1
|
Chen J, Yan L, Zhang Y, Liu X, Wei Y, Zhao Y, Li K, Shi Y, Liu H, Lai W, Tian L, Lin B. Maternal exposure to nanopolystyrene induces neurotoxicity in offspring through P53-mediated ferritinophagy and ferroptosis in the rat hippocampus. J Nanobiotechnology 2024; 22:651. [PMID: 39438901 PMCID: PMC11520165 DOI: 10.1186/s12951-024-02911-9] [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: 06/11/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
There are increasing concerns regarding the rapid expansion of polystyrene nanoplastics (PS-NPs), which could impact human health. Previous studies have shown that nanoplastics can be transferred from mothers to offspring through the placenta and breast milk, resulting in cognitive deficits in offspring. However, the neurotoxic effects of maternal exposure on offspring and its mechanisms remain unclear. In this study, PS-NPs (50 nm) were gavaged to female rats throughout gestation and lactation to establish an offspring exposure model to study the neurotoxicity and behavioral changes caused by PS-NPs on offspring. Neonatal rat hippocampal neuronal cells were used to investigate the pathways through which NPs induce neurodevelopmental toxicity in offspring rats, using iron inhibitors, autophagy inhibitors, reactive oxygen species (ROS) scroungers, P53 inhibitors, and NCOA4 inhibitors. We found that low PS-NPs dosages can cause ferroptosis in the hippocampus of the offspring, resulting in a decline in the cognitive, learning, and memory abilities of the offspring. PS-NPs induced NOCA4-mediated ferritinophagy and promoted ferroptosis by inciting ROS production to activate P53-mediated ferritinophagy. Furthermore, the levels of the antioxidant factors glutathione peroxidase 4 (GPX4) and glutathione (GSH), responsible for ferroptosis, were reduced. In summary, this study revealed that consumption of PS-NPs during gestation and lactation can cause ferroptosis and damage the hippocampus of offspring. Our results can serve as a basis for further research into the neurodevelopmental effects of nanoplastics in offspring.
Collapse
Affiliation(s)
- Jiang Chen
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Licheng Yan
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Yaping Zhang
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Xuan Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yizhe Wei
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yiming Zhao
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Kang Li
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yue Shi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Huanliang Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Wenqing Lai
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Lei Tian
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| | - Bencheng Lin
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| |
Collapse
|
2
|
Kim JB, Hyung H, Bae JE, Jang S, Park NY, Jo DS, Kim YH, Choi DK, Ryu HY, Lee HS, Ryoo ZY, Cho DH. Increased ER stress by depletion of PDIA6 impairs primary ciliogenesis and enhances sensitivity to ferroptosis in kidney cells. BMB Rep 2024; 57:453-458. [PMID: 39044457 PMCID: PMC11524824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/16/2024] [Accepted: 03/18/2024] [Indexed: 07/25/2024] Open
Abstract
Primary cilia are crucial for cellular balance, serving as sensors for external conditions. Nephronophthisis and related ciliopathies, which are hereditary and degenerative, stem from genetic mutations in cilia-related genes. However, the precise mechanisms of these conditions are still not fully understood. Our research demonstrates that downregulating PDIA6, leading to cilia removal, makes cells more sensitive to ferroptotic death caused by endoplasmic reticulum (ER) stress. The reduction of PDIA6 intensifies the ER stress response, while also impairing the regulation of primary cilia in various cell types. PDIA6 loss worsens ER stress, hastening ferroptotic death in proximal tubule epithelial cells, HK2 cells. Counteracting this ER stress can mitigate PDIA6 depletion effects, restoring both the number and length of cilia. Moreover, preventing ferroptosis corrects the disrupted primary ciliogenesis due to PDIA6 depletion in HK2 cells. Our findings emphasize the role of PDIA6 in primary ciliogenesis, and suggest its absence enhances ER stress and ferroptosis. These insights offer new therapeutic avenues for treating nephronophthisis and similar ciliopathies. [BMB Reports 2024; 57(10): 453-458].
Collapse
Affiliation(s)
- Joon Bum Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Hyejin Hyung
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Ji-Eun Bae
- KNU LAMP Research Center, KNU Institute of Basic Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Soyoung Jang
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Na Yeon Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | | | - Yong Hwan Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Dong Kyu Choi
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Hong-Yeoul Ryu
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- KNU LAMP Research Center, KNU Institute of Basic Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- KNU LAMP Research Center, KNU Institute of Basic Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Zae Young Ryoo
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Dong-Hyung Cho
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- ORGASIS Corp., Suwon 16229, Korea
- Organelle Institute, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
3
|
Miller MR, Landis HE, Miller RE, Tizabi Y. Intercellular Adhesion Molecule 1 (ICAM-1): An Inflammatory Regulator with Potential Implications in Ferroptosis and Parkinson's Disease. Cells 2024; 13:1554. [PMID: 39329738 PMCID: PMC11430830 DOI: 10.3390/cells13181554] [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: 08/09/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Intercellular adhesion molecule 1 (ICAM-1/CD54), a transmembrane glycoprotein, has been considered as one of the most important adhesion molecules during leukocyte recruitment. It is encoded by the ICAM1 gene and plays a central role in inflammation. Its crucial role in many inflammatory diseases such as ulcerative colitis and rheumatoid arthritis are well established. Given that neuroinflammation, underscored by microglial activation, is a key element in neurodegenerative diseases such as Parkinson's disease (PD), we investigated whether ICAM-1 has a role in this progressive neurological condition and, if so, to elucidate the underpinning mechanisms. Specifically, we were interested in the potential interaction between ICAM-1, glial cells, and ferroptosis, an iron-dependent form of cell death that has recently been implicated in PD. We conclude that there exist direct and indirect (via glial cells and T cells) influences of ICAM-1 on ferroptosis and that further elucidation of these interactions can suggest novel intervention for this devastating disease.
Collapse
Affiliation(s)
| | - Harold E. Landis
- Integrative Medicine Fellow, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | | | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| |
Collapse
|
4
|
Khan S, Bano N, Ahamad S, John U, Dar NJ, Bhat SA. Excitotoxicity, Oxytosis/Ferroptosis, and Neurodegeneration: Emerging Insights into Mitochondrial Mechanisms. Aging Dis 2024:AD.2024.0125-1. [PMID: 39122453 DOI: 10.14336/ad.2024.0125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Mitochondrial dysfunction plays a pivotal role in the development of age-related diseases, particularly neurodegenerative disorders. The etiology of mitochondrial dysfunction involves a multitude of factors that remain elusive. This review centers on elucidating the role(s) of excitotoxicity, oxytosis/ferroptosis and neurodegeneration within the context of mitochondrial bioenergetics, biogenesis, mitophagy and oxidative stress and explores their intricate interplay in the pathogenesis of neurodegenerative diseases. The effective coordination of mitochondrial turnover processes, notably mitophagy and biogenesis, is assumed to be critically important for cellular resilience and longevity. However, the age-associated decrease in mitophagy impedes the elimination of dysfunctional mitochondria, consequently impairing mitochondrial biogenesis. This deleterious cascade results in the accumulation of damaged mitochondria and deterioration of cellular functions. Both excitotoxicity and oxytosis/ferroptosis have been demonstrated to contribute significantly to the pathophysiology of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS) and Multiple Sclerosis (MS). Excitotoxicity, characterized by excessive glutamate signaling, initiates a cascade of events involving calcium dysregulation, energy depletion, and oxidative stress and is intricately linked to mitochondrial dysfunction. Furthermore, emerging concepts surrounding oxytosis/ferroptosis underscore the importance of iron-dependent lipid peroxidation and mitochondrial engagement in the pathogenesis of neurodegeneration. This review not only discusses the individual contributions of excitotoxicity and ferroptosis but also emphasizes their convergence with mitochondrial dysfunction, a key driver of neurodegenerative diseases. Understanding the intricate crosstalk between excitotoxicity, oxytosis/ferroptosis, and mitochondrial dysfunction holds potential to pave the way for mitochondrion-targeted therapeutic strategies. Such strategies, with a focus on bioenergetics, biogenesis, mitophagy, and oxidative stress, emerge as promising avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh-202002, India
| | - Urmilla John
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India; School of Studies in Zoology, Jiwaji University, Gwalior, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | | |
Collapse
|
5
|
Xiao L, Wen H, Peng S, Chen B, Tang B, Liu B. Polygonatum polysaccharide ameliorates D-galactose-induced cognitive dysfunction in aging rats by inhibiting ferroptosis through activation of Nrf2. Neurosci Lett 2024; 836:137873. [PMID: 38871020 DOI: 10.1016/j.neulet.2024.137873] [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: 01/04/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
CONTEXT Aging is a major risk factor for various neurodegenerative diseases, and ferroptosis has been identified as an important mode of cell death during accelerated aging. As the main component of the edible plant YuZhu in China, Polygonatum polysaccharide (POP) is an important natural compound with anti-aging properties. OBJECTIVE To evaluate the anti-aging effects of POP and the underlying molecular mechanisms involved and to evaluate the overall anti-aging effects of POP on cognitive impairment due to accelerated aging. MATERIALS AND METHODS A D-galactose (D-gal)-induced accelerated aging rat model was established to evaluate the anti-aging effects of POP and the underlying molecular mechanisms involved. In turn, Morris water maze and open field experiments were used to evaluate the anti-aging effects of POP on cognitive impairment due to accelerated aging. RESULTS The mechanism by which POP affects nuclear factor E2-related factor 2 (Nrf2), an essential transcription factor, was confirmed. POP significantly improved d-gal-induced cognitive dysfunction in treated model rats, which exhibited reduced pathological changes in the hippocampus, reduced latency of the water maze platform, and increased exploration time in the central area in the open field experiment compared to those of untreated model rats. Furthermore, POP intervention downregulated ferroptosis-related proteins and upregulated Nrf2 expression, and selective inhibition of Nrf2 eliminated the ability of POP to reduce ferroptosis. CONCLUSIONS POP is a natural ingredient with therapeutic potential due to its ability to alleviate aging by activating Nrf2, inhibiting ferroptosis, and alleviating cognitive dysfunction.
Collapse
Affiliation(s)
- Lan Xiao
- School of Pharmacy, Hunan University of Traditional Chinese Medicine, ChangSha, China
| | - Huiqiao Wen
- XiangXing College, Hunan University of Traditional Chinese Medicine, ChangSha, China
| | - Sha Peng
- School of Pharmacy, Hunan University of Traditional Chinese Medicine, ChangSha, China
| | - Bowei Chen
- Hunan Academy of Chinese Medicine, Changsha, China
| | - Biao Tang
- School of Medicine, Hunan University of Traditional Chinese Medicine, ChangSha, China
| | - Baiyan Liu
- Hunan Academy of Chinese Medicine, Changsha, China
| |
Collapse
|
6
|
Yan Y, Huang W, Lu X, Chen X, Shan Y, Luo X, Li Y, Yang X, Li C. Zinc oxide nanoparticles induces cell death and consequently leading to incomplete neural tube closure through oxidative stress during embryogenesis. Cell Biol Toxicol 2024; 40:51. [PMID: 38958792 PMCID: PMC11222284 DOI: 10.1007/s10565-024-09894-1] [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: 01/29/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
The implementation of Zinc oxide nanoparticles (ZnO NPs) raises concerns regarding their potential toxic effects on human health. Although more and more researches have confirmed the toxic effects of ZnO NPs, limited attention has been given to their impact on the early embryonic nervous system. This study aimed to explore the impact of exposure to ZnO NPs on early neurogenesis and explore its underlying mechanisms. We conducted experiments here to confirm the hypothesis that exposure to ZnO NPs causes neural tube defects in early embryonic development. We first used mouse and chicken embryos to confirm that ZnO NPs and the Zn2+ they release are able to penetrate the placental barrier, influence fetal growth and result in incomplete neural tube closure. Using SH-SY5Y cells, we determined that ZnO NPs-induced incomplete neural tube closure was caused by activation of various cell death modes, including ferroptosis, apoptosis and autophagy. Moreover, dissolved Zn2+ played a role in triggering widespread cell death. ZnO NPs were accumulated within mitochondria after entering cells, damaging mitochondrial function and resulting in the over production of reactive oxygen species, ultimately inducing cellular oxidative stress. The N-acetylcysteine (NAC) exhibits significant efficacy in mitigating cellular oxidative stress, thereby alleviating the cytotoxicity and neurotoxicity brought about by ZnO NPs. These findings indicated that the exposure of ZnO NPs in early embryonic development can induce cell death through oxidative stress, resulting in a reduced number of cells involved in early neural tube closure and ultimately resulting in incomplete neural tube closure during embryo development. The findings of this study could raise public awareness regarding the potential risks associated with the exposure and use of ZnO NPs in early pregnancy.
Collapse
Affiliation(s)
- Yu Yan
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wenyi Huang
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Research Center of Integrative Medicine, School Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoting Lu
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Research Center of Integrative Medicine, School Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianxian Chen
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Research Center of Integrative Medicine, School Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingyi Shan
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center of Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Xin Luo
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Yu Li
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Xuesong Yang
- Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, China.
- Clinical Research Center, Clifford Hospital, Guangzhou, 511495, China.
| | - Chun Li
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
7
|
Fikry H, Saleh LA, Mahmoud FA, Gawad SA, Abd-Alkhalek HA. CoQ10 targeted hippocampal ferroptosis in a status epilepticus rat model. Cell Tissue Res 2024; 396:371-397. [PMID: 38499882 PMCID: PMC11144258 DOI: 10.1007/s00441-024-03880-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2024] [Indexed: 03/20/2024]
Abstract
Status epilepticus (SE), the most severe form of epilepsy, leads to brain damage. Uncertainty persists about the mechanisms that lead to the pathophysiology of epilepsy and the death of neurons. Overloading of intracellular iron ions has recently been identified as the cause of a newly recognized form of controlled cell death called ferroptosis. Inhibiting ferroptosis has shown promise as a treatment for epilepsy, according to recent studies. So, the current study aimed to assess the possible antiepileptic impact of CoQ10 either alone or with the standard antiepileptic drug sodium valproate (SVP) and to evaluate the targeted effect of COQ10 on hippocampal oxidative stress and ferroptosis in a SE rat model. Using a lithium-pilocarpine rat model of epilepsy, we evaluated the effect of SVP, CoQ10, or both on seizure severity, histological, and immunohistochemical of the hippocampus. Furthermore, due to the essential role of oxidative stress and lipid peroxidation in inducing ferroptosis, we evaluated malonaldehyde (MDA), reduced glutathione (GSH), glutathione peroxidase 4 (GPX4), and ferritin in tissue homogenate. Our work illustrated that ferroptosis occurs in murine models of lithium-pilocarpine-induced seizures (epileptic group). Nissl staining revealed significant neurodegeneration. A significant increase in the number of astrocytes stained with an astrocyte-specific marker was observed in the hippocampus. Effective seizure relief can be achieved in the seizure model by administering CoQ10 alone compared to SVP. This was accomplished by lowering ferritin levels and increasing GPX4, reducing MDA, and increasing GSH in the hippocampus tissue homogenate. In addition, the benefits of SVP therapy for regulating iron stores, GPX4, and oxidative stress markers were amplified by incorporating CoQ10 as compared to SVP alone. It was concluded that CoQ10 alone has a more beneficial effect than SVP alone in restoring histological structures and has a targeted effect on hippocampal oxidative stress and ferroptosis. In addition, COQ10 could be useful as an adjuvant to SVP in protecting against oxidative damage and ferroptosis-related damage that result from epileptic seizures.
Collapse
Affiliation(s)
- Heba Fikry
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon st, Abbasiya sq., Cairo, 11566, Egypt.
| | - Lobna A Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon st, Abbasiya sq., Cairo, 11566, Egypt
| | - Faten A Mahmoud
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon st, Abbasiya sq., Cairo, 11566, Egypt
| | - Sara Abdel Gawad
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon st, Abbasiya sq., Cairo, 11566, Egypt
| | - Hadwa Ali Abd-Alkhalek
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Khalifa El-Maamon st, Abbasiya sq., Cairo, 11566, Egypt
| |
Collapse
|
8
|
Zeng L, Liu Y, Wang Q, Wan H, Meng X, Tu P, Chen H, Luo A, Hu P, Ding X. Botulinum toxin A attenuates osteoarthritis development via inhibiting chondrocyte ferroptosis through SLC7Al1/GPX4 axis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167215. [PMID: 38714267 DOI: 10.1016/j.bbadis.2024.167215] [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: 10/17/2023] [Revised: 04/07/2024] [Accepted: 04/30/2024] [Indexed: 05/09/2024]
Abstract
Osteoarthritis (OA) is a prevalent joint degenerative disease, resulting in a significant societal burden. However, there is currently a lack of effective treatment option available. Previous studies have suggested that Botulinum toxin A (BONT/A), a macromolecular protein extracted from Clostridium Botulinum, may improve the pain and joint function in OA patients, but the mechanism remains elusive. This study was to investigate the impact and potential mechanism of BONT/A on OA in vivo and in vitro experiment. LPS increased the levels of ROS, Fe2+and Fe3+, as well as decreased GSH levels, the ratio of GSH / GSSH and mitochondrial membrane potential. It also enhanced the degeneration of extracellular matrix (ECM) and altered the ferroptosis-related protein expression in chondrocytes. BONT/A rescued LPS-induced decrease in collagen type II (Collagen II) expression and increase in matrix metalloproteinase 13 (MMP13), mitigated LPS-induced cytotoxicity in chondrocytes, abolished the accumulation of ROS and iron, upregulated GSH and the ratio of GSH/ GSSH, improved mitochondrial function, and promoted SLC7A11/GPX4 anti-ferroptosis system activation. Additionally, intra-articular injection of BONT/A inhibited the degradation of cartilage in OA model rats. This chondroprotective effect of BONT/A was reversed by erastin (a classical ferroptosis agonist) and enhanced by liproxstatin-1 (a classic ferroptosis inhibitor). Our research confirms that BONT/A alleviates the OA development by inhibiting the ferroptosis of chondrocytes, which revealed to be a potential therapeutic mechanism for BONT/A treating the OA.
Collapse
Affiliation(s)
- Lian Zeng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanping Liu
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Qingsong Wang
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Hongmei Wan
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Xiran Meng
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Panwen Tu
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Huaxian Chen
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - PengChao Hu
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China.
| | - Xudong Ding
- Department of Rehabilitation Medicine, Clinical Medical Center for Rehabilitation Treatment of Dystonia Disease, Hubei Provincial Clinical Research Center for Parkinson's Disease, Central Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 44100, China.
| |
Collapse
|
9
|
Xu P, Xing H, Ma Y, Ding X, Li T, Zhang Y, Liu L, Ma J, Niu Q. Fluoride Induces Neurocytotoxicity by Disrupting Lysosomal Iron Metabolism and Membrane Permeability. Biol Trace Elem Res 2024:10.1007/s12011-024-04226-0. [PMID: 38760610 DOI: 10.1007/s12011-024-04226-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/06/2024] [Indexed: 05/19/2024]
Abstract
The detrimental effects of fluoride on neurotoxicity have been widely recorded, yet the detailed mechanisms underlying these effects remain unclear. This study explores lysosomal iron metabolism in fluoride-related neurotoxicity, with a focus on the Steap3/TRPML1 axis. Utilizing sodium fluoride (NaF)-treated human neuroblastoma (SH-SY5Y) and mouse hippocampal neuron (HT22) cell lines, our research demonstrates that NaF enhances the accumulation of ferrous ions (Fe2+) in these cells, disrupting lysosomal iron metabolism through the Steap3/TRPML1 axis. Notably, NaF exposure upregulated ACSL4 and downregulated GPX4, accompanied by reduced glutathione (GSH) levels and superoxide dismutase (SOD) activity and increased malondialdehyde (MDA) levels. These changes indicate increased vulnerability to ferroptosis within neuronal cells. The iron chelator deferoxamine (DFO) mitigates this disruption. DFO binds to lysosomal Fe2+ and inhibits the Steap3/TRPML1 axis, restoring normal lysosomal iron metabolism, preventing lysosomal membrane permeabilization (LMP), and reducing neuronal cell ferroptosis. Our findings suggest that interference in lysosomal iron metabolism may mitigate fluoride-induced neurotoxicity, underscoring the critical role of the Steap3/TRPML1 axis in this pathological process.
Collapse
Affiliation(s)
- Panpan Xu
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Hengrui Xing
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yue Ma
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Xueman Ding
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Tingting Li
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yue Zhang
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Li Liu
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jiaolong Ma
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China.
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| | - Qiang Niu
- Department of Preventive Medicine, School of Medicine, Shihezi University, North 2nd Road, Shihezi, Xinjiang, 832000, People's Republic of China.
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, Shihezi, Xinjiang, People's Republic of China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, People's Republic of China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence DiseasesFirst Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| |
Collapse
|
10
|
Mann V, Sundaresan A, Shishodia S. Overnutrition and Lipotoxicity: Impaired Efferocytosis and Chronic Inflammation as Precursors to Multifaceted Disease Pathogenesis. BIOLOGY 2024; 13:241. [PMID: 38666853 PMCID: PMC11048223 DOI: 10.3390/biology13040241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/25/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Overnutrition, driven by the consumption of high-fat, high-sugar diets, has reached epidemic proportions and poses a significant global health challenge. Prolonged overnutrition leads to the deposition of excessive lipids in adipose and non-adipose tissues, a condition known as lipotoxicity. The intricate interplay between overnutrition-induced lipotoxicity and the immune system plays a pivotal role in the pathogenesis of various diseases. This review aims to elucidate the consequences of impaired efferocytosis, caused by lipotoxicity-poisoned macrophages, leading to chronic inflammation and the subsequent development of severe infectious diseases, autoimmunity, and cancer, as well as chronic pulmonary and cardiovascular diseases. Chronic overnutrition promotes adipose tissue expansion which induces cellular stress and inflammatory responses, contributing to insulin resistance, dyslipidemia, and metabolic syndrome. Moreover, sustained exposure to lipotoxicity impairs the efferocytic capacity of macrophages, compromising their ability to efficiently engulf and remove dead cells. The unresolved chronic inflammation perpetuates a pro-inflammatory microenvironment, exacerbating tissue damage and promoting the development of various diseases. The interaction between overnutrition, lipotoxicity, and impaired efferocytosis highlights a critical pathway through which chronic inflammation emerges, facilitating the development of severe infectious diseases, autoimmunity, cancer, and chronic pulmonary and cardiovascular diseases. Understanding these intricate connections sheds light on potential therapeutic avenues to mitigate the detrimental effects of overnutrition and lipotoxicity on immune function and tissue homeostasis, thereby paving the way for novel interventions aimed at reducing the burden of these multifaceted diseases on global health.
Collapse
Affiliation(s)
| | | | - Shishir Shishodia
- Department of Biology, Texas Southern University, Houston, TX 77004, USA; (V.M.); (A.S.)
| |
Collapse
|
11
|
Xiang D, Zhou L, Yang R, Yuan F, Xu Y, Yang Y, Qiao Y, Li X. Advances in Ferroptosis-Inducing Agents by Targeted Delivery System in Cancer Therapy. Int J Nanomedicine 2024; 19:2091-2112. [PMID: 38476278 PMCID: PMC10929151 DOI: 10.2147/ijn.s448715] [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: 11/09/2023] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Currently, cancer remains one of the most significant threats to human health. Treatment of most cancers remains challenging, despite the implementation of diverse therapies in clinical practice. In recent years, research on the mechanism of ferroptosis has presented novel perspectives for cancer treatment. Ferroptosis is a regulated cell death process caused by lipid peroxidation of membrane unsaturated fatty acids catalyzed by iron ions. The rapid development of bio-nanotechnology has generated considerable interest in exploiting iron-induced cell death as a new therapeutic target against cancer. This article provides a comprehensive overview of recent advancements at the intersection of iron-induced cell death and bionanotechnology. In this respect, the mechanism of iron-induced cell death and its relation to cancer are summarized. Furthermore, the feasibility of a nano-drug delivery system based on iron-induced cell death for cancer treatment is introduced and analyzed. Secondly, strategies for inducing iron-induced cell death using nanodrug delivery technology are discussed, including promoting Fenton reactions, inhibiting glutathione peroxidase 4, reducing low glutathione levels, and inhibiting system Xc-. Additionally, the article explores the potential of combined treatment strategies involving iron-induced cell death and bionanotechnology. Finally, the application prospects and challenges of iron-induced nanoagents for cancer treatment are discussed.
Collapse
Affiliation(s)
- Debiao Xiang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| | - Lili Zhou
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Rui Yang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Fang Yuan
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| | - Yilin Xu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Yuan Yang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan Province, People’s Republic of China
| | - Yong Qiao
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| | - Xin Li
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan Province, People’s Republic of China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan Province, People’s Republic of China
- The Clinical Application Research Institute of Antibiotics in Changsha, Changsha, Hunan Province, People’s Republic of China
| |
Collapse
|
12
|
Punziano C, Trombetti S, Cesaro E, Grosso M, Faraonio R. Antioxidant Systems as Modulators of Ferroptosis: Focus on Transcription Factors. Antioxidants (Basel) 2024; 13:298. [PMID: 38539832 PMCID: PMC10967371 DOI: 10.3390/antiox13030298] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 10/28/2024] Open
Abstract
Ferroptosis is a type of programmed cell death that differs from apoptosis, autophagy, and necrosis and is related to several physio-pathological processes, including tumorigenesis, neurodegeneration, senescence, blood diseases, kidney disorders, and ischemia-reperfusion injuries. Ferroptosis is linked to iron accumulation, eliciting dysfunction of antioxidant systems, which favor the production of lipid peroxides, cell membrane damage, and ultimately, cell death. Thus, signaling pathways evoking ferroptosis are strongly associated with those protecting cells against iron excess and/or lipid-derived ROS. Here, we discuss the interaction between the metabolic pathways of ferroptosis and antioxidant systems, with a particular focus on transcription factors implicated in the regulation of ferroptosis, either as triggers of lipid peroxidation or as ferroptosis antioxidant defense pathways.
Collapse
Affiliation(s)
- Carolina Punziano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Silvia Trombetti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy
| | - Elena Cesaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (C.P.); (S.T.); (E.C.)
| |
Collapse
|
13
|
Mohan S, Alhazmi HA, Hassani R, Khuwaja G, Maheshkumar VP, Aldahish A, Chidambaram K. Role of ferroptosis pathways in neuroinflammation and neurological disorders: From pathogenesis to treatment. Heliyon 2024; 10:e24786. [PMID: 38314277 PMCID: PMC10837572 DOI: 10.1016/j.heliyon.2024.e24786] [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: 02/17/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Ferroptosis is a newly discovered non-apoptotic and iron-dependent type of cell death. Ferroptosis mainly takes place owing to the imbalance of anti-oxidation and oxidation in the body. It is regulated via a number of factors and pathways both inside and outside the cell. Ferroptosis is closely linked with brain and various neurological disorders (NDs). In the human body, the brain contains the highest levels of polyunsaturated fatty acids, which are known as lipid peroxide precursors. In addition, there is also a connection of glutathione depletion and lipid peroxidation with NDs. There is growing evidence regarding the possible link between neuroinflammation and multiple NDs, such as Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, and stroke. Recent studies have demonstrated that disruptions of lipid reactive oxygen species (ROS), glutamate excitatory toxicity, iron homeostasis, and various other manifestations linked with ferroptosis can be identified in various neuroinflammation-mediated NDs. It has also been reported that damage-associated molecular pattern molecules including ROS are generated during the events of ferroptosis and can cause glial activation via activating neuroimmune pathways, which subsequently leads to the generation of various inflammatory factors that play a role in various NDs. This review summarizes the regulation pathways of ferroptosis, the link between ferroptosis as well as inflammation in NDs, and the potential of a range of therapeutic agents that can be used to target ferroptosis and inflammation in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Hassan A Alhazmi
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Rym Hassani
- Department of Mathematics, University College AlDarb, Jazan University, Jazan, Saudi Arabia
| | - Gulrana Khuwaja
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - V P Maheshkumar
- Department of Pharmacy, Annamalai University, Annamalai Nagar 608002, Tamil Nadu, India
| | - Afaf Aldahish
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Kumarappan Chidambaram
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| |
Collapse
|
14
|
Mei H, Wu D, Yong Z, Cao Y, Chang Y, Liang J, Jiang X, Xu H, Yang J, Shi X, Xie R, Zhao W, Wu Y, Liu Y. PM 2.5 exposure exacerbates seizure symptoms and cognitive dysfunction by disrupting iron metabolism and the Nrf2-mediated ferroptosis pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 910:168578. [PMID: 37981141 DOI: 10.1016/j.scitotenv.2023.168578] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
In recent years, air pollution has garnered global attention due to its ability to traverse borders and regions, thereby impacting areas far removed from the emission sources. While prior studies predominantly focused on the deleterious effects of PM2.5 on the respiratory and cardiovascular systems, emerging evidence has highlighted the potential risks of PM2.5 exposure to the central nervous system. Nonetheless, research elucidating the potential influences of PM2.5 exposure on seizures, specifically in relation to neuronal ferroptosis, remains limited. In this study, we investigated the potential effects of PM2.5 exposure on seizure symptoms and seizures-induced hippocampal neuronal ferroptosis. Our findings suggest that seizure patients residing in regions with high PM2.5 levels are more likely to disturb iron homeostasis and the Nrf2 dependent ferroptosis pathway compared to those living in areas with lower PM2.5 levels. The Morris Water Maze test, Racine scores, and EEG recordings in epileptic mice suggest that PM2.5 exposure can exacerbate seizure symptoms and cognitive dysfunction. Neurotoxic effects of PM2.5 exposure were demonstrated via Nissl staining and CCK-8 assays. Direct evidence of PM2.5-induced hippocampal neuronal ferroptosis was provided through TEM images. Additionally, increased Fe2+ and lipid ROS levels indirectly supported the notion of PM2.5-induced hippocampal ferroptosis. Therefore, our study underscores the necessity of preventing and controlling PM2.5 levels, particularly for patients with seizures.
Collapse
Affiliation(s)
- Huiya Mei
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongqin Wu
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zenghua Yong
- Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yingsi Cao
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuanjin Chang
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Junjie Liang
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiaofan Jiang
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hua Xu
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jiatao Yang
- Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Environment and Health Research Division, Public Health Research Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xian Shi
- Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Environment and Health Research Division, Public Health Research Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ruijin Xie
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China; Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Wenjing Zhao
- Yangzhou Key Laboratory of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
| | - Yu Wu
- Lab of Modern Environmental Toxicology, Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China; Environment and Health Research Division, Public Health Research Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China; The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yueying Liu
- Department of Pediatrics, Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
15
|
Ehlinger JV, Goodrich JM, Dolinoy DC, Watkins DJ, Cantoral A, Mercado-García A, Téllez-Rojo MM, Peterson KE. Associations between blood leukocyte DNA methylation and sustained attention in mid-to-late childhood. Epigenomics 2023; 15:965-981. [PMID: 37942546 PMCID: PMC10718163 DOI: 10.2217/epi-2023-0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023] Open
Abstract
Aims: To identify associations between DNA methylation (DNAm) across the epigenome and symptoms related to attention-deficit/hyperactivity disorder in a population of Hispanic children. Materials & methods: Among 517 participants in the ELEMENT study aged 9-18 years, we conducted an epigenome-wide association study examining associations between blood leukocyte DNAm and performance on the Conners' continuous performance test (CPT3). Results: DNAm at loci in or near ZNF814, ELF4 and OR6K6 and functional enrichment for gene pathways pertaining to ferroptosis, inflammation, immune response and neurotransmission were significantly related to CPT3 scores. Conclusion: DNAm was associated with CPT3 performance. Further analysis is warranted to understand how these genes and enriched pathways contribute to attention-deficit/hyperactivity disorder.
Collapse
Affiliation(s)
- Jessa V Ehlinger
- Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jaclyn M Goodrich
- Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dana C Dolinoy
- Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Deborah J Watkins
- Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | - Karen E Peterson
- Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Gao X, Hu W, Qian D, Bai X, He H, Li L, Sun S. The Mechanisms of Ferroptosis Under Hypoxia. Cell Mol Neurobiol 2023; 43:3329-3341. [PMID: 37458878 PMCID: PMC10477166 DOI: 10.1007/s10571-023-01388-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/06/2023] [Indexed: 09/05/2023]
Abstract
Ferroptosis is a new form of programmed cell death, which is characterized by the iron-dependent accumulation of lipid peroxidation and increase of ROS, resulting in oxidative stress and cell death. Iron, lipid, and multiple signaling pathways precisely control the occurrence and implementation of ferroptosis. The pathways mainly include Nrf2/HO-1 signaling pathway, p62/Keap1/Nrf2 signaling pathway. Activating p62/Keap1/Nrf2 signaling pathway inhibits ferroptosis. Nrf2/HO-1 signaling pathway promotes ferroptosis. Furthermore, some factors also participate in the occurrence of ferroptosis under hypoxia, such as HIF-1, NCOA4, DMT1. Meanwhile, ferroptosis is related with hypoxia-related diseases, such as MIRI, cancers, and AKI. Accordingly, ferroptosis appears to be a therapeutic target for hypoxia-related diseases.
Collapse
Affiliation(s)
- Xin Gao
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China
- 2020 Clinical Medicine Class 6, Kunming Medical University, Kunming, 650500, China
| | - Wei Hu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China
| | - Dianlun Qian
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| | - Xiangfeng Bai
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| | - Huilin He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China
| | - Lin Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, 650032, China.
| |
Collapse
|
17
|
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: 0] [Impact Index Per Article: 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
|
18
|
Emmerson JT, Do Carmo S, Liu Y, Shalhoub A, Liu A, Bonomo Q, Malcolm JC, Breuillaud L, Cuello AC. Progressive human-like tauopathy with downstream neurodegeneration and neurovascular compromise in a transgenic rat model. Neurobiol Dis 2023; 184:106227. [PMID: 37454780 DOI: 10.1016/j.nbd.2023.106227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
Tauopathies, including frontotemporal dementia (FTD) and Alzheimer's disease (AD), clinically present with progressive cognitive decline and the deposition of neurofibrillary tangles (NFTs) in the brain. Neurovascular compromise is also prevalent in AD and FTD however the relationship between tau and the neurovascular unit is less understood relative to other degenerative phenotypes. Current animal models confer the ability to recapitulate aspects of the CNS tauopathies, however, existing models either display overaggressive phenotypes, or do not develop neuronal loss or genuine neurofibrillary lesions. In this report, we communicate the longitudinal characterization of brain tauopathy in a novel transgenic rat model, coded McGill-R955-hTau. The model expresses the longest isoform of human P301S tau. Homozygous R955-hTau rats displayed a robust, progressive accumulation of mutated human tau leading to the detection of tau hyperphosphorylation and cognitive deficits accelerating from 14 months of age. This model features extensive tau hyperphosphorylation with endogenous tau recruitment, authentic neurofibrillary lesions, and tau-associated neuronal loss, ventricular dilation, decreased brain volume, and gliosis in aged rats. Further, we demonstrate how neurovascular integrity becomes compromised at aged life stages using a combination of electron microscopy, injection of the tracer horseradish peroxidase and immunohistochemical approaches.
Collapse
Affiliation(s)
- Joshua T Emmerson
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - Yingying Liu
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - Ali Shalhoub
- Department of Biochemistry, McGill University, Montreal H3A 0C7, Canada
| | - Ai Liu
- Integrated Program in Neuroscience, McGill University, Montreal H3A 1A1, Canada
| | - Quentin Bonomo
- Integrated Program in Neuroscience, McGill University, Montreal H3A 1A1, Canada
| | - Janice C Malcolm
- Department of Anatomy and Cell Biology, McGill University, Montreal H3A 0C7, Canada
| | - Lionel Breuillaud
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - A Claudio Cuello
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada; Integrated Program in Neuroscience, McGill University, Montreal H3A 1A1, Canada; Department of Pharmacology, Oxford University, Oxford OX13QT, UK.
| |
Collapse
|
19
|
Pap R, Pandur E, Jánosa G, Sipos K, Fritz FR, Nagy T, Agócs A, Deli J. Protective Effects of 3'-Epilutein and 3'-Oxolutein against Glutamate-Induced Neuronal Damage. Int J Mol Sci 2023; 24:12008. [PMID: 37569384 PMCID: PMC10418699 DOI: 10.3390/ijms241512008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Dietary lutein can be naturally metabolized to 3'-epilutein and 3'-oxolutein in the human body. The epimerization of lutein can happen in acidic pH, and through cooking, 3'-epilutein can be the product of the direct oxidation of lutein in the retina, which is also present in human serum. The 3'-oxolutein is the main oxidation product of lutein. Thus, the allylic oxidation of dietary lutein can result in the formation of 3'-oxolutein, which may undergo reduction either to revert to dietary lutein or epimerize to form 3'-epilutein. We focused on the effects of 3'-epilutein and 3'-oxolutein itself and on glutamate-induced neurotoxicity on SH-SY5Y human neuroblastoma cells to identify the possible alterations in oxidative stress, inflammation, antioxidant capacity, and iron metabolism that affect neurological function. ROS measurements were performed in the differently treated cells. The inflammatory state of cells was followed by TNFα, IL-6, and IL-8 cytokine ELISA measurements. The antioxidant status of the cells was determined by the total antioxidant capacity kit assay. The alterations of genes related to ferroptosis and lipid peroxidation were followed by gene expression measurements; then, thiol measurements were performed. Lutein metabolites 3'-epilutein and 3'-oxolutein differently modulated the effect of glutamate on ROS, inflammation, ferroptosis-related iron metabolism, and lipid peroxidation in SH-SY5Y cells. Our results revealed the antioxidant and anti-inflammatory features of 3'-epilutein and 3'-oxolutein as possible protective agents against glutamate-induced oxidative stress in SH-SY5Y cells, with greater efficacy in the case of 3'-epilutein.
Collapse
Affiliation(s)
- Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Gergely Jánosa
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Ferenc Rómeó Fritz
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; (R.P.); (E.P.); (G.J.); (K.S.); (F.R.F.)
| | - Tamás Nagy
- Department of Laboratory Medicine, Faculty of Medical Sciences, University of Pécs, Ifjúság út 13, H-7624 Pécs, Hungary;
| | - Attila Agócs
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary;
| | - József Deli
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary;
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| |
Collapse
|
20
|
Singh A, Kukal S, Kanojia N, Singh M, Saso L, Kukreti S, Kukreti R. Lipid Mediated Brain Disorders: A Perspective. Prostaglandins Other Lipid Mediat 2023; 167:106737. [PMID: 37086954 DOI: 10.1016/j.prostaglandins.2023.106737] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/24/2023]
Abstract
The brain, one of the most resilient organs of the body is highly enriched in lipid content, suggesting the essential role of lipids in brain physiological activities. Lipids constitute an important structural part of the brain and act as a rich source of metabolic energy. Besides, lipids in their bioactive form (known as bioactive lipids) play an essential signaling and regulatory role, facilitating neurogenesis, synaptogenesis, and cell-cell communication. Brain lipid metabolism is thus a tightly regulated process. Any alteration/dysregulation of lipid metabolism greatly impact brain health and activity. Moreover, since central nervous system (CNS) is the most metabolically active system and lacks an efficient antioxidative defence system, it acts as a hub for the production of reactive oxygen species (ROS) and subsequent lipid peroxidation. These peroxidation events are reported during pathological changes such as neuronal tissue injury and inflammation. Present review is a modest attempt to gain insights into the role of dysregulated bioactive lipid levels and lipid oxidation status in the pathogenesis and progression of neurodegenerative disorders. This may open up new avenues exploiting lipids as the therapeutic targets for improving brain health, and treatment of nervous system disorders.
Collapse
Affiliation(s)
- Anju Singh
- Department of Chemistry, Ramjas College, University of Delhi, Delhi 110007, India; Nucleic Acids Research Lab, Department of Chemistry, University of Delhi (North Campus), Delhi 110007, India
| | - Samiksha Kukal
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India
| | - Neha Kanojia
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India
| | - Mahak Singh
- Department of Chemistry, Ramjas College, University of Delhi, Delhi 110007, India
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Shrikant Kukreti
- Nucleic Acids Research Lab, Department of Chemistry, University of Delhi (North Campus), Delhi 110007, India
| | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India.
| |
Collapse
|
21
|
Payne A, Taka E, Adinew GM, Soliman KFA. Molecular Mechanisms of the Anti-Inflammatory Effects of Epigallocatechin 3-Gallate (EGCG) in LPS-Activated BV-2 Microglia Cells. Brain Sci 2023; 13:632. [PMID: 37190597 PMCID: PMC10137201 DOI: 10.3390/brainsci13040632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Chronic neuroinflammation is associated with many neurodegenerative diseases, such as Alzheimer's. Microglia are the brain's primary immune cells, and when activated, they release various proinflammatory cytokines. Several natural compounds with anti-inflammatory and antioxidant properties, such as epigallocatechin 3-gallate (EGCG), may provide a promising strategy for inflammation-related neurodegenerative diseases involving activated microglia cells. The objective of the current study was to examine the molecular targets underlying the anti-inflammatory effects of EGCG in activated microglia cells. BV-2 microglia cells were grown, stimulated, and treated with EGCG. Cytotoxicity and nitric oxide (NO) production were evaluated. Immunoassay, PCR array, and WES™ Technology were utilized to evaluate inflammatory, neuroprotective modulators as well as signaling pathways involved in the mechanistic action of neuroinflammation. Our findings showed that EGCG significantly inhibited proinflammatory mediator NO production in LPS-stimulated BV-2 microglia cells. In addition, ELISA analysis revealed that EGCG significantly decreases the release of proinflammatory cytokine IL-6 while it increases the release of TNF-α. PCR array analysis showed that EGCG downregulated MIF, CCL-2, and CSF2. It also upregulated IL-3, IL-11, and TNFS10. Furthermore, the analysis of inflammatory signaling pathways showed that EGCG significantly downregulated mRNA expression of mTOR, NF-κB2, STAT1, Akt3, CCL5, and SMAD3 while significantly upregulating the expression of mRNA of Ins2, Pld2, A20/TNFAIP3, and GAB1. Additionally, EGCG reduced the relative protein expression of NF-κB2, mTOR, and Akt3. These findings suggest that EGCG may be used for its anti-inflammatory effects to prevent neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health (COPPS, IPH), Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
22
|
Cao Y, Li M, Gu L, Zhao X, Zhou A, Miao Y, Wu Y, Ke Z, Hu R, Wang Z, Wu X. Chinese traditional formula Kaixin San suppressed ferroptosis of hippocampal neurons and cardiomyocytes in mice with paradoxical sleep deprivation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:116034. [PMID: 36529245 DOI: 10.1016/j.jep.2022.116034] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kaixin San (KXS) is one of the most famous traditional Chinese formulas prescribed by Sun Simiao in 652 Christian era. It is composed of Panax ginseng C.A.Mey, Polygala tenuifolia, Poria cocos and Acorus calamus var. angustatus Besser. KXS is widely used for the treatment of emotion-thought disease, such as settling fright, quieting the spirit and nourishing the heart. However, whether KXS benefits hippocampal neurons and myocardial cells of mice impaired by paradoxical sleep deprivation (PSD) and its mechanism remains unclear. AIM OF THE STUDY This study was aimed to investigate the effect of KXS on hippocampal neuron and cardiac ferroptosis in rapid-eye-movement (REM) sleep deprived mice and clarify its potential mechanism. MATERIALS AND METHODS PSD was induced by a modified multi-platform method. Morris water maze (MWM) was used to detect the ability of learning and memory. Cardiac morphological changes were assessed by hematoxylin and eosin (HE) staining. Heart rate was detected by a PowerLab multichannel physiological recorder. Serum levels of atrial natriuretic peptide (ANP) and lactate dehydrogenase (LDH) were measured with biochemical kits. Transmission electron microscopy (TEM), immunofluorescent, and Western blotting analysis were used to observe the process and pathway of ferrotosis in hippocampus tissue and heart tissue of PSD mice. RESULTS KXS administration improved the impaired learning and memory of PSD mice. It prevented the damage of mitochondria in the hippocampus and heart of PSD mice. KXS also alleviated the myocardial injury, such as morphological damage, abnormal heart rate, serum ANP, and serum LDH induced by PSD. Further study disclosed that KXS reversed the expressions of proteins involved in ferroptosis such as TFRC, SLC7A11/xCT, GPX-4, ACSL4, and FTH1 in hippocampus and heart tissues. CONCLUSIONS KXS improved learning and memory of mice with REM sleep deprivation, which was closely associated with suppressed ferroptosis in hippocampal neurons and myocardiocytes.
Collapse
Affiliation(s)
- Yin Cao
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Mingrui Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Lihua Gu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The Sate Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Zhao
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The Sate Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - An Zhou
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Yuping Miao
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Yi Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Zunji Ke
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Rongfeng Hu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China.
| | - Zhengtao Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The Sate Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The Sate Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
23
|
Role of Ferroptosis in Regulating the Epithelial-Mesenchymal Transition in Pulmonary Fibrosis. Biomedicines 2023; 11:biomedicines11010163. [PMID: 36672671 PMCID: PMC9856078 DOI: 10.3390/biomedicines11010163] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Idiopathic pulmonary fibrosis is a chronic interstitial lung disease whose pathogenesis involves a complex interaction of cell types and signaling pathways. Lung epithelial cells responding to repeated injury experience persistent inflammation and sustained epithelial-mesenchymal transition (EMT). The persistence of EMT-induced signals generates extracellular matrix accumulation, thereby causing fibrosis. Ferroptosis is a newly characterized iron-dependent non-apoptotic regulated cell death. Increased iron accumulation can increase iron-induced oxidant damage in alveolar epithelial cells. Studies have demonstrated that iron steady states and oxidation steady states play an important role in the iron death regulation of EMT. This review summarizes the role of ferroptosis in regulating EMT in pulmonary fibrosis, aiming to provide a new idea for the prevention and treatment of this disease.
Collapse
|
24
|
Yu S, Zeng Y, Ruan C, Bai L, Liang Z. Protective effects of brain and muscle ARNT-like gene 1 on oxidized low-density lipoprotein-induced human brain microvascular endothelial cell injury by alleviating ferroptosis. Hum Exp Toxicol 2023; 42:9603271231184630. [PMID: 37343012 DOI: 10.1177/09603271231184630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023]
Abstract
Ferroptosis plays an important role in atherosclerotic cerebrovascular diseases. The brain and muscle ARNT-like gene 1 (BMAL1) is an important mediator in the progression of cerebrovascular diseases. However, whether BMAL1 regulates ferroptosis in atherosclerotic cerebrovascular diseases remains obscure. Here, human brain microvascular endothelial cells (HBMECs) were exposed to oxidized low-density lipoprotein (ox-LDL) to imitate cerebrovascular atherosclerosis. It was found that ox-LDL treatment induced ferroptosis events and reduced BMAL1 expression in HBMECs, which could be reversed by ferroptosis inhibitor ferrostatin-1. Furthermore, BMAL1 overexpression markedly mitigated ox-LDL-induced ferroptosis events and cell damage. Moreover, BMAL1 overexpression significantly promoted nuclear factor erythroid 2-related factor 2 (Nrf2) expression in HBMECs under ox-LDL conditions. And, Nrf2 silencing attenuated the protective effects of BMAL1 on ox-LDL-stimulated HBMEC damage and ferroptosis. Altogether, our findings delineate the cerebrovascular protective role of BMAL1/Nrf2 by antagonizing ferroptosis in response to ox-LDL stimulation and provide novel perspectives for therapeutic strategies for atherosclerotic cerebrovascular diseases.
Collapse
Affiliation(s)
- Shui Yu
- Department of Neurosurgery, The People's Hospital of Dujiangyan, Dujiangyan, Sichuan Province, China
| | - Yijun Zeng
- Department of Neurosurgery, The People's Hospital of Dujiangyan, Dujiangyan, Sichuan Province, China
| | - Chenbin Ruan
- Department of Neurology, The People's Hospital of Dujiangyan, Dujiangyan, Sichuan Province, China
| | - Lei Bai
- Department of Neurology, The People's Hospital of Dujiangyan, Dujiangyan, Sichuan Province, China
| | - Zhang Liang
- Department of Neurosurgery, The People's Hospital of Dujiangyan, Dujiangyan, Sichuan Province, China
| |
Collapse
|
25
|
Bammidi S, Hose S, Handa JT, Sinha D, Ghosh S. Thermal Shift Assay in Ferroptosis. Methods Mol Biol 2023; 2712:179-186. [PMID: 37578706 PMCID: PMC11059966 DOI: 10.1007/978-1-0716-3433-2_16] [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] [Indexed: 08/15/2023]
Abstract
Ferroptosis is a recently described process of cell death that is dependent on unregulated cellular iron accumulation with induction of oxidative stress. Ferroptosis has been linked to several human diseases; therefore, investigations aimed at better understanding the pathway and elucidating avenues for future drug development are warranted. Current assays that target ferroptosis/oxidative stress in cells is limited to western blotting and imaging techniques, and unfortunately provide only a broad understanding that is insufficient to effectively assess novel drugs (ligands). Specifically, these assays do not provide insights about ligand interactions with specific proteins associated with these processes. Herein, we discuss a cell-based thermal shift assay that enables screening of ligands under specific cellular conditions for targeting ferroptosis and/or oxidative stress pathways. These data would provide detailed preliminary evidence required for drug development that targets this pathway.
Collapse
Affiliation(s)
- Sridhar Bammidi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - James T Handa
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
26
|
Zhao J, Wang C, Sun W, Li C. Tailoring Materials for Epilepsy Imaging: From Biomarkers to Imaging Probes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2203667. [PMID: 35735191 DOI: 10.1002/adma.202203667] [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] [Received: 04/24/2022] [Revised: 06/11/2022] [Indexed: 06/15/2023]
Abstract
Excising epileptic foci (EF) is the most efficient approach for treating drug-resistant epilepsy (DRE). However, owing to the vast heterogeneity of epilepsies, EF in one-third of patients cannot be accurately located, even after exhausting all current diagnostic strategies. Therefore, identifying biomarkers that truly represent the status of epilepsy and fabricating probes with high targeting specificity are prerequisites for identifying the "concealed" EF. However, no systematic summary of this topic has been published. Herein, the potential biomarkers of EF are first summarized and classified into three categories: functional, molecular, and structural aberrances during epileptogenesis, a procedure of nonepileptic brain biasing toward epileptic tissue. The materials used to fabricate these imaging probes and their performance in defining the EF in preclinical and clinical studies are highlighted. Finally, perspectives for developing the next generation of probes and their challenges in clinical translation are discussed. In general, this review can be helpful in guiding the development of imaging probes defining EF with improved accuracy and holds promise for increasing the number of DRE patients who are eligible for surgical intervention.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Zhangheng Road 826, Shanghai, 201203, China
| | - Cong Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Zhangheng Road 826, Shanghai, 201203, China
- Academy for Engineering and Technology, Fudan University, 20 Handan Road, Yangpu District, Shanghai, 200433, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 200031, China
| | - Wanbing Sun
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Zhangheng Road 826, Shanghai, 201203, China
- State Key Laboratory of Medical Neurobiology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
27
|
Villalón-García I, Povea-Cabello S, Álvarez-Córdoba M, Talaverón-Rey M, Suárez-Rivero JM, Suárez-Carrillo A, Munuera-Cabeza M, Reche-López D, Cilleros-Holgado P, Piñero-Pérez R, Sánchez-Alcázar JA. Vicious cycle of lipid peroxidation and iron accumulation in neurodegeneration. Neural Regen Res 2022; 18:1196-1202. [PMID: 36453394 PMCID: PMC9838166 DOI: 10.4103/1673-5374.358614] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Lipid peroxidation and iron accumulation are closely associated with neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, or neurodegeneration with brain iron accumulation disorders. Mitochondrial dysfunction, lipofuscin accumulation, autophagy disruption, and ferroptosis have been implicated as the critical pathomechanisms of lipid peroxidation and iron accumulation in these disorders. Currently, the connection between lipid peroxidation and iron accumulation and the initial cause or consequence in neurodegeneration processes is unclear. In this review, we have compiled the known mechanisms by which lipid peroxidation triggers iron accumulation and lipofuscin formation, and the effect of iron overload on lipid peroxidation and cellular function. The vicious cycle established between both pathological alterations may lead to the development of neurodegeneration. Therefore, the investigation of these mechanisms is essential for exploring therapeutic strategies to restrict neurodegeneration. In addition, we discuss the interplay between lipid peroxidation and iron accumulation in neurodegeneration, particularly in PLA2G6-associated neurodegeneration, a rare neurodegenerative disease with autosomal recessive inheritance, which belongs to the group of neurodegeneration with brain iron accumulation disorders.
Collapse
Affiliation(s)
- Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Juan M. Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - José A. Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain,Correspondence to: José A. Sánchez-Alcázar, MD, PhD, .
| |
Collapse
|
28
|
Li X, Zhu S, Li Z, Meng Y, Huang S, Yu Q, Li B. Melittin induces ferroptosis and ER stress-CHOP-mediated apoptosis in A549 cells. Free Radic Res 2022; 56:398-410. [PMID: 36194238 DOI: 10.1080/10715762.2022.2131551] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Melittin is a natural polypeptide present in bee venom, with significant anti-tumor activity. Melittin has been reported to induce cell death in lung carcinoma cell line A549 cells, suggesting an excellent potential for treating lung cancer. However, the core mechanism underlying melittin-induced cell death in A549 cells remains unclear. This work reports that melittin induces reactive oxygen species (ROS) burst, upregulates intracellular Fe2+ levels, disrupts the glutathione-glutathione peroxidase 4 antioxidant system, and increases lipid peroxide accumulation, eventually inducing cell death, indicating that ferroptosis may be involved in the antitumor effects of melittin in A549 cells. Furthermore, A549 cells treated with the ferroptosis inhibitors ferrostatin-1 and deferoxamine demonstrated that these inhibitors could reverse the cell death induced by melittin, further confirming that melittin induces A549 cell death via ferroptosis. Furthermore, the results also illustrated that melittin activated the endoplasmic reticulum (ER) stress-CHOP (C/EBP homologous protein) apoptotic signal, closely associated with high-level intracellular ROS. The ER stress inhibitor, 4-Phenyl butyric acid, was used to confirm that ER stress-CHOP apoptotic signaling is another molecular mechanism of melittin-induced A549 cell death. Thus, our results demonstrate that ferroptosis and ER stress-CHOP signaling are key molecular mechanisms of melittin-induced cell death in lung cancer.Key policy highlightsMelittin upregulates intracellular Fe2+ levels, leading to the accumulation of lipid peroxides in A549 cells.Melittin disrupts the glutathione-glutathione peroxidase 4 antioxidant system in A549 cells.Melittin induces activation of endoplasmic reticulum stress-C/EBP homologous protein apoptosis signal.Ferroptosis and ER stress are the core molecular mechanisms underlying melittin-induced apoptosis in A549 cells.
Collapse
Affiliation(s)
- Xuan Li
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Sen Zhu
- School of Life Sciences, Lanzhou University, 730030, Lanzhou, China
| | - Zheng Li
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Yuqi Meng
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Sujie Huang
- School of Basic Medical Sciences, Lanzhou University, 730030, Lanzhou, China
| | - Qiyao Yu
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| | - Bin Li
- Department of Thoracic Surgery, Lanzhou University Second Hospital, Lanzhou University Second Clinical Medical College, Lanzhou University 730030, Lanzhou, China
| |
Collapse
|
29
|
Li C, Wu Z, Xue H, Gao Q, Zhang Y, Wang C, Zhao P. Ferroptosis contributes to hypoxic-ischemic brain injury in neonatal rats: Role of the SIRT1/Nrf2/GPx4 signaling pathway. CNS Neurosci Ther 2022; 28:2268-2280. [PMID: 36184790 PMCID: PMC9627393 DOI: 10.1111/cns.13973] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS Hypoxic-ischemic brain injury (HIBI) often results in cognitive impairments. Herein, we investigated the roles of ferroptosis in HIBI and the underlying signaling pathways. METHODS Ferrostatin-1 (Fer-1) or resveratrol (Res) treatments were administered intracerebroventricularly 30 min before HIBI in 7-day-old rats. Glutathione peroxidase 4 (GPx4) expression, malondialdehyde (MDA) concentration, iron content, mitochondrial morphology, and the expression of silent information regulator factor 2-related enzyme 1 (SIRT1) and nuclear factor erythroid-2-related factor 2 (Nrf2) were measured after HIBI. Additionally, the weight ratio of left/right hemisphere, brain morphology, Nissl staining, and the Morris water maze test were conducted to estimate brain damage. RESULTS At 24-h post-HIBI, GPx4 expression was decreased, and MDA concentration and iron content were increased in the hippocampus. HIBI led to mitochondrial atrophy, brain atrophy/damage, and resultant learning and memory impairments, which were alleviated by Fer-1-mediated inhibition of ferroptosis. Furthermore, Res-mediated SIRT1 upregulation increased Nrf2 and GPx4 expression, thereby attenuating ferroptosis, reducing brain atrophy/damage, and improving learning and memory abilities. CONCLUSION The results demonstrated that during HIBI, ferroptosis occurs via the SIRT1/Nrf2/GPx4 signaling pathway, suggesting it as a potential therapeutic target for inhibiting ferroptosis and ameliorating HIBI-induced cognitive impairments.
Collapse
Affiliation(s)
- Chang Li
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ziyi Wu
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Hang Xue
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Qiushi Gao
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yahan Zhang
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Changming Wang
- Department of AnesthesiologyPeople's Hospital of China Medical University (Liaoning Provincial People's Hospital)ShenyangLiaoningChina
| | - Ping Zhao
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
30
|
Zhang C, Liu N. Ferroptosis, necroptosis, and pyroptosis in the occurrence and development of ovarian cancer. Front Immunol 2022; 13:920059. [PMID: 35958626 PMCID: PMC9361070 DOI: 10.3389/fimmu.2022.920059] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common malignancies that causes death in women and is a heterogeneous disease with complex molecular and genetic changes. Because of the relatively high recurrence rate of OC, it is crucial to understand the associated mechanisms of drug resistance and to discover potential target for rational targeted therapy. Cell death is a genetically determined process. Active and orderly cell death is prevalent during the development of living organisms and plays a critical role in regulating life homeostasis. Ferroptosis, a novel type of cell death discovered in recent years, is distinct from apoptosis and necrosis and is mainly caused by the imbalance between the production and degradation of intracellular lipid reactive oxygen species triggered by increased iron content. Necroptosis is a regulated non-cysteine protease–dependent programmed cell necrosis, morphologically exhibiting the same features as necrosis and occurring via a unique mechanism of programmed cell death different from the apoptotic signaling pathway. Pyroptosis is a form of programmed cell death that is characterized by the formation of membrane pores and subsequent cell lysis as well as release of pro-inflammatory cell contents mediated by the abscisin family. Studies have shown that ferroptosis, necroptosis, and pyroptosis are involved in the development and progression of a variety of diseases, including tumors. In this review, we summarized the recent advances in ferroptosis, necroptosis, and pyroptosis in the occurrence, development, and therapeutic potential of OC.
Collapse
|
31
|
Feng B, Wang K, Wang Z, Niu H, Wang G, Chen Y, Zhang H. Mitochondrial-Targeted Ratiometric Fluorescent Probe to Monitor ClO - Induced by Ferroptosis in Living Cells. Front Chem 2022; 10:909670. [PMID: 35755249 PMCID: PMC9218690 DOI: 10.3389/fchem.2022.909670] [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: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 11/24/2022] Open
Abstract
Ferroptosis is a type of iron-dependent programmed cell death. Once such kind of death occurs, an individual cell would undergo a series of changes related to reactive oxygen species (ROS) in mitochondria. A mitochondrial-targeted ratiometric fluorescent probe (MBI-OMe) was developed to specifically detect ferroptosis-induced ClO−, whose recognition group is p-methoxyphenol, and the mitochondrial-targeted group is benzimidazole. The fluorescence of MBI-OMe was first quenched by 30 μM of Fe3+, and then MBI-OMe appeared as a ratiometric signal at 477 nm and 392 nm in response to ferroptosis-induced ClO− in living cells. MBI-OMe was successfully used to evaluate changes in ClO− induced by ferroptosis.
Collapse
Affiliation(s)
- Beidou Feng
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Kui Wang
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Zhe Wang
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Huiyu Niu
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Ge Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yuehua Chen
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Hua Zhang
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| |
Collapse
|
32
|
Assessing the neurotoxicity of airborne nano-scale particulate matter in human iPSC-derived neurons using a transcriptomics benchmark dose model. Toxicol Appl Pharmacol 2022; 449:116109. [DOI: 10.1016/j.taap.2022.116109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022]
|
33
|
Wu X, Li R, Hong Q, Chi X. Development and Validation of a Novel Diagnostic Model for Childhood Autism Spectrum Disorder Based on Ferroptosis-Related Genes. Front Psychiatry 2022; 13:886055. [PMID: 35633787 PMCID: PMC9133509 DOI: 10.3389/fpsyt.2022.886055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/26/2022] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder (ASD) is a highly genetic heterogeneous neurodevelopmental disorder, which is usually considered a heritable and heterogeneous neurodevelopmental disorder and has caused a great burden to society and families. Emerging roles of ferroptosis have been observed in neurological disorders. This study aimed to construct a diagnostic model based on ferroptosis-related genes (FRGs) to contribute to the early and precise diagnosis of childhood ASD. In the candidate FRGs, we identified 27 differentially expressed genes (DEGs) between ASD patients and typically developing (TD) controls. Four key FRGs were identified using the random forest analysis for further analysis. Utilization of the four gene expression, we constructed a diagnostic model and the AUC value in the training dataset (GSE18123) is 0.7002. We deem that a patient with a score less than 0.9904 is likely to have ASD. Three validation datasets (GSE111176, GSE113834, and GSE28521) were collected and the AUC value is 0.7442, 0.7444, and 0.6474, respectively. A multi-factor regulatory network based on four FRGs indicated that RORA, EAF1, NFYB, miR-4703-3p, and miR-6073 may play a role in the development of ASD. In addition, we found piperaquine may have the potential to be a promising drug for the treatment of ASD. Overall, we constructed a diagnostic model of childhood ASD, which could contribute to the precision diagnosis and timely treatment of childhood ASD.
Collapse
Affiliation(s)
- Xiaolu Wu
- Department of Child Health Care, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Ran Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Hong
- Department of Child Health Care, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Xia Chi
- Department of Child Health Care, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|