality and long-term disability worldwide, with ferroptosis and necroptosis emerging as key drivers of secondary neuronal damage. Ferroptosis, characterized by iron-dependent lipid peroxidation and mitochondrial dysfunction, exacerbates oxidative stress and neuronal cell death. In parallel, necroptosis, mediated by receptor-interacting protein kinases (RIPK1 and RIPK3), amplifies inflammation through membrane rupture and the release of cellular components. Mitochondrial dynamics, involving fission and fusion processes, play a dual role in regulating these pathways. While mitochondrial fusion preserves cellular integrity and reduces oxidative stress, excessive mitochondrial fission driven by dynamin-related protein 1 (DRP1) accelerates necroptotic signaling and neuronal injury. This intricate interplay between ferroptosis, necroptosis, and mitochondrial dynamics highlights potential therapeutic targets. Modulating these pathways through tailored interventions could reduce neuronal damage, mitigate neuroinflammation, and improve functional outcomes in TBI patients. Advancing our understanding of these mechanisms is essential for developing precision therapies that address the complex pathology of traumatic brain injury.
Collapse
Affiliation(s)
- Mohd Anash
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Kousik Maparu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
2
|
Liu ZH, Zhai Y, Zhang J, Huang W, Li W, Qin W. Mitochondrial iron deficiency mediated inhibition of ecdysone synthesis underlies lead (Pb) induced developmental toxicity in Drosophila melanogaster. Toxicol Appl Pharmacol 2025; 497:117283. [PMID: 40020975 DOI: 10.1016/j.taap.2025.117283] [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: 08/13/2024] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Lead (Pb) is a pervasive heavy metal possessing developmental toxicity, at least in part, by disrupting iron homeostasis. In this study, we aimed to elucidate the underlying mechanism of iron deficiency mediated developmental defects in Pb exposed Drosophila melanogaster, mainly focusing on iron-dependent synthesis of ecdysone signaling, which plays a key role in the development of insects. Herein, we found Pb exposure resulted in iron deficiency in mitochondria by inhibiting expression of mitoferrin (evidenced by qPCR assay), the mitochondrial iron importer. Further study demonstrated that biosynthesis of ecdysone, a hormone synthesized with the help of iron-containing cytochrome P450s in mitochondria, was inhibited following Pb exposure. Ecdysone supplementation, to some extent, rescued Pb induced developmental delay and reproductive defects in Drosophila melanogaster. Furthermore, we found that disruption of mitoferrin and ecdysone synthesis was restored by NAC (N-Acetylcysteine, a well-known ROS scavenger), suggesting that oxidative stress plays a key role in Pb mediated mitochondrial iron dys-homeostasis and developmental toxicity. This study therefore revealed that mitochondrial iron deficiency mediated inhibition of ecdysone synthesis is a key event associated with iron dys-homeostasis mediated developmental defects caused by Pb exposure. Meanwhile, our study indicated that mitochondria may act as an important target of Pb, thus providing potential protective strategies against Pb toxicity.
Collapse
Affiliation(s)
- Zhi-Hua Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China.
| | - YuYin Zhai
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Jiakai Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Wei Huang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Wanrong Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Wenting Qin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| |
Collapse
|
3
|
Jia D, Huang W, Yin Q, Wang H, Wang Z, Zhang M, Gong W, Wang R, Zhu Y, Ji Y. Melatonin alleviates ferroptosis triggered by cadmium via regulating ferritinophagy and iron metabolism in spermatogonia. Sci Rep 2025; 15:8910. [PMID: 40087488 PMCID: PMC11909107 DOI: 10.1038/s41598-025-93822-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
Melatonin (Mel), a classical antioxidant, has the potential to mediate ferroptosis. Cadmium (Cd) poses a substantial threat to the male reproductive system, as it can induce testicular injury by triggering ferroptosis. The study aimed to explore the protective role and mechanism of Mel in Cd-induced ferroptosis in spermatogonia (spg). Our results demonstrated that Cd disrupted the mitochondrial ultrastructure and induced more autophagosomes in spg. Exposure to Cd resulted in a reduction of the mitochondrial membrane potential of the cells. The transcriptomics analysis revealed significant differences in gene expression associated with ferroptosis and autophagy. Mel could reverse the changes caused by Cd in the genes mentioned above. Furthermore, Cd increased cellular iron content and elevated reactive oxygen species levels, which induced oxidative stress in spg. Mel pretreatment reduced iron accumulation and oxidative damage caused by Cd exposure. Additional studies demonstrated that Cd exposure activated NCOA4-mediated ferritinophagy in spg. Mel pretreatment, as anticipated, inhibited the increased the mRNA and protein expression of ATG5, LC3B, and NCOA4 caused by Cd, ameliorated Cd-caused iron overload and oxidative stress, and protected spg from ferroptosis. Our study provides a therapeutic basis for the use of Mel to treat Cd-induced testicular injury.
Collapse
Affiliation(s)
- Didi Jia
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Wei Huang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Qizi Yin
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Han Wang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Ziyue Wang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Mingming Zhang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Wenjing Gong
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Rong Wang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Yan Zhu
- The Second Affiliated Hospital of Anhui, University of Traditional Chinese Medicine, Hefei, Anhui, China.
| | - Yanli Ji
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics / Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
4
|
Chen Z, Zheng N, Wang F, Zhou Q, Chen Z, Xie L, Sun Q, Li L, Li B. The role of ferritinophagy and ferroptosis in Alzheimer's disease. Brain Res 2025; 1850:149340. [PMID: 39586368 DOI: 10.1016/j.brainres.2024.149340] [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: 07/02/2024] [Revised: 10/17/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
Iron is a crucial mineral element within human cells, serving as a pivotal cofactor for diverse biological enzymes. Ferritin plays a crucial role in maintaining iron homeostasis within the body through its ability to sequester and release iron. Ferritinophagy is a selective autophagic process in cells that specifically facilitates the degradation of ferritin and subsequent release of free iron, thereby regulating intracellular iron homeostasis. The nuclear receptor coactivator 4 (NCOA4) serves as a pivotal regulator in the entire process of ferritinophagy, facilitating its binding to ferritin and subsequent delivering to lysosomes for degradation, thereby enabling the release of free iron. The free iron ions within the cell undergo catalysis through the Fenton reaction, resulting in a substantial generation of reactive oxygen species (ROS). This process induces lipid peroxidation, thereby stimulating a cascade leading to cellular tissue damage and subsequent initiation of ferroptosis. Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive deterioration of emotional memory and cognitive function, accompanied by mental and behavioral aberrations. The pathology of the disease is characterized by aberrant deposition of amyloid β-protein (Aβ) and hyperphosphorylated tau protein. It has been observed that evident iron metabolism disorders and accumulation of lipid peroxides occur in AD, indicating a significant impact of ferritinophagy and ferroptosis on the pathogenesis and progression of AD. This article elucidates the process and mechanism of ferritinophagy and ferroptosis, investigating their implications in AD to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ziwen Chen
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Nan Zheng
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Fuwei Wang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Qiong Zhou
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zihao Chen
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Lihua Xie
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Qiang Sun
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Li Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| | - Baohong Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
5
|
Gil J, Kim D, Choi S, Bae ON. Cadmium-induced iron dysregulation contributes to functional impairment in brain endothelial cells via the ferroptosis pathway. Toxicol Appl Pharmacol 2025; 495:117233. [PMID: 39842614 DOI: 10.1016/j.taap.2025.117233] [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: 10/04/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Cadmium (Cd2+) is a heavy metal that is a major hazardous environmental contaminant, ubiquitously present in the environment. Cd2+ exposure has been closely associated with an increased prevalence and severity of neurological and cardiovascular diseases (CVD). The blood-brain barrier (BBB) plays a crucial role in protecting the brain from external environmental factors. Mitochondria play an important role in maintaining the barrier function of brain endothelial cells by regulating energy metabolism and redox homeostasis. In this study, we aimed to assess the cytotoxic effects of Cd2+ on the integrity and function of brain endothelial cells. After 24 h of exposure, Cd2+ reduced cell survival, tight junction protein expression, and trans-endothelial electrical resistance (TEER) in bEnd.3 cells suggest a potential BBB integrity disruption by Cd2+ exposure. To clarify the underlying mechanism, we further investigated the role of mitochondria in iron overload-mediated cell death following Cd2+ exposure. Cd2+ induced a substantial reduction in mitochondrial basal respiration and ATP production in brain endothelial cells, suggesting mitochondrial dysfunction. In addition, Cd2+ exposure led to impaired autophagy, elevated iron levels, and increased lipid peroxidation, indicating the initiation of ferroptosis, a form of cell death triggered by iron. In summary, our research suggests that Cd2+ exposure can disrupt BBB function by causing mitochondrial dysfunction and disrupting iron homeostasis.
Collapse
Affiliation(s)
- Junkyung Gil
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Donghyun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Sungbin Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| |
Collapse
|
6
|
Zhao H, Ji QH, Jia ZZ, Shen LH. Association between deep gray matter iron deposition and clinical symptoms in Parkinson's disease: a quantitative susceptibility mapping study. Front Neurol 2025; 15:1442903. [PMID: 39835146 PMCID: PMC11743366 DOI: 10.3389/fneur.2024.1442903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Purpose This study aimed to assess the association between motor and non-motor symptoms of Parkinson's disease (PD) and iron accumulation within the deep gray matter of the brain by Quantitative Susceptibility Mapping (QSM). Methods Fifty-six PD patients and twenty-nine healthy controls were recruited in this study. According to the Hoehn and Yahr (H-Y) stage score, PD patients were divided into early stage (H-Y ≤ 2) and advanced stage (H-Y > 2) groups. Specifically, the Regions of Interest (ROIs) encompassed the substantia nigra (SN), red nucleus (RN), caudate nucleus (CN), globus pallidus (GP) and putamen (PT). Meanwhile, various rating scales were used to assess the clinical symptoms of PD. Results Compared to healthy controls (HCs), PD patients showed a significant increase in magnetic susceptibility values (MSVs) within the SN and GP. Further comparisons indicated that the MSVs of the SN, PT, GP and CN are all higher in advanced stages than in early stages. Significant positive correlations were observed between the MSVs of the SN and scores on the UPDRS-III, HAMA, and HAMD (r = 0.310, p = 0.020; r = 0.273, p = 0.042; r = 0.342, p = 0.010, respectively). Likewise, the MSVs of the GP demonstrated notable correlations with HAMA and HAMD scores (r = 0.275, p = 0.040; r = 0.415, p = 0.001). Additionally, a significant correlation was found between the MSVs of the PT and HAMD scores (r = 0.360, p = 0.006). Furthermore, we identified a significant negative correlation between MMSE scores and the MSVs of both the PT and GP (r = -0.268, p = 0.046; r = -0.305, p = 0.022). Conclusion Our study revealed that QSM possesses the capability to serve as a biomarker for PD. Significant correlations were found between clinical features and the iron deposition in the nigrostriatal system.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Department of Neurology, Affiliated Rudong Hospital of Xinglin College, Nantong University, Nantong, China
| | - Qiu-Hong Ji
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhong-Zheng Jia
- Department of Medical Imaging, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Li-Hua Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
7
|
Chipofya E, Docrat TF, Marnewick JL. The Neuroprotective Effect of Rooibos Herbal Tea Against Alzheimer's Disease: A Review. Mol Nutr Food Res 2025; 69:e202400670. [PMID: 39703045 PMCID: PMC11704843 DOI: 10.1002/mnfr.202400670] [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: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
The world is experiencing a demographic shift toward an increasing proportion of elderly persons. Alzheimer's disease (AD) and other neurological disorders are far more likely to develop as people age. AD is a gradual, irreversible, and degenerative brain disorder that progressively deteriorates memory and cognitive function, eventually leading to death. Treatment for AD is the most significant unmet clinical need in neurology. There are no effective treatment options to prevent or reverse the degenerative process. The current medical management focuses primarily on temporarily easing symptoms, with little or no overall improvement. Although genetic predisposition and lifestyle factors influence the risk of neurodegenerative disorders, recent research suggests that dietary polyphenols with solid antioxidant capacities play crucial roles in determining brain health and aging. Aspalathus linearis is used to produce Rooibos, a popular South African herbal tea, which may modulate neurodegenerative mechanisms such as oxidative stress, tau protein, amyloid plaques, inflammation, and metals, all of which have been associated with AD. We reviewed the literature to evaluate the potential neuroprotective effects of Rooibos and its major flavonoids and to understand the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Elias Chipofya
- Applied Microbial and Health Biotechnology InstituteCape Peninsula University of TechnologyCape TownSouth Africa
- Department of Biomedical SciencesFaculty of Health Sciences and Wellness SciencesCape Peninsula University of TechnologyCape TownSouth Africa
| | - Taskeen F. Docrat
- Applied Microbial and Health Biotechnology InstituteCape Peninsula University of TechnologyCape TownSouth Africa
| | - Jeanine L. Marnewick
- Applied Microbial and Health Biotechnology InstituteCape Peninsula University of TechnologyCape TownSouth Africa
| |
Collapse
|
8
|
Zhou X, Wang H, Yan B, Nie X, Chen Q, Yang X, Lei M, Guo X, Ouyang C, Ren Z. Ferroptosis in Cardiovascular Diseases and Ferroptosis-Related Intervention Approaches. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07642-5. [PMID: 39641901 DOI: 10.1007/s10557-024-07642-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Cardiovascular diseases (CVDs) are major public health problems that threaten the lives and health of individuals. The article has reviewed recent progresses about ferroptosis and ferroptosis-related intervention approaches for the treatment of CVDs and provided more references and strategies for targeting ferroptosis to prevent and treat CVDs. METHODS A comprehensive review was conducted using the literature researches. RESULTS AND DISCUSSION Many ferroptosis-targeted compounds and ferroptosis-related genes may be prospective targets for treating CVDs and our review provides a solid foundation for further studies about the detailed pathological mechanisms of CVDs. CONCLUSION There are challenges and limitations about the translation of ferroptosis-targeted potential therapies from experimental research to clinical practice. It warrants further exploration to pursure safer and more effective ferroptosis-targeted thereapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Xianpeng Zhou
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Hao Wang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Biao Yan
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xinwen Nie
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Qingjie Chen
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiaosong Yang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Min Lei
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiying Guo
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Changhan Ouyang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Zhanhong Ren
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China.
| |
Collapse
|
9
|
Zhao Y, Yang M, Liang X. The role of mitochondria in iron overload-induced damage. J Transl Med 2024; 22:1057. [PMID: 39587666 PMCID: PMC11587765 DOI: 10.1186/s12967-024-05740-4] [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: 08/20/2024] [Accepted: 10/06/2024] [Indexed: 11/27/2024] Open
Abstract
Iron overload is a pathological condition characterized by the abnormal accumulation of iron within the body, which may result from excessive iron intake, disorders of iron metabolism, or specific disease states. This condition can lead to significant health complications and may pose life-threatening risks. The excessive accumulation of iron can induce cellular stress, adversely affecting the structure and function of mitochondria, thereby compromising overall organ function. Given the critical role of mitochondria in cellular metabolism and homeostasis, it is imperative to investigate how mitochondrial dysfunction induced by iron overload contributes to disease progression, as well as to explore mitochondrial-related pathways as potential therapeutic targets for various iron overload disorders. This review examines the mechanisms by which mitochondria are implicated in iron overload-induced damage, including increased oxidative stress, mitochondrial DNA damage, and disruptions in energy metabolism. Additionally, it addresses the relationship between these processes and various forms of programmed cell death, as well as alterations in mitochondrial dynamics. Furthermore, the review discusses strategies aimed at alleviating and mitigating the complications associated with iron overload in patients by targeting mitochondrial pathways.
Collapse
Affiliation(s)
- Yangyang Zhao
- Department of Transfusion, Affiliated Hospital of North Sichuan Medical college, Nanchong, Sichuan, P.R. China
| | - Mengjiao Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical college, Nanchong, Sichuan, P.R. China
- Graduate School of Comprehensive Human Science, University of Tsukuba, Tsukuba, Japan
| | - Xiaoxue Liang
- Chengdu Qingbaijiang District People's Hospital, Chengdu, 610300, Sichuan, P.R. China.
| |
Collapse
|
10
|
Chen L, Yang Y, Zhang N, Che H, Wang Z, Han J, Wen M. DHA and EPA alleviate depressive-like behaviors in chronic sleep-deprived mice: Involvement of iron metabolism, oligodendrocyte-lipids peroxidation and the LCN2-NLRP3 signaling axis. Free Radic Biol Med 2024; 225:654-664. [PMID: 39447994 DOI: 10.1016/j.freeradbiomed.2024.10.298] [Citation(s) in RCA: 1] [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] [Received: 08/13/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024]
Abstract
Mounting evidence suggests that eicosapentaenoic acid (EPA) is superior to docosahexaenoic acid (DHA) in the treatment of depression, but the underlying mechanisms remain elusive. In the present study, the effect of DHA and EPA on depressive-like behaviors was investigated in chronic sleep-deprived (CSD) mice. Following the administration of EPA or DHA, investigations were conducted on depression-like behavior, myelin damage, iron dyshomeostasis, oligodendrocyte-lipids peroxidation, and neuroinflammation. As anticipated, EPA was more effective than DHA in ameliorating CSD-induced depression by increasing center preference and immobility time and concurrently shortening immobility latency. Both DHA and EPA mitigated myelin damage with EPA demonstrating superior benefits characterized by higher levels of Olig2, MBP, and FTH, as well as decreased oligodendrocyte-lipid peroxidation. The inhibition of activated astrocytes and the associated LCN2-NLRP3 signaling pathway was observed following both EPA and DHA supplementation. However, the inhibitory effect was more pronounced with EPA. Additionally, EPA outperformed DHA in mitigating microglial activation and M1/M2 polarization imbalance. Overall, this present study provides valuable insights into the anti-depressive effects of DHA and EPA, highlighting their role in inhibiting oligodendrocyte-lipids peroxidation and the LCN2-NLRP3 axis and corroborating the superiority of EPA in mediating antidepressant effects.
Collapse
Affiliation(s)
- Lu Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, Shandong Province, China
| | - Yueqi Yang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, Shandong Province, China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, Shandong Province, China
| | - Hongxia Che
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, Shandong Province, China
| | - Jun Han
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, Shandong Province, China
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, Shandong Province, China; Pet Nutrition Research and Development Center Gambol Pet Group Co.,Ltd, Liaocheng, 252000, Shandong Province, China.
| |
Collapse
|
11
|
Sun Y, Tian B, Liang J, Bu M, Deng X, Deng K, Jiang M, Chen BT. Hippocampal iron overload and spatial reference memory impairment: Insights from a rat model. Neurosci Lett 2024; 843:138014. [PMID: 39447723 DOI: 10.1016/j.neulet.2024.138014] [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: 08/09/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Brain iron overload may induce neuronal death and lead to cognitive impairment. The hippocampus is a critical limbic structure involved in memory. This study aimed to investigate iron overload and its role in hippocampal damage and memory impairment using a rat model. METHODS Young rats (2 weeks old) received intraperitoneal injections of high-dose iron solution (Group H, n = 10), low-dose iron solution (Group L, n = 10) and normal saline as control (Group D, n = 5). The Morris water maze (MWM) test was performed on all rats to evaluate their spatial reference memory by assessing their escape latency time and number of platform crossing. The iron content and neuronal damage in hippocampal tissue sections of the rats were assessed semi-quantitatively using diaminobenzidine (DAB)-enhanced Perl's Prussian blue (PPB) staining, and their correlation with spatial reference memory performance was evaluated. RESULTS The escape latency in Group H was significantly longer compared to Groups L and D (P < 0.05). The number of platform crossings was significantly lower in Group H than in Group L or D (P < 0.001). The neuronal cells in Group H had more brown iron deposits than those of Groups L and D. There were significant correlations between the severity of structural damage in the hippocampal tissue and the number of platform crossings (P1 = 0.001 for Group H; P2 = 0.043 for Group L). CONCLUSION This study showed an association between hippocampal iron-induced structural damage and spatial reference memory impairment in a rat model. This work should advance our understanding of hippocampal iron overload on cognitive functioning.
Collapse
Affiliation(s)
- Yihao Sun
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Bin Tian
- Department of Radiology, The Affiliated Jinyang Hospital of Guizhou Medical University, Guiyang, PR China
| | - Jiali Liang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Meiru Bu
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Xi Deng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Kemei Deng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China
| | - Muliang Jiang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China.
| | - Bihong T Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
12
|
Mohammadi S, Ghaderi S, Fatehi F. Putamen iron quantification in diseases with neurodegeneration: a meta-analysis of the quantitative susceptibility mapping technique. Brain Imaging Behav 2024; 18:1239-1255. [PMID: 38758278 DOI: 10.1007/s11682-024-00895-6] [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] [Accepted: 05/02/2024] [Indexed: 05/18/2024]
Abstract
Quantitative susceptibility mapping (QSM) is an MRI technique that accurately measures iron concentration in brain tissues. This meta-analysis synthesized evidence from 30 studies that used QSM to quantify the iron levels in the putamen. The PRISMA statement was adhered to when conducting the systematic reviews and meta-analyses. We conducted a meta-analysis using a random-effects model, as well as subgroup analyses (disease type, geographic region, field strength, coil, disease type, age, and sex) and sensitivity analysis. A total of 1247 patients and 1035 controls were included in the study. Pooled results showed a standardized mean difference (SMD) of 0.41 (95% CI 0.19 to 0.64), with the strongest effect seen in Alzheimer's disease (AD) at 1.01 (95% CI 0.50 to 1.52). Relapsing-remitting multiple sclerosis (RRMS) also showed increased putaminal iron at 0.37 (95% CI 0.177 to 0.58). No significant differences were observed in Parkinson's disease (PD). No significant differences were found between subgroups based on geographic region, field strength, coil, disease type, age, and sex. The studies revealed significant heterogeneity, with field strength as the primary source, while other factors, such as disease type, location, age, sex, and coil type, may have contributed. The sensitivity analysis showed that these factors did not have a significant influence on the overall results. In summary, this meta-analysis supports abnormalities in putaminal iron content across different diseases with neurodegeneration, especially AD and RRMS, as measured by QSM. This highlights the potential of QSM as an imaging biomarker to better understand disease mechanisms involving disturbances in brain iron homeostasis.
Collapse
Affiliation(s)
- Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Ghaderi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
- Neurology Department, University Hospitals of Leicester NHS Trust, Leicester, UK.
| |
Collapse
|
13
|
Khoshdooz S, Abbasi H, Abbasi MM. Iron-Status Indicators and HFE Gene Polymorphisms in Individuals with Amyotrophic Lateral Sclerosis: An Umbrella Review of Meta-analyses and Systematic Reviews. Biol Trace Elem Res 2024:10.1007/s12011-024-04391-2. [PMID: 39317854 DOI: 10.1007/s12011-024-04391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the progressive loss of motor neurons. Recent meta-analyses and systematic reviews suggest that HFE gene polymorphisms and iron-associated biomarkers may play a key role in the risk and occurrence of ALS. This umbrella study aimed to explore the roles of HFE gene polymorphisms and iron-associated biomarkers in individuals with ALS. A thorough search of three online scientific databases, namely Scopus, Web of Science, and PubMed, was conducted from their inception until September 13, 2024. The screening and selection processes were executed based on the PICO framework and eligibility criteria, followed by two independent reviewers. The Assessment of Multiple Systematic Reviews (AMSTAR)-2 and GRADE tools were utilized to assess the methodological quality and the certainty of evidence. Through an advanced search, 101 records were retrieved, of which eight meta-analyses and systematic reviews were selected for this umbrella review. A significant increase in iron concentrations was found in individuals with ALS compared to healthy controls (SMD, 0.26; 95% CI - 0.05, 0.57). Conversely, selected meta-analyses reported that serum transferrin concentrations in ALS patients were lower compared to healthy controls (SMD, - 0.15; 95% CI - 0.36, 0.05). Furthermore, mutations in H63D polymorphisms resulted in a 13% significant increase in the risk of ALS (OR, 1.13; 95% CI 1.05, 1.22). Our umbrella study of meta-analyses and systematic reviews reveals that individuals with ALS have lower serum concentrations of transferrin compared to healthy controls. Additionally, the H63D polymorphism in the HFE gene is associated with a slight increase in the risk of ALS. Future research should investigate broader aspects of iron-related biomarkers and HFE genes to elucidate their roles in ALS pathogenesis. Registration: Our umbrella study was registered in the International Prospective Register of Systematic Reviews (PROSPERO) with the identification number CRD42024559032 ( https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42024559032 ).
Collapse
Affiliation(s)
- Sara Khoshdooz
- Faculty of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Hamid Abbasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran.
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Mehdi Abbasi
- Student Research Committee, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Bouchab H, Ishaq A, Limami Y, Saretzki G, Nasser B, El Kebbaj R. Antioxidant Effects of Cactus Seed Oil against Iron-Induced Oxidative Stress in Mouse Liver, Brain and Kidney. Molecules 2024; 29:4463. [PMID: 39339457 PMCID: PMC11433720 DOI: 10.3390/molecules29184463] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
In recent times, exploring the protective potential of medicinal plants has attracted increasing attention. To fight reactive oxygen species (ROS), which are key players in hepatic, cerebral and renal diseases, scientists have directed their efforts towards identifying novel compounds with antioxidant effects. Due to its unique composition, significant attention has been given to Cactus Seed Oil (CSO). Iron, as a metal, can be a potent generator of reactive oxygen species, especially hydroxyl radicals, via the Fenton and Haber-Weiss reactions. Here, we employed ferrous sulfate (FeSO4) to induce oxidative stress and DNA damage in mice. Then, we used CSO and Colza oil (CO) and evaluated the levels of the antioxidants (superoxide dismutase [SOD], glutathione peroxidase [GPx] and glutathione [GSH]) as well as a metabolite marker for lipid peroxidation (malondialdehyde [MDA]) relating to the antioxidant balance in the liver, brain and kidney. In addition, we measured DNA damage levels in hepatic tissue and the effects of CSO on it. Our study found that iron-dependent GPx activity decreases in the liver and the kidney tissues. Additionally, while iron decreased SOD activity in the liver, it increased it in the kidney. Interestingly, iron treatment resulted in a significant increase in hepatic MDA levels. In contrast, in brain tissue, there was a significant decrease under iron treatment. In addition, we found varying protective effects of CSO in alleviating oxidative stress in the different tissues with ameliorating DNA damage after iron overload in a mouse liver model, adding compelling evidence to the protective potential of CSO.
Collapse
Affiliation(s)
- Habiba Bouchab
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (H.B.); (Y.L.)
- Higher Institute of Nursing Professions and Technical Health (ISPITS), Errachidia 52000, Morocco
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Science and Technology, Hassan First University of Settat, Settat 26000, Morocco;
- Biosciences Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE2 4HH, UK; (A.I.); (G.S.)
| | - Abbas Ishaq
- Biosciences Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE2 4HH, UK; (A.I.); (G.S.)
| | - Youness Limami
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (H.B.); (Y.L.)
| | - Gabriele Saretzki
- Biosciences Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE2 4HH, UK; (A.I.); (G.S.)
| | - Boubker Nasser
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Science and Technology, Hassan First University of Settat, Settat 26000, Morocco;
| | - Riad El Kebbaj
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (H.B.); (Y.L.)
| |
Collapse
|
15
|
Kerr NR, Mossman CW, Chou CH, Bunten JM, Kelty TJ, Childs TE, Rector RS, Arnold WD, Grisanti LA, Du X, Booth FW. Hindlimb immobilization induces insulin resistance and elevates mitochondrial ROS production in the hippocampus of female rats. J Appl Physiol (1985) 2024; 137:512-526. [PMID: 38961821 PMCID: PMC11424180 DOI: 10.1152/japplphysiol.00234.2024] [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: 04/01/2024] [Revised: 06/11/2024] [Accepted: 07/02/2024] [Indexed: 07/05/2024] Open
Abstract
Alzheimer's disease (AD) is the fifth leading cause of death in older adults, and treatment options are severely lacking. Recent findings demonstrate a strong relationship between skeletal muscle and cognitive function, with evidence supporting that muscle quality and cognitive function are positively correlated in older adults. Conversely, decreased muscle function is associated with a threefold increased risk of cognitive decline. Based on these observations, the purpose of this study was to investigate the negative effects of muscle disuse [via a model of hindlimb immobilization (HLI)] on hippocampal insulin sensitivity and mitochondrial function and identify the potential mechanisms involved. HLI for 10 days in 4-mo-old female Wistar rats resulted in the following novel findings: 1) hippocampal insulin resistance and deficits in whole body glucose homeostasis, 2) dramatically increased mitochondrial reactive oxygen species (ROS) production in the hippocampus, 3) elevated markers for amyloidogenic cleavage of amyloid precursor protein (APP) and tau protein in the hippocampus, 4) and reduced brain-derived neurotrophic factor (BDNF) expression. These findings were associated with global changes in iron homeostasis, with muscle disuse producing muscle iron accumulation in association with decreased serum and whole brain iron levels. We report the novel finding that muscle disuse alters brain iron homeostasis and reveal a strong negative correlation between muscle and brain iron content. Overall, HLI-induced muscle disuse has robust negative effects on hippocampal insulin sensitivity and ROS production in association with altered brain iron homeostasis. This work provides potential novel mechanisms that may help explain how loss of muscle function contributes to cognitive decline and AD risk.NEW & NOTEWORTHY Muscle disuse via hindlimb immobilization increased oxidative stress and insulin resistance in the hippocampus. These findings were in association with muscle iron overload in connection with iron dysregulation in the brain. Overall, our work identifies muscle disuse as a contributor to hippocampal dysfunction, potentially through an iron-based muscle-brain axis, highlighting iron dysregulation as a potential novel mechanism in the relationship between muscle health, cognitive function, and Alzheimer's disease risk.
Collapse
Affiliation(s)
- Nathan R Kerr
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Chandler W Mossman
- Veterinary Medical Diagnostic Laboratory, University of Missouri, Columbia, Missouri, United States
| | - Chih-Hsuan Chou
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Joshua M Bunten
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Taylor J Kelty
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | - Thomas E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Randy Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Research Service, Harry S. Truman Memorial Veterans Medical Center, University of Missouri, Columbia, Missouri, United States
- Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - William David Arnold
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Physical Medicine and Rehabilitation, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Department of Neurology, University of Missouri, Columbia, Missouri, United States
| | - Laurel A Grisanti
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Xiangwei Du
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Veterinary Medical Diagnostic Laboratory, University of Missouri, Columbia, Missouri, United States
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
16
|
Luo Y, Zhu J, Hu Z, Luo W, Du X, Hu H, Peng S. Progress in the Pathogenesis of Diabetic Encephalopathy: The Key Role of Neuroinflammation. Diabetes Metab Res Rev 2024; 40:e3841. [PMID: 39295168 DOI: 10.1002/dmrr.3841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 09/21/2024]
Abstract
Diabetic encephalopathy (DE) is a severe complication that occurs in the central nervous system (CNS) and leads to cognitive impairment. DE involves various pathophysiological processes, and its pathogenesis is still unclear. This review summarised current research on the pathogenesis of diabetic encephalopathy, which involves neuroinflammation, oxidative stress, iron homoeostasis, blood-brain barrier disruption, altered gut microbiota, insulin resistance, etc. Among these pathological mechanisms, neuroinflammation has been focused on. This paper summarises some of the molecular mechanisms involved in neuroinflammation, including the Mammalian Target of Rapamycin (mTOR), Lipocalin-2 (LCN-2), Pyroptosis, Advanced Glycosylation End Products (AGEs), and some common pro-inflammatory factors. In addition, we discuss recent advances in the study of potential therapeutic targets for the treatment of DE against neuroinflammation. The current research on the pathogenesis of DE is progressing slowly, and more research is needed in the future. Further study of neuroinflammation as a mechanism is conducive to the discovery of more effective treatments for DE in the future.
Collapse
Affiliation(s)
- Yifan Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jinxi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haijun Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Bogdańska-Chomczyk E, Wojtacha P, Tsai ML, Huang ACW, Kozłowska A. Age-related changes in the architecture and biochemical markers levels in motor-related cortical areas of SHR rats-an ADHD animal model. Front Mol Neurosci 2024; 17:1414457. [PMID: 39246601 PMCID: PMC11378348 DOI: 10.3389/fnmol.2024.1414457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/25/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder whose exact pathophysiology has not been fully understood yet. Numerous studies have suggested disruptions in the cellular architecture and neuronal activity within brain structures of individuals with ADHD, accompanied by imbalances in the immune system, oxidative stress, and metabolism. Methods This study aims to assess two functionally and histologically distinct brain areas involved in motor control and coordination: the motor cortex (MC) and prefrontal cortex (PFC). Namely, the morphometric analysis of the MC throughout the developmental stages of Spontaneously Hypertensive Rats (SHRs) and Wistar Kyoto Rats (WKYs). Additionally, the study aimed to investigate the levels and activities of specific immune, oxidative stress, and metabolic markers in the PFC of juvenile and maturing SHRs in comparison to WKYs. Results The most significant MC volume reductions occurred in juvenile SHRs, accompanied by alterations in neuronal density in these brain areas compared to WKYs. Furthermore, juvenile SHRs exhibit heightened levels and activity of various markers, including interleukin-1α (IL-1α), IL-6, serine/threonine-protein mammalian target of rapamycin, RAC-alpha serine/threonine-protein kinase, glucocorticoid receptor β, malondialdehyde, sulfhydryl groups, superoxide dismutase, peroxidase, glutathione reductase, glutathione S-transferase, glucose, fructosamine, iron, lactic acid, alanine, aspartate transaminase, and lactate dehydrogenase. Discussion Significant changes in the MC morphometry and elevated levels of inflammatory, oxidative, and metabolic markers in PFC might be associated with disrupted brain development and maturation in ADHD.
Collapse
Affiliation(s)
- E Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - P Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - M L Tsai
- Department of Biomechatronic Engineering, National Ilan University, Yilan, Taiwan
| | - A C W Huang
- Department of Psychology, Fo Guang University, Yilan, Taiwan
| | - A Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
18
|
Agostini F, Sgalletta B, Bisaglia M. Iron Dyshomeostasis in Neurodegeneration with Brain Iron Accumulation (NBIA): Is It the Cause or the Effect? Cells 2024; 13:1376. [PMID: 39195264 PMCID: PMC11352641 DOI: 10.3390/cells13161376] [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: 07/22/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Iron is an essential metal ion implicated in several cellular processes. However, the reactive nature of iron renders this metal ion potentially dangerous for cells, and its levels need to be tightly controlled. Alterations in the intracellular concentration of iron are associated with different neuropathological conditions, including neurodegeneration with brain iron accumulation (NBIA). As the name suggests, NBIA encompasses a class of rare and still poorly investigated neurodegenerative disorders characterized by an abnormal accumulation of iron in the brain. NBIA is mostly a genetic pathology, and to date, 10 genes have been linked to familial forms of NBIA. In the present review, after the description of the principal mechanisms implicated in iron homeostasis, we summarize the research data concerning the pathological mechanisms underlying the genetic forms of NBIA and discuss the potential involvement of iron in such processes. The picture that emerges is that, while iron overload can contribute to the pathogenesis of NBIA, it does not seem to be the causal factor in most forms of the pathology. The onset of these pathologies is rather caused by a combination of processes involving the interplay between lipid metabolism, mitochondrial functions, and autophagic activity, eventually leading to iron dyshomeostasis.
Collapse
Affiliation(s)
- Francesco Agostini
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Bibiana Sgalletta
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Marco Bisaglia
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, 35121 Padova, Italy
| |
Collapse
|
19
|
Guo C, Peng J, Cheng P, Yang C, Gong S, Zhang L, Zhang T, Peng J. Mechanistic elucidation of ferroptosis and ferritinophagy: implications for advancing our understanding of arthritis. Front Physiol 2024; 15:1290234. [PMID: 39022306 PMCID: PMC11251907 DOI: 10.3389/fphys.2024.1290234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/23/2024] [Indexed: 07/20/2024] Open
Abstract
In recent years, the emerging phenomenon of ferroptosis has garnered significant attention as a distinctive mode of programmed cell death. Distinguished by its reliance on iron and dependence on reactive oxygen species (ROS), ferroptosis has emerged as a subject of extensive investigation. Mechanistically, this intricate process involves perturbations in iron homeostasis, dampening of system Xc-activity, morphological dynamics within mitochondria, and the onset of lipid peroxidation. Additionally, the concomitant phenomenon of ferritinophagy, the autophagic degradation of ferritin, assumes a pivotal role by facilitating the liberation of iron ions from ferritin, thereby advancing the progression of ferroptosis. This discussion thoroughly examines the detailed cell structures and basic processes behind ferroptosis and ferritinophagy. Moreover, it scrutinizes the intricate web of regulators that orchestrate these processes and examines their intricate interplay within the context of joint disorders. Against the backdrop of an annual increase in cases of osteoarthritis, rheumatoid arthritis, and gout, these narrative sheds light on the intriguing crossroads of pathophysiology by dissecting the intricate interrelationships between joint diseases, ferroptosis, and ferritinophagy. The newfound insights contribute fresh perspectives and promising therapeutic avenues, potentially revolutionizing the landscape of joint disease management.
Collapse
Affiliation(s)
- Caopei Guo
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Jiaze Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Piaotao Cheng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Chengbing Yang
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Shouhang Gong
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Lin Zhang
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiachen Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, China
| |
Collapse
|
20
|
Wei H, Chen C, Di F, Sun C, Wang X, Sun M, Liu N, Zhang M, Li M, Zhang J, Zhang S, Liang X. PM 2.5-induced ferroptosis by Nrf2/Hmox1 signaling pathway led to inflammation in microglia. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 352:124130. [PMID: 38729511 DOI: 10.1016/j.envpol.2024.124130] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Particulate matter (PM) has been a dominant contributor to air contamination, which will enter the central nervous system (CNS), causing neurotoxicity. However, the biological mechanism is poorly identified. In this study, C57BL/6J mice were applied to evaluate the neurotoxicity of collected fine particulate matter (PM2.5), via oropharyngeal aspiration at two ambient equivalent concentrations. The Y-maze results showed that PM2.5 exposure in mice would lead to the damage in hippocampal-dependent working memory. In addition, cell neuroinflammation, microglial activation were detected in hippocampus of PM2.5-exposure mice. To confirm the underlying mechanism, the microarray assay was conducted to screen the differentially expressed genes (DEGs) in microglia after PM2.5 exposure, and the results indicated the enrichment of DEGs in ferroptosis pathways. Furthermore, Heme oxygenase-1 (Hmox1) was found to be one of the most remarkably upregulated genes after PM2.5 exposure for 24 h. And PM2.5 exposure induced ferroptosis with iron accumulation through heme degradation by Nrf2-mediated Hmox1 upregulation, which could be eliminated by Nrf2-inhibition. Meanwhile, Hmox1 antagonist zinc protoporphyrin IX (ZnPP) could protect BV2 cells from ferroptosis. The results taken together indicated that PM2.5 resulted in the ferroptosis by causing iron overload through Nrf2/Hmox1 signaling pathway, which could account for the inflammation in microglia.
Collapse
Affiliation(s)
- Haiyun Wei
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Chao Chen
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Fanglin Di
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Changhua Sun
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, 250014, China
| | - Xinzhi Wang
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Meng Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Natong Liu
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Min Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Meng Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xue Liang
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
21
|
Marupudi N, Xiong MP. Genetic Targets and Applications of Iron Chelators for Neurodegeneration with Brain Iron Accumulation. ACS BIO & MED CHEM AU 2024; 4:119-130. [PMID: 38911909 PMCID: PMC11191567 DOI: 10.1021/acsbiomedchemau.3c00066] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 06/25/2024]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of neurodegenerative diseases that are typically caused by a monogenetic mutation, leading to development of disordered movement symptoms such as dystonia, hyperreflexia, etc. Brain iron accumulation can be diagnosed through MRI imaging and is hypothesized to be the cause of oxidative stress, leading to the degeneration of brain tissue. There are four main types of NBIA: pantothenate kinase-associated neurodegeneration (PKAN), PLA2G6-associated neurodegeneration (PLAN), mitochondrial membrane protein-associated neurodegeneration (MKAN), and beta-propeller protein-associated neurodegeneration (BPAN). There are no causative therapies for these diseases, but iron chelators have been shown to have potential toward treating NBIA. Three chelators are investigated in this Review: deferoxamine (DFO), desferasirox (DFS), and deferiprone (DFP). DFO has been investigated to treat neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD); however, dose-related toxicity in these studies, as well as in PKAN studies, have shown that the drug still requires more development before it can be applied toward NBIA cases. Iron chelation therapies other than the ones currently in clinical use have not yet reached clinical studies, but they may possess characteristics that would allow them to access the brain in ways that current chelators cannot. Intranasal formulations are an attractive dosage form to study for chelation therapy, as this method of delivery can bypass the blood-brain barrier and access the CNS. Gene therapy differs from iron chelation therapy as it is a causal treatment of the disease, whereas iron chelators only target the disease progression of NBIA. Because the pathophysiology of NBIA diseases is still unclear, future courses of action should be focused on causative treatment; however, iron chelation therapy is the current best course of action.
Collapse
Affiliation(s)
- Neharika Marupudi
- Department of Pharmaceutical
& Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602-2352, United States
| | - May P. Xiong
- Department of Pharmaceutical
& Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602-2352, United States
| |
Collapse
|
22
|
Buoso C, Seifert M, Lang M, Griffith CM, Talavera Andújar B, Castelo Rueda MP, Fischer C, Doerrier C, Talasz H, Zanon A, Pramstaller PP, Schymanski EL, Pichler I, Weiss G. Dopamine‑iron homeostasis interaction rescues mitochondrial fitness in Parkinson's disease. Neurobiol Dis 2024; 196:106506. [PMID: 38648865 DOI: 10.1016/j.nbd.2024.106506] [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: 12/21/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Imbalances of iron and dopamine metabolism along with mitochondrial dysfunction have been linked to the pathogenesis of Parkinson's disease (PD). We have previously suggested a direct link between iron homeostasis and dopamine metabolism, as dopamine can increase cellular uptake of iron into macrophages thereby promoting oxidative stress responses. In this study, we investigated the interplay between iron, dopamine, and mitochondrial activity in neuroblastoma SH-SY5Y cells and human induced pluripotent stem cell (hiPSC)-derived dopaminergic neurons differentiated from a healthy control and a PD patient with a mutation in the α-synuclein (SNCA) gene. In SH-SY5Y cells, dopamine treatment resulted in increased expression of the transmembrane iron transporters transferrin receptor 1 (TFR1), ferroportin (FPN), and mitoferrin2 (MFRN2) and intracellular iron accumulation, suggesting that dopamine may promote iron uptake. Furthermore, dopamine supplementation led to reduced mitochondrial fitness including decreased mitochondrial respiration, increased cytochrome c control efficiency, reduced mtDNA copy number and citrate synthase activity, increased oxidative stress and impaired aconitase activity. In dopaminergic neurons derived from a healthy control individual, dopamine showed comparable effects as observed in SH-SY5Y cells. The hiPSC-derived PD neurons harboring an endogenous SNCA mutation demonstrated altered mitochondrial iron homeostasis, reduced mitochondrial capacity along with increased oxidative stress and alterations of tricarboxylic acid cycle linked metabolic pathways compared with control neurons. Importantly, dopamine treatment of PD neurons promoted a rescue effect by increasing mitochondrial respiration, activating antioxidant stress response, and normalizing altered metabolite levels linked to mitochondrial function. These observations provide evidence that dopamine affects iron homeostasis, intracellular stress responses and mitochondrial function in healthy cells, while dopamine supplementation can restore the disturbed regulatory network in PD cells.
Collapse
Affiliation(s)
- Chiara Buoso
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy; Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Martin Lang
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy
| | - Corey M Griffith
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Begoña Talavera Andújar
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | | | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Heribert Talasz
- Institute of Medical Biochemistry, Protein Core Facility, Biocenter Innsbruck, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | | | - Emma L Schymanski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy.
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
23
|
Song K, Liu X, Xu H, Li M, Zheng Q, Qi C, Wang X, Liu Y, Zheng P, Liu J. Cr(VI) induces ferroptosis in DF-1 cells by simultaneously perturbing iron homeostasis of ferritinophagy and mitophagy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 925:171818. [PMID: 38508245 DOI: 10.1016/j.scitotenv.2024.171818] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Hexavalent chromium [Cr(VI)] is an environmental pollutant known for its strong oxidizing and carcinogenic effects. However, its potential to induce ferroptosis in poultry remains poorly understood. This study aims to investigate the induction of ferroptosis by Cr(VI) in DF-1 cells and elucidate the underlying mechanisms. DF-1 cells exposed to Cr(VI) showed increased lipid reactive oxygen species and changes in ferroptosis marker genes (decreased expression of GPX4 and increased expression of COX2). Notably, the addition of the ferroptosis-specific inhibitor ferrostatin-1 (Fer-1) can reverse this effect. During the cell death process, Cr(VI) induced ferritinophagy, disrupting iron homeostasis and releasing labile iron ions. We predicted by docking that these iron ions would bind to mitochondrial membrane proteins through virtual docking. This binding was validated through colocalization analysis. In addition, Cr(VI) caused mitophagy, which releases additional ferrous ions. Therefore, Cr(VI) can induce the simultaneous release of ferrous ions through these pathways, thereby exacerbating lipid peroxidation and ultimately triggering ferroptosis in DF-1 cells. This study demonstrates that Cr(VI) can induce ferroptosis in DF-1 cells by disrupting intracellular iron homeostasis and providing valuable insights into the toxic effects of Cr(VI) in poultry and potentially other organisms.
Collapse
Affiliation(s)
- Kaimin Song
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Xiaoting Liu
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Huiling Xu
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Muzi Li
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Qi Zheng
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Changxi Qi
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Xiaozhou Wang
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Yongxia Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Pimiao Zheng
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China.
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, Shandong 271018, China.
| |
Collapse
|
24
|
Forma A, Grunwald A, Zembala P, Januszewski J, Brachet A, Zembala R, Świątek K, Baj J. Micronutrient Status and Breast Cancer: A Narrative Review. Int J Mol Sci 2024; 25:4968. [PMID: 38732186 PMCID: PMC11084730 DOI: 10.3390/ijms25094968] [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: 03/14/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Breast cancer is one of the most common cancers worldwide, at the same time being one of the most prevalent causes of women's death. Many factors such as alcohol, weight fluctuations, or hormonal replacement therapy can potentially contribute to breast cancer development and progression. Another important factor in breast cancer onset includes micronutrient status. In this narrative review, we analyzed 23 micronutrients and their possible influence on breast cancer onset and progression. Further, the aim of this study was to investigate the impact of micronutrient status on the prevention of breast cancer and its possible influence on various therapeutic pathways. We researched meta-analyses, systemic and narrative reviews, retrospective studies, as well as original studies on human and animal models. The results of these studies indicate a possible correlation between the different levels of micronutrients and a decreased risk of breast cancer as well as a better survival rate. However, further studies are necessary to establish adequate doses of supplementation of the chosen micronutrients and the exact mechanisms of micronutrient impact on breast cancer therapy.
Collapse
Affiliation(s)
- Alicja Forma
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Arkadiusz Grunwald
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Patryk Zembala
- Faculty of Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Jacek Januszewski
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| | - Adam Brachet
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Roksana Zembala
- Faculty of Medicine, Cardinal Stefan Wyszynski University in Warsaw, Wóycickiego 1/3, 01-938 Warsaw, Poland;
| | - Kamila Świątek
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| |
Collapse
|
25
|
Fan Y, Ma J, Yang D, Li X, Liang K, She Z, Qi X, Shi X, Gu Q, Zheng J, Li D. Clinical findings of hyperechoic substantia nigra in patients with Parkinson's disease. Eur J Neurosci 2024; 59:2702-2714. [PMID: 38469656 DOI: 10.1111/ejn.16308] [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: 07/12/2023] [Revised: 01/06/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024]
Abstract
This study aims to analyse hyperechoic substantia nigra (HSN) characteristics and the correlation of HSN with clinical features and blood biomarkers in patients with Parkinson's disease (PD). Transcranial sonography (TCS) evaluations of the substantia nigra (SN) were performed in 40 healthy controls and 71 patients with PD, including patients with SN hyperechogenicity (SN+) and those with normal SN echogenicity (SN-). Evaluation of motor and non-motor symptoms was assessed by a series of rating scales. The uricase method was used to determine serum uric acid (UA) levels, and enzyme-linked immunosorbent assay (ELISA) was used to measure plasma interleukin (IL)-1β levels. TCS showed 92.50% specificity and 61.97% sensitivity in differentiating PD patients from controls. The area of SN+ contralateral to the side of initial motor symptoms (SNcontra) was larger than that ipsilateral to the side of initial motor symptoms (SNipsi). The PDSN+ group had lower Argentine Hyposmia Rating Scale (AHRS) scores and UA levels than the PDSN- group. Binary logistic regression analysis revealed that AHRS scores and UA levels could be independent predictors for HSN. The larger SN echogenic area (SNL) sizes positively correlated with plasma IL-1β levels in PD patients with SN+. The present study provides further evidence of the potential of SN echogenicity as an imaging biomarker for PD diagnosis. PD patients with HSN have more severe non-motor symptoms of hyposmia. HSN in PD patients is related to the mechanism of abnormal iron metabolism and microglial activation.
Collapse
Affiliation(s)
- Yongyan Fan
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jianjun Ma
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, China
| | - Dawei Yang
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiaohuan Li
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Keke Liang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, China
| | - Zonghan She
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xuelin Qi
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiaoxue Shi
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, China
| | - Qi Gu
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, China
| | - Jinhua Zheng
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, China
| | - Dongsheng Li
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, China
| |
Collapse
|
26
|
Kapper C, Oppelt P, Ganhör C, Gyunesh AA, Arbeithuber B, Stelzl P, Rezk-Füreder M. Minerals and the Menstrual Cycle: Impacts on Ovulation and Endometrial Health. Nutrients 2024; 16:1008. [PMID: 38613041 PMCID: PMC11013220 DOI: 10.3390/nu16071008] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The role of minerals in female fertility, particularly in relation to the menstrual cycle, presents a complex area of study that underscores the interplay between nutrition and reproductive health. This narrative review aims to elucidate the impacts of minerals on key aspects of the reproductive system: hormonal regulation, ovarian function and ovulation, endometrial health, and oxidative stress. Despite the attention given to specific micronutrients in relation to reproductive disorders, there is a noticeable absence of a comprehensive review focusing on the impact of minerals throughout the menstrual cycle on female fertility. This narrative review aims to address this gap by examining the influence of minerals on reproductive health. Each mineral's contribution is explored in detail to provide a clearer picture of its importance in supporting female fertility. This comprehensive analysis not only enhances our knowledge of reproductive health but also offers clinicians valuable insights into potential therapeutic strategies and the recommended intake of minerals to promote female reproductive well-being, considering the menstrual cycle. This review stands as the first to offer such a detailed examination of minerals in the context of the menstrual cycle, aiming to elevate the understanding of their critical role in female fertility and reproductive health.
Collapse
Affiliation(s)
- Celine Kapper
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Peter Oppelt
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
- Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Clara Ganhör
- Division of Pathophysiology, Institute of Physiology and Pathophysiology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- Clinical Research Institute for Cardiovascular and Metabolic Diseases, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Ayberk Alp Gyunesh
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Barbara Arbeithuber
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Patrick Stelzl
- Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Marlene Rezk-Füreder
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| |
Collapse
|
27
|
Mikulska J, Pietrzak D, Rękawek P, Siudaj K, Walczak-Nowicka ŁJ, Herbet M. Celiac disease and depressive disorders as nutritional implications related to common factors - A comprehensive review. Behav Brain Res 2024; 462:114886. [PMID: 38309373 DOI: 10.1016/j.bbr.2024.114886] [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: 12/14/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/05/2024]
Abstract
Celiac disease (CD) is an immune-mediated disease affecting the small intestine. The only treatment strategy for CD is the gluten-free diet (GFD). One of the more common mental disorders in CD patients is major depressive disorder (MDD). The influence of GFD on the occurrence of MDD symptoms in patients with CD will be evaluated. This diet often reduces nutritional deficiencies in these patients and also helps to reduce depressive symptoms. Both disease entities are often dominated by the same deficiencies of nutrients such as iron, zinc, selenium, iodine, or B and D vitamins. Deficiencies of particular components in CD can favor MDD and vice versa. Gluten can adversely affect the mental state of patients without CD. Also, intestinal microbiota may play an important role in the described process. This work aims to comprehensively assess the common factors involved in the pathomechanisms of MDD and CD, with particular emphasis on nutrient imbalances. Given the complexity of both disease entities, and the many common links, more research related to improving mental health in these patients and the implementation of a GFD would need to be conducted, but it appears to be a viable pathway to improving the quality of life and health of people struggling with CD and MDD. Therefore, probiotics, micronutrients, macronutrients, and vitamin supplements are recommended to reduce the risk of MDD, given that they may alleviate the symptoms of both these disease entities. In turn, in patients with MDD, it is worth considering testing for CD.
Collapse
Affiliation(s)
- Joanna Mikulska
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093 Lublin, Poland
| | - Diana Pietrzak
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093 Lublin, Poland
| | - Paweł Rękawek
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093 Lublin, Poland
| | - Krystian Siudaj
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093 Lublin, Poland
| | - Łucja Justyna Walczak-Nowicka
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093 Lublin, Poland.
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093 Lublin, Poland
| |
Collapse
|
28
|
Mohammadi S, Ghaderi S, Sayehmiri F, Fathi M. Quantitative susceptibility mapping for iron monitoring of multiple subcortical nuclei in type 2 diabetes mellitus: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1331831. [PMID: 38510699 PMCID: PMC10950952 DOI: 10.3389/fendo.2024.1331831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Iron accumulation in the brain has been linked to diabetes, but its role in subcortical structures involved in motor and cognitive functions remains unclear. Quantitative susceptibility mapping (QSM) allows the non-invasive quantification of iron deposition in the brain. This systematic review and meta-analysis examined magnetic susceptibility measured by QSM in the subcortical nuclei of patients with type 2 diabetes mellitus (T2DM) compared with controls. Methods PubMed, Scopus, and Web of Science databases were systematically searched [following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines] for studies reporting QSM values in the deep gray matter (DGM) regions of patients with T2DM and controls. Pooled standardized mean differences (SMDs) for susceptibility were calculated using fixed-effects meta-analysis models, and heterogeneity was assessed using I2. Sensitivity analyses were conducted, and publication bias was evaluated using Begg's and Egger's tests. Results Six studies including 192 patients with T2DM and 245 controls were included. This study found a significant increase in iron deposition in the subcortical nuclei of patients with T2DM compared to the control group. The study found moderate increases in the putamen (SMD = 0.53, 95% CI 0.33 to 0.72, p = 0.00) and dentate nucleus (SMD = 0.56, 95% CI 0.27 to 0.85, p = 0.00) but weak associations between increased iron levels in the caudate nucleus (SMD = 0.32, 95% CI 0.13 to 0.52, p = 0.00) and red nucleus (SMD = 0.22, 95% CI 0.00 0.44, p = 0.05). No statistical significance was found for iron deposition alterations in the globus pallidus (SMD = 0.19; 95% CI -0.01 to 0.38; p = 0.06) and substantia nigra (SMD = 0.12, 95% CI -0.10, 0.34, p = 0.29). Sensitivity analysis showed that the findings remained unaffected by individual studies, and consistent increases were observed in multiple subcortical areas. Discussion QSM revealed an increase in iron in the DGM/subcortical nuclei in T2DM patients versus controls, particularly in the motor and cognitive nuclei, including the putamen, dentate nucleus, caudate nucleus, and red nucleus. Thus, QSM may serve as a potential biomarker for iron accumulation in T2DM patients. However, further research is needed to validate these findings.
Collapse
Affiliation(s)
- Sana Mohammadi
- Department of Medical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sayehmiri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Kang T, Han Z, Zhu L, Cao B. TFR1 knockdown alleviates iron overload and mitochondrial dysfunction during neural differentiation of Alzheimer's disease-derived induced pluripotent stem cells by interacting with GSK3B. Eur J Med Res 2024; 29:101. [PMID: 38321571 PMCID: PMC10845644 DOI: 10.1186/s40001-024-01677-y] [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: 10/12/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Iron metabolism disorders are implicated in the pathogenesis of Alzheimer's disease (AD). It was previously reported that transferrin receptor (TFR1) expression was upregulated in AD mouse model. However, the precise biological functions of TFR1 in AD progression remains unclear. Herein, we observed a gradual increase in TFR1 protein expression during the differentiation of AD patient-derived induced pluripotent stem cells (AD-iPS). TFR1 knockdown inhibited the protein expression of ferritin and ferritin heavy chain 1 (FTH1), enhanced the expression of ferroportin 1 (FPN1), and decreased intracellular levels of total iron, labile iron, and reactive oxygen species (ROS). Moreover, TFR1 knockdown improved mitochondrial membrane potential (MMP), increased adenosine triphosphate (ATP) content, downregulated mitochondrial fission proteins, and upregulated mitochondrial fusion proteins. TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS, while TFR1 overexpression showed the opposite results. Additionally, TFR1interacted with glycogen synthase kinase 3 beta (GSK3B) and promoted GSK3B expression. GSK3B overexpression reversed the inhibitory effects of TFR1 knockdown on iron overload and mitochondrial dysfunction in AD-iPS differentiated neural cells. In conclusion, TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS by promoting GSK3B expression. Our findings provide a potential therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Tao Kang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Zheng Han
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Lijuan Zhu
- Department of Anesthesia, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Bingqing Cao
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| |
Collapse
|
30
|
Zhen D, Zhang S, Yang A, Li L, Cai Q, Grimes CA, Liu Y. A PEDOT enhanced covalent organic framework (COF) fluorescent probe for in vivo detection and imaging of Fe 3. Int J Biol Macromol 2024; 259:129104. [PMID: 38161014 DOI: 10.1016/j.ijbiomac.2023.129104] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/13/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Simple and accurate in vivo monitoring of Fe3+ is essential for gaining a better understanding of its role in physiological and pathological processes. A novel fluorescent probe was synthesized via in situ solid-state polymerization of 3,4-ethylenedioxythiophene (PEDOT) in the pore channels of a covalent organic framework (COF). The PEDOT@COF fluorescent probe exhibited an absolute quantum yield (QY) 3 times higher than COF. In the presence of Fe3+ the PEDOT@COF 475 nm fluorescence emission, 365 nm excitation, is quenched within 180 s. Fluorescence quenching is linear with Fe3+ in the concentration range of 0-960 μM, with a detection limit of 0.82 μM. The fluorescence quenching mechanism was attributed to inner filter effect (IEF), photoinduced electron transfer (PET) and static quenching (SQE) between PEDOT@COF and Fe3+. A paper strip-based detector was designed to facilitate practical applicability, and the PEDOT@COF probe successfully applied to fluorescence imaging of Fe3+ levels in vivo. This work details a tool of great promise for enabling detailed investigations into the role of Fe3+ in physiological and pathological diseases.
Collapse
Affiliation(s)
- Deshuai Zhen
- Hunan Key Laboratory of Typical Environment Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, PR China; State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Shaoqi Zhang
- Hunan Key Laboratory of Typical Environment Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, PR China; State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Aofeng Yang
- Hunan Key Laboratory of Typical Environment Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Le Li
- Hunan Key Laboratory of Typical Environment Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Craig A Grimes
- Flux Photon Corporation, 5950 Shiloh Road East, Alpharetta, GA 30005, United States
| | - Yu Liu
- Hunan Key Laboratory of Typical Environment Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, PR China; State Key Laboratory of Chemo/Biosensing and Chemometrics, School of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China.
| |
Collapse
|
31
|
Ago Y, Rintz E, Musini KS, Ma Z, Tomatsu S. Molecular Mechanisms in Pathophysiology of Mucopolysaccharidosis and Prospects for Innovative Therapy. Int J Mol Sci 2024; 25:1113. [PMID: 38256186 PMCID: PMC10816168 DOI: 10.3390/ijms25021113] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Mucopolysaccharidoses (MPSs) are a group of inborn errors of the metabolism caused by a deficiency in the lysosomal enzymes required to break down molecules called glycosaminoglycans (GAGs). These GAGs accumulate over time in various tissues and disrupt multiple biological systems, including catabolism of other substances, autophagy, and mitochondrial function. These pathological changes ultimately increase oxidative stress and activate innate immunity and inflammation. We have described the pathophysiology of MPS and activated inflammation in this paper, starting with accumulating the primary storage materials, GAGs. At the initial stage of GAG accumulation, affected tissues/cells are reversibly affected but progress irreversibly to: (1) disruption of substrate degradation with pathogenic changes in lysosomal function, (2) cellular dysfunction, secondary/tertiary accumulation (toxins such as GM2 or GM3 ganglioside, etc.), and inflammatory process, and (3) progressive tissue/organ damage and cell death (e.g., skeletal dysplasia, CNS impairment, etc.). For current and future treatment, several potential treatments for MPS that can penetrate the blood-brain barrier and bone have been proposed and/or are in clinical trials, including targeting peptides and molecular Trojan horses such as monoclonal antibodies attached to enzymes via receptor-mediated transport. Gene therapy trials with AAV, ex vivo LV, and Sleeping Beauty transposon system for MPS are proposed and/or underway as innovative therapeutic options. In addition, possible immunomodulatory reagents that can suppress MPS symptoms have been summarized in this review.
Collapse
Affiliation(s)
- Yasuhiko Ago
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
| | - Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland;
| | - Krishna Sai Musini
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Zhengyu Ma
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
| | - Shunji Tomatsu
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA; (Y.A.); (K.S.M.); (Z.M.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1112, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
32
|
Zhang G, Lv S, Zhong X, Li X, Yi Y, Lu Y, Yan W, Li J, Teng J. Ferroptosis: a new antidepressant pharmacological mechanism. Front Pharmacol 2024; 14:1339057. [PMID: 38259274 PMCID: PMC10800430 DOI: 10.3389/fphar.2023.1339057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
The incidence rate of depression, a mental disorder, is steadily increasing and has the potential to become a major global disability factor. Given the complex pathological mechanisms involved in depression, the use of conventional antidepressants may lead to severe complications due to their side effects. Hence, there is a critical need to explore the development of novel antidepressants. Ferroptosis, a newly recognized form of cell death, has been found to be closely linked to the onset of depression. Several studies have indicated that certain active ingredients can ameliorate depression by modulating the ferroptosis signaling pathway. Notably, traditional Chinese medicine (TCM) active ingredients and TCM prescriptions have demonstrated promising antidepressant effects in previous investigations owing to their unique advantages in antidepressant therapy. Building upon these findings, our objective was to review recent relevant research and provide new insights and directions for the development and application of innovative antidepressant strategies.
Collapse
Affiliation(s)
- Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xia Zhong
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangyu Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yunhao Yi
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitong Lu
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Yan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiamin Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Teng
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
33
|
Liu T, Wu H, Wei J. The Construction and Validation of a Novel Ferroptosis-Related Gene Signature in Parkinson's Disease. Int J Mol Sci 2023; 24:17203. [PMID: 38139032 PMCID: PMC10742934 DOI: 10.3390/ijms242417203] [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: 11/20/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
As a newly discovered regulated cell death mode, ferroptosis is associated with the development of Parkinson's disease (PD) and has attracted much attention. Nonetheless, the relationship between ferroptosis and PD pathogenesis remains unclear. The GSE8397 dataset includes GPL96 and GPL97 platforms. The differential genes were analyzed by immune infiltration and Gene Set Enrichment Analysis (GSEA) (p < 0.05), and differential multiple |logFC| > 1 and weighted gene coexpression network analysis (WGCNA) were used to screen differential expression genes (DEGs). The intersection with 368 ferroptosis-related genes (FRGs) was conducted for gene ontology/Kyoto encyclopedia of gene and genome (GO/KEGG) enrichment analysis, gene expression analysis, correlation analysis, single-cell sequencing analysis, and prognosis analysis (area under the curve, AUC) and to predict relevant miRNAs and construct network diagrams using Cytoscape. The intersection genes of differentially expressed ferroptosis-related genes (DEFRGs) and mitochondrial dysfunction genes were validated in the substantia nigra of MPTP-induced PD mice models by Western blotting and immunohistochemistry, and the protein-binding pocket was predicted using the DoGSiteScorer database. According to the results, the estimated scores were positively correlated with the stromal scores or immune scores in the GPL96 and GPL97 platforms. In the GPL96 platform, the GSEA showed that differential genes were mainly involved in the GnRH signaling pathway, B cell receptor signaling pathway, inositol phosphate metabolism, etc. In the GPL97 platform, the GSEA showed that differential genes were mainly involved in the ubiquitin-mediated proteolysis, axon guidance, Wnt signaling pathway, MAPK signaling pathway, etc. We obtained 26 DEFRGs, including 12 up-regulated genes and 14 down-regulated genes, with good correlation. The area under the prognostic analysis curve (AUC > 0.700) showed a good prognostic ability. We found that they were enriched in different neuronal cells, oligodendrocytes, astrocytes, oligodendrocyte precursor cells, and microglial cells, and their expression scores were positively correlated, and selected genes with an AUC curve ≥0.9 were used to predict miRNA, including miR-214/761/3619-5p, miR-203, miR-204/204b/211, miR-128/128ab, miR-199ab-5p, etc. For the differentially expressed ferroptosis-mitochondrial dysfunction-related genes (DEF-MDRGs) (AR, ISCU, SNCA, and PDK4), in the substantia nigra of mice, compared with the Saline group, the expression of AR and ISCU was decreased (p < 0.05), and the expression of α-Syn and PDK4 was increased (p < 0.05) in the MPTP group. Therapeutic drugs that target SNCA include ABBV-0805, Prasinezumab, Cinpanemab, and Gardenin A. The results of this study suggest that cellular DEF-MDRGs might play an important role in PD. AR, ISCU, SNCA, and PDK4 have the potential to be specific biomarkers for the early diagnosis of PD.
Collapse
Affiliation(s)
| | | | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; (T.L.)
| |
Collapse
|
34
|
Yang K, Zeng L, Zeng J, Deng Y, Wang S, Xu H, He Q, Yuan M, Luo Y, Ge A, Ge J. Research progress in the molecular mechanism of ferroptosis in Parkinson's disease and regulation by natural plant products. Ageing Res Rev 2023; 91:102063. [PMID: 37673132 DOI: 10.1016/j.arr.2023.102063] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder of the central nervous system after Alzheimer's disease. The current understanding of PD focuses mainly on the loss of dopamine neurons in the substantia nigra region of the midbrain, which is attributed to factors such as oxidative stress, alpha-synuclein aggregation, neuroinflammation, and mitochondrial dysfunction. These factors together contribute to the PD phenotype. Recent studies on PD pathology have introduced a new form of cell death known as ferroptosis. Pathological changes closely linked with ferroptosis have been seen in the brain tissues of PD patients, including alterations in iron metabolism, lipid peroxidation, and increased levels of reactive oxygen species. Preclinical research has demonstrated the neuroprotective qualities of certain iron chelators, antioxidants, Fer-1, and conditioners in Parkinson's disease. Natural plant products have shown significant potential in balancing ferroptosis-related factors and adjusting their expression levels. Therefore, it is vital to understand the mechanisms by which natural plant products inhibit ferroptosis and relieve PD symptoms. This review provides a comprehensive look at ferroptosis, its role in PD pathology, and the mechanisms underlying the therapeutic effects of natural plant products focused on ferroptosis. The insights from this review can serve as useful references for future research on novel ferroptosis inhibitors and lead compounds for PD treatment.
Collapse
Affiliation(s)
- Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Academy of Chinese Medicine, Changsha, Hunan, China.
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Deng
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hao Xu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Mengxia Yuan
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, Shantou, China
| | - Yanfang Luo
- The Central Hospital of Shaoyang, Shaoyang, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Academy of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
35
|
Fu D, Liang X, Jiang Y, Liu J, Lin X, Yang Q, Chen X, Huang P, Wang W, Wu W. Iron blocks autophagic flux and induces autophagosomes accumulation in microglia. Food Chem Toxicol 2023; 181:114054. [PMID: 37777083 DOI: 10.1016/j.fct.2023.114054] [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: 06/09/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Iron is an essential dietary micronutrient for maintaining physiological homeostasis. However, disruption of cerebral iron regulation with the accumulation of iron in different brain structures appears to have a role in the pathogenesis of various neurodegenerative disorders. Studies have reported that autophagy induction could potentially mitigate progression in neurodegenerative diseases with iron deposition, but the relationship between autophagy and iron remains poorly understood. Meanwhile, abnormal autophagy in microglia is closely related to the occurrence of neurodegenerative diseases. Therefore, the effect of iron on microglia autophagy needs to be elaborated. In the present study, we found that iron induces autophagosome accumulation but inhibits its initiation in an Akt-mTOR pathway independent manner. Meanwhile, it caused autophagy flux defects and dysfunction of lysosomes. We also found that iron overload reduced the expression of Rab7, which is an essential protein for the fusion of autophagosomes and lysosomes. These results suggest that iron induces the accumulation of autophagosome in microglia and disrupts the autophagic flux in late stage of autophagy. Therefore, our work provides new insights into the molecular mechanisms of iron neurotoxicity.
Collapse
Affiliation(s)
- Deqiang Fu
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xingyue Liang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China; School of Advance Manufacture, Fuzhou University, Jinjiang, 362251, China
| | - Yuxuan Jiang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Jieping Liu
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xiaosi Lin
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Quan Yang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xue Chen
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Ping Huang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Wei Wang
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China.
| | - Wenlin Wu
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China; School of Advance Manufacture, Fuzhou University, Jinjiang, 362251, China.
| |
Collapse
|
36
|
Li Y, Liu SB, Ni W, Gurzadyan GG, Wu Y, Wang J, Kuang GC, Jiang W. Near-Infrared BODIPY Photosensitizer for Modulating Mitochondrial Fusion Proteins and Inhibiting Choroidal Neovascularization. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48027-48037. [PMID: 37812497 DOI: 10.1021/acsami.3c11053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Photosensitizers have emerged as cytotoxic reactive oxygen species (ROS) activators in photodynamic therapy (PDT), which induced cell apoptosis. As the major contributors to ROS and oxidative stress, mitochondria play an important role in cell apoptosis. Although there are many reports about near-infrared 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) as photosensitizers (PSs) for PDT, this kind of PS has rarely been used for treating mitochondrial function and choroidal neovascularization application at the same time. Herein, a novel near-infrared PS (BDP2) characterized by good water solubility, long wavelength excitation, and high ROS quantum yield has been made. Under near-infrared light irradiation, BDP2 would generate ROS with high yield, induce a mitochondrial morphology change, and trigger cell apoptosis by changing the fusion protein level. Deep investigation revealed that BDP2 can cause oxidative stress, break the balance between fusion and fission of mitochondrial dynamics protein through decreasing fusion protein MFN2 and OPA1 expression, and finally cause cell apoptosis. Due to these characteristics, the BDP2 PS was used to treat choroidal neovascularization in animal models and can inhibit neovascularization.
Collapse
Affiliation(s)
- Yue Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, The People's Republic of China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha 410011, Hunan, The People's Republic of China
| | - Shi-Bo Liu
- State Key Laboratory of Powder Metallurgy, Central South University, Lushan South Road 932, Yuelu District, Changsha 410083, Hunan, The People's Republic of China
| | - Wenjun Ni
- Institute of Artificial Photosynthesis, State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, Liaoning, The People's Republic of China
| | - Gagik G Gurzadyan
- Institute of Artificial Photosynthesis, State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, Liaoning, The People's Republic of China
| | - Yongquan Wu
- Key Laboratory of Organo-pharmaceutical Chemistry, School of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou 341000, Jiangxi, The People's Republic of China
| | - Jun Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, The People's Republic of China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha 410011, Hunan, The People's Republic of China
| | - Gui-Chao Kuang
- State Key Laboratory of Powder Metallurgy, Central South University, Lushan South Road 932, Yuelu District, Changsha 410083, Hunan, The People's Republic of China
| | - Wenmin Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, The People's Republic of China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha 410011, Hunan, The People's Republic of China
| |
Collapse
|
37
|
Wang J, Yang J, Liu K, Yuan J, Shi Y, Li H, Zhao L. Tumor targeted cancer membrane-camouflaged ultra-small Fe nanoparticles for enhanced collaborative apoptosis and ferroptosis in glioma. Mater Today Bio 2023; 22:100780. [PMID: 37680585 PMCID: PMC10480784 DOI: 10.1016/j.mtbio.2023.100780] [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: 05/31/2023] [Revised: 07/28/2023] [Accepted: 08/27/2023] [Indexed: 09/09/2023] Open
Abstract
Glioma is recognized as the most common and aggressive primary brain tumor in adults. Owing to the occurrence of drug resistance and the failure of drug to penetrate the blood-brain barrier (BBB), there is no effective strategy for the treatment of glioma. The main objective of this study was to develop a biomimetic glioma C6 cell membrane (C6M) derived nanovesicles (DOX-FN/C6M-NVs) loaded with doxorubicin (DOX) and ultra-small Fe nanoparticles (FN) for accomplishing the effective brain tumor-targeted delivery of DOX and improving anti-cancer efficacy via inducing collaborative apoptosis and ferroptosis. The findings revealed that employing C6M-NVs as a carrier significantly improved the therapeutic efficacy by enabling evasion of immune surveillance, facilitating targeted drug delivery to tumor sites, and minimizing cardiotoxicity and adverse effects associated with DOX. DOX-FN/C6M-NVs exhibited more potent anti-tumor effects as compared with free DOX by promoting DOX-mediated apoptosis and accelerating ferroptosis via the mediation of FN. This study suggested that DOX-FN/C6M-NVs as the potential inducer of ferroptosis and apoptosis conferred effective tumor suppression in the treatment of glioma.
Collapse
Affiliation(s)
- Jingchen Wang
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Jian Yang
- Life Science Institution, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Kang Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Jiayu Yuan
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Hongdan Li
- Life Science Institution, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| |
Collapse
|
38
|
Ghaderi S, Batouli SAH, Mohammadi S, Fatehi F. Iron quantification in basal ganglia using quantitative susceptibility mapping in a patient with ALS: a case report and literature review. Front Neurosci 2023; 17:1229082. [PMID: 37877011 PMCID: PMC10593460 DOI: 10.3389/fnins.2023.1229082] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/04/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Quantitative susceptibility mapping (QSM) is a magnetic resonance imaging (MRI) technique that can measure the magnetic susceptibility of tissues, which can reflect their iron content. QSM has been used to detect iron accumulation in cortical and subcortical brain regions. However, its application in subcortical regions such as the basal ganglia, particularly the putamen, is rare in patients with amyotrophic lateral sclerosis (ALS). CASE PRESENTATION AND LITERATURE REVIEW We present the case of a 40-year-old male patient with ALS who underwent an MRI for QSM. We compared his QSM images with those of a control subject and performed a quantitative analysis of the magnetic susceptibility values in the putamen regions. We also reviewed the literature on previous QSM studies in ALS and summarized their methods and findings. Our QSM analysis revealed increased magnetic susceptibility values in the bilateral putamen of the ALS patient compared to controls, indicating iron overload. This finding is consistent with previous studies reporting iron dysregulation in subcortical nuclei in ALS. We also discussed the QSM processing techniques used in our study and in the literature, highlighting their advantages and limitations. CONCLUSION This case report demonstrates the potential of QSM as a sensitive MRI biomarker for evaluating iron levels in subcortical regions of ALS patients. QSM can provide quantitative information on iron deposition patterns in both motor and extra-motor areas of ALS patients, which may help understand the pathophysiology of ALS and monitor disease progression. Further studies with larger samples are needed to validate these results and explore the clinical implications of QSM in ALS.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Hossein Batouli
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Mohammadi
- Department of Medical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Mukem S, Sayoh I, Maungchanburi S, Thongbuakaew T. Ebselen, Iron Uptake Inhibitor, Alleviates Iron Overload-Induced Senescence-Like Neuronal Cells SH-SY5Y via Suppressing the mTORC1 Signaling Pathway. Adv Pharmacol Pharm Sci 2023; 2023:6641347. [PMID: 37731679 PMCID: PMC10509000 DOI: 10.1155/2023/6641347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/17/2023] [Accepted: 09/02/2023] [Indexed: 09/22/2023] Open
Abstract
Increasing evidence highlights that excessive iron accumulation in the brain plays a vital role in neuronal senescence and is implicated in the pathogenesis of age-related neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Therefore, the chemical compounds that eliminate an iron overload may provide better protection against oxidative stress conditions that cause the accumulation of senescent cells during brain aging. Ebselen has been identified as a strongly useful compound in the research on redox biology mechanisms. We hypothesized that ebselen could alleviate an iron overload-induced oxidative stress and consequently reverses the senescence-like phenotypes in the neuronal cells. In the present study, SH-SY5Y cells were treated with ferric ammonium citrate (FAC) before ebselen, and the evaluation of the cellular iron homeostasis, the indicators of oxidative stress, and the onset of senescence phenotypes and mechanisms were carried out accordingly. Our findings showed that ebselen ameliorated the FAC-mediated iron overload by decreasing the expression of divalent metal transporter 1 (DMT1) and ferritin light chain (FT-L) proteins. In contrast, it increased the expression of ferroportin 1 (FPN1) protein and its correlation led to a decrease in the expression of the cytosolic labile iron pool (LIP). Furthermore, ebselen significantly reduced reactive oxygen species (ROS) and rescued the mitochondrial membrane potential (ΔΨm). Notably, ebselen restored the biomarkers of cellular senescence by reducing the number of senescence-associated β-galactosidase (SA-β-gal) positive cells and senescence-associated secretory phenotypes (SASP). This also suppressed the expression of p53 protein targeting DNA damage response (DDR)/p21 cyclin-dependent kinase (CDK) inhibitor through a mTORC1 signaling pathway. Potentially, ebselen could be a therapeutic agent for treating brain aging and AD by mitigating iron accumulation and restoring senescence in SH-SY5Y cells.
Collapse
Affiliation(s)
- Sirirak Mukem
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Ibrahim Sayoh
- Department of Anatomy, Faculty of Science and Technology, Princess of Naradhiwas University, Narathiwat 96000, Thailand
| | - Saowanee Maungchanburi
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | | |
Collapse
|
40
|
Moos WH, Faller DV, Glavas IP, Kanara I, Kodukula K, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Sampani K, Steliou K, Vavvas DG. Epilepsy: Mitochondrial connections to the 'Sacred' disease. Mitochondrion 2023; 72:84-101. [PMID: 37582467 DOI: 10.1016/j.mito.2023.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/03/2023] [Accepted: 08/12/2023] [Indexed: 08/17/2023]
Abstract
Over 65 million people suffer from recurrent, unprovoked seizures. The lack of validated biomarkers specific for myriad forms of epilepsy makes diagnosis challenging. Diagnosis and monitoring of childhood epilepsy add to the need for non-invasive biomarkers, especially when evaluating antiseizure medications. Although underlying mechanisms of epileptogenesis are not fully understood, evidence for mitochondrial involvement is substantial. Seizures affect 35%-60% of patients diagnosed with mitochondrial diseases. Mitochondrial dysfunction is pathophysiological in various epilepsies, including those of non-mitochondrial origin. Decreased ATP production caused by malfunctioning brain cell mitochondria leads to altered neuronal bioenergetics, metabolism and neurological complications, including seizures. Iron-dependent lipid peroxidation initiates ferroptosis, a cell death pathway that aligns with altered mitochondrial bioenergetics, metabolism and morphology found in neurodegenerative diseases (NDDs). Studies in mouse genetic models with seizure phenotypes where the function of an essential selenoprotein (GPX4) is targeted suggest roles for ferroptosis in epilepsy. GPX4 is pivotal in NDDs, where selenium protects interneurons from ferroptosis. Selenium is an essential central nervous system micronutrient and trace element. Low serum concentrations of selenium and other trace elements and minerals, including iron, are noted in diagnosing childhood epilepsy. Selenium supplements alleviate intractable seizures in children with reduced GPX activity. Copper and cuproptosis, like iron and ferroptosis, link to mitochondria and NDDs. Connecting these mechanistic pathways to selenoproteins provides new insights into treating seizures, pointing to using medicines including prodrugs of lipoic acid to treat epilepsy and to potential alternative therapeutic approaches including transcranial magnetic stimulation (transcranial), photobiomodulation and vagus nerve stimulation.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.
| | - Douglas V Faller
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Ioannis P Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, NY, USA
| | | | | | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Carl A Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Whitney R Powers
- Department of Health Sciences, Boston University, Boston, MA, USA; Department of Anatomy, Boston University School of Medicine, Boston, MA, USA
| | - Konstantina Sampani
- Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA; PhenoMatriX, Inc., Natick, MA, USA
| | - Demetrios G Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| |
Collapse
|
41
|
Martins AC, Virgolini MB, Ávila DS, Scharf P, Li J, Tinkov AA, Skalny AV, Bowman AB, Rocha JBT, Aschner M. Mitochondria in the Spotlight: C. elegans as a Model Organism to Evaluate Xenobiotic-Induced Dysfunction. Cells 2023; 12:2124. [PMID: 37681856 PMCID: PMC10486742 DOI: 10.3390/cells12172124] [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: 07/18/2023] [Revised: 08/19/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023] Open
Abstract
Mitochondria play a crucial role in cellular respiration, ATP production, and the regulation of various cellular processes. Mitochondrial dysfunctions have been directly linked to pathophysiological conditions, making them a significant target of interest in toxicological research. In recent years, there has been a growing need to understand the intricate effects of xenobiotics on human health, necessitating the use of effective scientific research tools. Caenorhabditis elegans (C. elegans), a nonpathogenic nematode, has emerged as a powerful tool for investigating toxic mechanisms and mitochondrial dysfunction. With remarkable genetic homology to mammals, C. elegans has been used in studies to elucidate the impact of contaminants and drugs on mitochondrial function. This review focuses on the effects of several toxic metals and metalloids, drugs of abuse and pesticides on mitochondria, highlighting the utility of C. elegans as a model organism to investigate mitochondrial dysfunction induced by xenobiotics. Mitochondrial structure, function, and dynamics are discussed, emphasizing their essential role in cellular viability and the regulation of processes such as autophagy, apoptosis, and calcium homeostasis. Additionally, specific toxins and toxicants, such as arsenic, cadmium, and manganese are examined in the context of their impact on mitochondrial function and the utility of C. elegans in elucidating the underlying mechanisms. Furthermore, we demonstrate the utilization of C. elegans as an experimental model providing a promising platform for investigating the intricate relationships between xenobiotics and mitochondrial dysfunction. This knowledge could contribute to the development of strategies to mitigate the adverse effects of contaminants and drugs of abuse, ultimately enhancing our understanding of these complex processes and promoting human health.
Collapse
Affiliation(s)
- Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Miriam B. Virgolini
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Daiana Silva Ávila
- Laboratory of Biochemistry and Toxicology in Caenorhabditis Elegans, Universidade Federal do Pampa, Campus Uruguaiana, BR-472 Km 592, Uruguaiana 97500-970, RS, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Jung Li
- College of Osteopathic Medicine, Des Moines University, Des Moines, IA 50312, USA
| | - Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Anatoly V. Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - João B. T. Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| |
Collapse
|
42
|
Zheng Q, Ma P, Yang P, Zhai S, He M, Zhang X, Tu Q, Jiao L, Ye L, Feng Z, Zhang C. Alpha lipoic acid ameliorates motor deficits by inhibiting ferroptosis in Parkinson's disease. Neurosci Lett 2023; 810:137346. [PMID: 37308056 DOI: 10.1016/j.neulet.2023.137346] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/07/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease. Ferroptosis shares several features with PD pathophysiology, and anti-ferroptosis molecules are neuroprotective in PD animal models. As an antioxidant and iron chelating agent, alpha lipoic acid (ALA) has a neuroprotective effect on PD; however, the influence of ALA on ferroptosis in PD remains unclear. This study aimed to determine the mechanism of ALA in regulating ferroptosis in PD models. Results showed that ALA could ameliorate motor deficits in PD models and regulate iron metabolism by upregulating ferroportin (FPN) and ferritin heavy chain 1 (FTH1) and downregulating iron importer divalent metal transporter 1 (DMT1). Moreover, ALA decreased the accumulation of reactive oxygen species (ROS) and lipid peroxidation, rescued mitochondrial damage, and prevented ferroptosis effectively by inhibiting the downregulation of glutathione peroxidase 4 (GPX4) and cysteine/glutamate transporter (xCT) in PD. Mechanistic study indicated that the activation of SIRT1/NRF2 pathway was involved in the upregulation effect of GPX4 and FTH1. Thus, ALA ameliorates motor deficits in PD models by regulating iron metabolism and mitigating ferroptosis through the SIRT1/NRF2 signaling pathway.
Collapse
Affiliation(s)
- Qian Zheng
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China
| | - Pengfei Ma
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China
| | - Pan Yang
- Engineering Research Center for Molecular Medicine, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China
| | - Suzhen Zhai
- Engineering Research Center for Molecular Medicine, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China
| | - Meina He
- Engineering Research Center for Molecular Medicine, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China
| | - Xiangming Zhang
- Engineering Research Center for Molecular Medicine, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China
| | - Qiuxia Tu
- Engineering Research Center for Molecular Medicine, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China
| | - Ling Jiao
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China
| | - Lan Ye
- Engineering Research Center for Molecular Medicine, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China
| | - Zhanhui Feng
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China.
| | - Chunlin Zhang
- Engineering Research Center for Molecular Medicine, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China; Department of Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
43
|
Casanova A, Wevers A, Navarro-Ledesma S, Pruimboom L. Mitochondria: It is all about energy. Front Physiol 2023; 14:1114231. [PMID: 37179826 PMCID: PMC10167337 DOI: 10.3389/fphys.2023.1114231] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/29/2023] [Indexed: 05/15/2023] Open
Abstract
Mitochondria play a key role in both health and disease. Their function is not limited to energy production but serves multiple mechanisms varying from iron and calcium homeostasis to the production of hormones and neurotransmitters, such as melatonin. They enable and influence communication at all physical levels through interaction with other organelles, the nucleus, and the outside environment. The literature suggests crosstalk mechanisms between mitochondria and circadian clocks, the gut microbiota, and the immune system. They might even be the hub supporting and integrating activity across all these domains. Hence, they might be the (missing) link in both health and disease. Mitochondrial dysfunction is related to metabolic syndrome, neuronal diseases, cancer, cardiovascular and infectious diseases, and inflammatory disorders. In this regard, diseases such as cancer, Alzheimer's, Parkinson's, amyotrophic lateral sclerosis (ALS), chronic fatigue syndrome (CFS), and chronic pain are discussed. This review focuses on understanding the mitochondrial mechanisms of action that allow for the maintenance of mitochondrial health and the pathways toward dysregulated mechanisms. Although mitochondria have allowed us to adapt to changes over the course of evolution, in turn, evolution has shaped mitochondria. Each evolution-based intervention influences mitochondria in its own way. The use of physiological stress triggers tolerance to the stressor, achieving adaptability and resistance. This review describes strategies that could recover mitochondrial functioning in multiple diseases, providing a comprehensive, root-cause-focused, integrative approach to recovering health and treating people suffering from chronic diseases.
Collapse
Affiliation(s)
- Amaloha Casanova
- Department of Physiotherapy, University of Granada, Granada, Spain
- Faculty of Health Sciences, Melilla, Spain
- PNI Europe, The Hague, Netherlands
- Chair of Clinical Psychoneuroimmunology, University of Granada and PNI Europe, Granada, Spain
| | - Anne Wevers
- Department of Physiotherapy, University of Granada, Granada, Spain
- Faculty of Health Sciences, Melilla, Spain
- PNI Europe, The Hague, Netherlands
- Chair of Clinical Psychoneuroimmunology, University of Granada and PNI Europe, Granada, Spain
| | - Santiago Navarro-Ledesma
- Department of Physiotherapy, University of Granada, Granada, Spain
- Faculty of Health Sciences, Melilla, Spain
- PNI Europe, The Hague, Netherlands
- Chair of Clinical Psychoneuroimmunology, University of Granada and PNI Europe, Granada, Spain
| | - Leo Pruimboom
- PNI Europe, The Hague, Netherlands
- Chair of Clinical Psychoneuroimmunology, University of Granada and PNI Europe, Granada, Spain
| |
Collapse
|
44
|
Exploring Whether Iron Sequestration within the CNS of Patients with Alzheimer’s Disease Causes a Functional Iron Deficiency That Advances Neurodegeneration. Brain Sci 2023; 13:brainsci13030511. [PMID: 36979320 PMCID: PMC10046656 DOI: 10.3390/brainsci13030511] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
The involvement of iron in the pathogenesis of Alzheimer’s disease (AD) may be multifaceted. Besides potentially inducing oxidative damage, the bioavailability of iron may be limited within the central nervous system, creating a functionally iron-deficient state. By comparing staining results from baseline and modified iron histochemical protocols, iron was found to be more tightly bound within cortical sections from patients with high levels of AD pathology compared to subjects with a diagnosis of something other than AD. To begin examining whether the bound iron could cause a functional iron deficiency, a protein-coding gene expression dataset of initial, middle, and advanced stages of AD from olfactory bulb tissue was analyzed for iron-related processes with an emphasis on anemia-related changes in initial AD to capture early pathogenic events. Indeed, anemia-related processes had statistically significant alterations, and the significance of these changes exceeded those for AD-related processes. Other changes in patients with initial AD included the expressions of transcripts with iron-responsive elements and for genes encoding proteins for iron transport and mitochondrial-related processes. In the latter category, there was a decreased expression for the gene encoding pitrilysin metallopeptidase 1 (PITRM1). Other studies have shown that PITRM1 has an altered activity in patients with AD and is associated with pathological changes in this disease. Analysis of a gene expression dataset from PITRM1-deficient or sufficient organoids also revealed statistically significant changes in anemia-like processes. These findings, together with supporting evidence from the literature, raise the possibility that a pathogenic mechanism of AD could be a functional deficiency of iron contributing to neurodegeneration.
Collapse
|
45
|
Sarkar A, Rasheed MSU, Singh MP. Redox Modulation of Mitochondrial Proteins in the Neurotoxicant Models of Parkinson's Disease. Antioxid Redox Signal 2023; 38:824-852. [PMID: 36401516 DOI: 10.1089/ars.2022.0106] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Significance: Mitochondrial proteins regulate the oxidative phosphorylation, cellular metabolism, and free radical generation. Redox modulation alters the mitochondrial proteins and instigates the damage to dopaminergic neurons. Toxicants contribute to Parkinson's disease (PD) pathogenesis in conjunction with aging and genetic factors. While oxidative modulation of a number of mitochondrial proteins is linked to xenobiotic exposure, little is known about its role in the toxicant-induced PD. Understanding the role of redox modulation of mitochondrial proteins in complex cellular events leading to neurodegeneration is highly relevant. Recent Advances: Many toxicants are shown to inhibit complex I or III and elicit free radical production that alters the redox status of mitochondrial proteins. Implication of redox modulation of the mitochondrial proteins makes them a target to comprehend the underlying mechanism of toxicant-induced PD. Critical Issues: Owing to multifactorial etiology, exploration of onset and progression and treatment outcomes needs a comprehensive approach. The article explains about a few mitochondrial proteins that undergo redox changes along with the promising strategies, which help to alleviate the toxicant-induced redox imbalance leading to neurodegeneration. Future Directions: Although mitochondrial proteins are linked to PD, their role in toxicant-induced parkinsonism is not yet completely known. Preservation of antioxidant defense machinery could alleviate the redox modulation of mitochondrial proteins. Targeted antioxidant delivery, use of metal chelators, and activation of nuclear factor erythroid 2-related factor 2, and combinational therapy that encounters multiple free radicals, could ameliorate the redox modulation of mitochondrial proteins and thereby PD progression.
Collapse
Affiliation(s)
- Alika Sarkar
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Mohd Sami Ur Rasheed
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Mahendra Pratap Singh
- Toxicogenomics and Predictive Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
46
|
Fukuhara K, Nakanishi I, Imai K, Mizuno M, Matsumoto KI, Ohno A. DTPA-Bound Planar Catechin with Potent Antioxidant Activity Triggered by Fe 3+ Coordination. Antioxidants (Basel) 2023; 12:antiox12020225. [PMID: 36829782 PMCID: PMC9952317 DOI: 10.3390/antiox12020225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
In diseases related to oxidative stress, accumulation of metal ions at the site of pathogenesis results in the generation of reactive oxygen species (ROS) through the reductive activation of oxygen molecules catalyzed by the metal ions. If these metals can be removed and the generated ROS can be strongly scavenged, such diseases can be prevented and treated. Planar catechins exhibit stronger radical scavenging activity than natural catechins and can efficiently scavenge hydroxyl radicals generated by the Fenton reaction without showing pro-oxidant effects, even in the presence of iron ions. Hence, in the current study, we designed a compound in which diethylenetriaminepentaacetic acid (DTPA), a metal chelator, was bound to a planar catechin with enhanced radical scavenging activity by immobilizing the steric structure of a natural catechin to be planar. This compound showed almost no radical scavenging activity due to intramolecular hydrogen bonding of DTPA with the planar catechins; however, when coordinated with Fe3+, it showed more potent radical scavenging activity than planar catechins. Owing to its potent antioxidant activity triggered by metal coordination and its inhibition of ROS generation by trapping metal ions, this compound might exert excellent preventive and therapeutic effects against oxidative stress-related diseases.
Collapse
Affiliation(s)
- Kiyoshi Fukuhara
- Division of Organic and Medicinal Chemistry, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo 142-8555, Japan
- Correspondence:
| | - Ikuo Nakanishi
- Quantum RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Inage-ku, Chiba 263-8555, Japan
| | - Kohei Imai
- Division of Organic and Medicinal Chemistry, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Mirei Mizuno
- Division of Organic and Medicinal Chemistry, School of Pharmacy, Showa University, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Ken-ichiro Matsumoto
- Quantitative RedOx Sensing Group, Department of Radiation Regulatory Science Research, National Institute of Radiological Sciences (NIRS), Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Inage-ku, Chiba 263-8555, Japan
| | - Akiko Ohno
- Division of Risk Assessment, Center for Biological Safety & Research, National Institute of Health Sciences, Kawasaki-ku, Kawasaki, Kanagawa, Yokohama 210-9501, Japan
| |
Collapse
|
47
|
Gérard C, Archambault AF, Bouchard C, Tremblay JP. A promising mouse model for Friedreich Ataxia progressing like human patients. Behav Brain Res 2023; 436:114107. [DOI: 10.1016/j.bbr.2022.114107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/27/2022]
|
48
|
Duan G, Li J, Duan Y, Zheng C, Guo Q, Li F, Zheng J, Yu J, Zhang P, Wan M, Long C. Mitochondrial Iron Metabolism: The Crucial Actors in Diseases. Molecules 2022; 28:29. [PMID: 36615225 PMCID: PMC9822237 DOI: 10.3390/molecules28010029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Iron is a trace element necessary for cell growth, development, and cellular homeostasis, but insufficient or excessive level of iron is toxic. Intracellularly, sufficient amounts of iron are required for mitochondria (the center of iron utilization) to maintain their normal physiologic function. Iron deficiency impairs mitochondrial metabolism and respiratory activity, while mitochondrial iron overload promotes ROS production during mitochondrial electron transport, thus promoting potential disease development. This review provides an overview of iron homeostasis, mitochondrial iron metabolism, and how mitochondrial iron imbalances-induced mitochondrial dysfunction contribute to diseases.
Collapse
Affiliation(s)
- Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianjun Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changbing Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengliao Wan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Cimin Long
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
49
|
Ferroptosis is involved in regulating perioperative neurocognitive disorders: emerging perspectives. J Neuroinflammation 2022; 19:219. [PMID: 36068571 PMCID: PMC9450301 DOI: 10.1186/s12974-022-02570-3] [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] [Received: 06/09/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
Since the twenty-first century, the development of technological advances in anesthesia and surgery has brought benefits to human health. However, the adverse neurological effects of perioperative-related factors (e.g., surgical trauma, anesthesia, etc.) as stressors cannot be ignored as well. The nervous system appears to be more "fragile" and vulnerable to damage in developing and aging individuals. Ferroptosis is a novel form of programmed cell death proposed in 2012. In recent years, the regulation of ferroptosis to treat cancer, immune system disorders, and neurodegenerative diseases have seen an unprecedented surge of interest. The association of ferroptosis with perioperative neurocognitive disorders has also received much attention. Cognitive impairment can not only affect the individual's quality of life, but also impose a burden on the family and society. Therefore, the search for effective preventive and therapeutic methods to alleviate cognitive impairment caused by perioperative-related factors is a challenge that needs to be urgently addressed. In our review, we first briefly describe the connection between iron accumulation in neurons and impairment of brain function during development and aging. It is followed by a review of the pathways of ferroptosis, mainly including iron metabolism, amino acid metabolism, and lipid metabolism pathway. Furthermore, we analyze the connection between ferroptosis and perioperative-related factors. The surgery itself, general anesthetic drugs, and many other relevant factors in the perioperative period may affect neuronal iron homeostasis. Finally, we summarize the experimental evidence for ameliorating developmental and degenerative neurotoxicity by modulating ferroptosis. The suppression of ferroptosis seems to provide the possibility to prevent and improve perioperative neurocognitive impairment.
Collapse
|
50
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kamperi N, Kanara I, Kodukula K, Mavrakis AN, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Sampani K, Steliou K, Tamvakopoulos C, Vavvas DG, Zamboni RJ, Chen X. Treatment and prevention of pathological mitochondrial dysfunction in retinal degeneration and in photoreceptor injury. Biochem Pharmacol 2022; 203:115168. [PMID: 35835206 DOI: 10.1016/j.bcp.2022.115168] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022]
Abstract
Pathological deterioration of mitochondrial function is increasingly linked with multiple degenerative illnesses as a mediator of a wide range of neurologic and age-related chronic diseases, including those of genetic origin. Several of these diseases are rare, typically defined in the United States as an illness affecting fewer than 200,000 people in the U.S. population, or about one in 1600 individuals. Vision impairment due to mitochondrial dysfunction in the eye is a prominent feature evident in numerous primary mitochondrial diseases and is common to the pathophysiology of many of the familiar ophthalmic disorders, including age-related macular degeneration, diabetic retinopathy, glaucoma and retinopathy of prematurity - a collection of syndromes, diseases and disorders with significant unmet medical needs. Focusing on metabolic mitochondrial pathway mechanisms, including the possible roles of cuproptosis and ferroptosis in retinal mitochondrial dysfunction, we shed light on the potential of α-lipoyl-L-carnitine in treating eye diseases. α-Lipoyl-L-carnitine is a bioavailable mitochondria-targeting lipoic acid prodrug that has shown potential in protecting against retinal degeneration and photoreceptor cell loss in ophthalmic indications.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.
| | - Douglas V Faller
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Ioannis P Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, NY, USA
| | - David N Harpp
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Natalia Kamperi
- Center for Clinical, Experimental Surgery and Translational Research Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | | | - Anastasios N Mavrakis
- Department of Medicine, Tufts University School of Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, MA, USA
| | - Carl A Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Whitney R Powers
- Department of Health Sciences, Boston University, Boston, MA, USA; Department of Anatomy, Boston University School of Medicine, Boston, MA, USA
| | - Konstantina Sampani
- Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA; PhenoMatriX, Inc., Natick, MA, USA
| | - Constantin Tamvakopoulos
- Center for Clinical, Experimental Surgery and Translational Research Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Demetrios G Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Robert J Zamboni
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Xiaohong Chen
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, USA; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China.
| |
Collapse
|