1
|
Yao Z, Jiao Q, Du X, Jia F, Chen X, Yan C, Jiang H. Ferroptosis in Parkinson's disease -- The iron-related degenerative disease. Ageing Res Rev 2024; 101:102477. [PMID: 39218077 DOI: 10.1016/j.arr.2024.102477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) is a prevalent and advancing age-related neurodegenerative disorder, distinguished by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Iron regional deposit in SNpc is a significant pathological characteristic of PD. Brain iron homeostasis is precisely regulated by iron metabolism related proteins, whereas disorder of these proteins can damage neurons and glial cells in the brain. Additionally, growing studies have reported iron metabolism related proteins are involved in the ferroptosis progression in PD. However, the effect of these proteins in the ferroptosis of PD has not been systematically summarized. This review focuses on the roles of iron metabolism related proteins in the ferroptosis of PD. Finally, we put forward the iron early diagnosis according to the observation of iron deposits in the brain and showed the recent advances in iron chelation therapy in PD.
Collapse
Affiliation(s)
- Zhengyang Yao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Fengju Jia
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Chunling Yan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hong Jiang
- Qingdao Key Laboratory of Neurorehabilitation, University of Health and Rehabilitation Sciences, Qingdao, 266113, China.
| |
Collapse
|
2
|
Zhang X, Liu Z, Li Z, Qi L, Huang T, Li F, Li M, Wang Y, Ma Z, Gao Y. Ferroptosis pathways: Unveiling the neuroprotective power of cistache deserticola phenylethanoid glycosides. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118465. [PMID: 38944360 DOI: 10.1016/j.jep.2024.118465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/22/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cistanche deserticola is a kind of parasitic plant living in the roots of desert trees. It is a rare Chinese medicine, which has the effect of tonifying kidney Yang, benefiting essence and blood and moistening the intestinal tract. Cistache deserticola phenylethanoid glycoside (PGS), an active component found in Cistanche deserticola Ma, have potential kidney tonifying, intellectual enhancing, and neuroprotective effects. Cistanche total glycoside capsule has been marketed to treat vascular dementia disease. AIM OF THE STUDY To identify the potential renal, intellectual enhancing and neuroprotective effects of PGS and explore the exact targets and mechanisms of PGS. MATERIALS AND METHODS This study systematically investigated the four types of pathways leading to ferroptosis through transcriptome, metabolome, ultrastructure and molecular biology techniques and explored the molecular mechanism by which multiple PGS targets and pathways synergistically exert neuroprotective effects on hypoxia. RESULTS PGS alleviated learning and memory dysfunction and pathological injury in mice exposed to hypobaric hypoxia by attenuating hypobaric hypoxia-induced hippocampal histopathological damage, impairing blood‒brain barrier integrity, increasing oxidative stress levels, and increasing the expression of cognitive proteins. PGS reduced the formation of lipid peroxides and improved ferroptosis by upregulating the GPX-4/SCL7A311 axis and downregulating the ACSL4/LPCAT3/LOX axis. PGS also reduced ferroptosis by facilitating cellular Fe2+ efflux and regulating mitochondrial Fe2+ transport and effectively antagonized cell ferroptosis induced by erastin (a ferroptosis inducer). CONCLUSIONS This study demonstrated the mechanism by which PGS prevents hypobaric hypoxic nerve injury through four types of ferroptosis pathways, achieved neuroprotective effects and alleviated learning and memory dysfunction in hypobaric hypoxia mice. This study provides a theoretical basis for the development and application of PGS.
Collapse
Affiliation(s)
- Xianxie Zhang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zuoxu Liu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zhihui Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Ling Qi
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Tianke Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Fang Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Maoxing Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Yuguang Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zengchun Ma
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Yue Gao
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China.
| |
Collapse
|
3
|
Tang S, Chen L. The recent advancements of ferroptosis of gynecological cancer. Cancer Cell Int 2024; 24:351. [PMID: 39462352 PMCID: PMC11520064 DOI: 10.1186/s12935-024-03537-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
Ovarian, endometrial, and cervical cancer are the most common types of gynecologic tumor in women. Surgery, combined with radiotherapy and chemotherapy, is commonly used to treat these tumors. Unfortunately, difficulties in early diagnosis and acquired drug resistance have resulted in poor outcomes for most patients. Ferroptosis is a form of regulated cell death that depends on iron and is characterized by iron accumulation, reactive oxygen species production, and lipid peroxidation. The strong association between ferroptosis and many diseases, especially tumor diseases, has been confirmed by numerous studies. Many studies have demonstrated that ferroptosis is involved in initiating, progressing and metastasizing gynecologic tumors. This review summarizes the pathogenesis of ferroptosis and its association with the development, treatment, and prognosis of gynecologic tumors, and further explore the potential utility of ferroptosis in treating gynecologic tumors.
Collapse
Affiliation(s)
- Shenglan Tang
- Department of the First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People's Republic of China
| | - Li Chen
- Department of Obstetrics and Gynecology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, 261 Huansha Road, Shangcheng, Hangzhou, Zhejiang, 310006, People's Republic of China.
| |
Collapse
|
4
|
Wang S, Wu Z, Bu X, Peng X, Zhou Q, Song W, Gao W, Wang W, Xia Z. MEF2C Alleviates Postoperative Cognitive Dysfunction by Repressing Ferroptosis. CNS Neurosci Ther 2024; 30:e70066. [PMID: 39350345 PMCID: PMC11442332 DOI: 10.1111/cns.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Ferroptosis, a form of programmed cell death featured by lipid peroxidation, has been proposed as a potential etiology for postoperative cognitive dysfunction (POCD). Myocyte-specific enhancer factor 2C (MEF2C), a transcription factor expressed in various brain cell types, has been implicated in cognitive disorders. This study sought to ascertain whether MEF2C governs postoperative cognitive capacity by affecting ferroptosis. METHODS Transcriptomic analysis of public data was used to identify MEF2C as a candidate differentially expressed gene in the hippocampus of POCD mice. The POCD mouse model was established via aseptic laparotomy under isoflurane anesthesia after treatment with recombinant adeno-associated virus 9 (AAV9)-mediated overexpression of MEF2C and/or the glutathione peroxidase 4 (GPX4) inhibitor RSL3. Cognitive performance, Nissl staining, and ferroptosis-related parameters were assessed. Dual-luciferase reporter gene assays and chromatin immunoprecipitation assays were implemented to elucidate the mechanism by which MEF2C transcriptionally activates GPX4. RESULTS MEF2C mRNA and protein levels decreased in the mouse hippocampus following anesthesia and surgery. MEF2C overexpression ameliorated postoperative memory decline, hindered lipid peroxidation and iron accumulation, and enhanced antioxidant capacity, which were reversed by RSL3. Additionally, MEF2C was found to directly bind to the Gpx4 promoter and activate its transcription. CONCLUSIONS Our findings suggest that MEF2C may be a promising therapeutic target for POCD through its negative modulation of ferroptosis.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zankai Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xueshan Bu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuan Peng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qin Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenqin Song
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenwei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
5
|
Zheng D, Jin S, Liu PS, Ye J, Xie X. Targeting ferroptosis by natural products in pathophysiological conditions. Arch Toxicol 2024; 98:3191-3208. [PMID: 38987487 DOI: 10.1007/s00204-024-03812-4] [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: 05/09/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Ferroptosis is a form of cell death that is induced by iron-mediated accumulation of lipid peroxidation. The involvement of ferroptosis in different pathophysiological conditions has offered new perspectives on potential therapeutic interventions. Natural products, which are widely recognized for their significance in drug discovery and repurposing, have shown great promise in regulating ferroptosis by targeting various ferroptosis players. In this review, we discuss the regulatory mechanisms of ferroptosis and its implications in different pathological conditions. We dissect the interactions between natural products and ferroptosis in cancer, ischemia/reperfusion, neurodegenerative diseases, acute kidney injury, liver injury, and cardiomyopathy, with an emphasis on the relevance of ferroptosis players to disease targetability.
Collapse
Affiliation(s)
- Daheng Zheng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Pu-Ste Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Jianping Ye
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
6
|
Huan H, Lyamzaev KG, Panteleeva AA, Chernyak BV. Mitochondrial lipid peroxidation is necessary but not sufficient for induction of ferroptosis. Front Cell Dev Biol 2024; 12:1452824. [PMID: 39324070 PMCID: PMC11422198 DOI: 10.3389/fcell.2024.1452824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
Ferroptosis, a form of regulated cell death mediated by lipid peroxidation (LPO), has become the subject of intense research due to its potential therapeutic applications in cancer chemotherapy as well as its pathophysiological role in ischemic organ injury. The role of mitochondrial lipid peroxidation (LPO) in ferroptosis remains poorly understood. We show that supplementation of exogenous iron in the form of ferric ammonium citrate (FAC) in combination with buthionine sulfoximine (BSO, an inhibitor of glutathione biosynthesis) induces mitochondrial lipid peroxidation that precedes ferroptosis in normal human fibroblasts. The mitochondrial-targeted antioxidant SkQ1 and the redox mediator methylene blue, which inhibits the production of reactive oxygen species (ROS) in complex I of the mitochondrial electron transport chain, prevent both mitochondrial lipid peroxidation and ferroptosis, but do not affect the cytosolic ROS accumulation. These data indicate that mitochondrial lipid peroxidation is required for ferroptosis induced by exogenous iron. FAC in the absence of BSO stimulates mitochondrial peroxidation without reducing cell viability. Glutathione depletion by BSO does not affect FAC-induced mitochondrial LPO but strongly stimulates the accumulation of ROS in the cytosol. These data allow us to conclude that mitochondrial LPO is not sufficient for ferroptosis and that cytosolic ROS mediates additional oxidative events that stimulate ferroptosis in conjunction with mitochondrial LPO.
Collapse
Affiliation(s)
- He Huan
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin G. Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- The “Russian Clinical Research Center for Gerontology” of the Ministry of Healthcare of the Russian Federation, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alisa A. Panteleeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Boris V. Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
7
|
Shi Y, Xu N, Liu B, Ma Y, Fu X, Shang Y, Huang Q, Yao Q, Chen J, Li H. Mifepristone protects acetaminophen induced liver injury through NRF2/GSH/GST mediated ferroptosis suppression. Free Radic Biol Med 2024; 222:229-243. [PMID: 38906233 DOI: 10.1016/j.freeradbiomed.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Ferroptosis is a form of iron-dependent cell death that has attracted significant attention for its potential role in numerous diseases. Targeted inhibition of ferroptosis could be of potential use in treating diseases: such as drug induced liver injury (DILI). Ferroptosis can be antagonized by the xCT/GSH/GPX4, FSP1/CoQ10, DHODH/CoQ10, GCH1/BH4, and NRF2 pathways. Identifying novel anti-ferroptosis pathways will further promote our understanding of the biological nature of ferroptosis and help discover new drugs targeting ferroptosis related human diseases. In this study, we identified the clinically used drug mifepristone (RU486) as a novel ferroptosis inhibitor. Mechanistically, RU486 inhibits ferroptosis by inducing GSH synthesis pathway, which supplies GSH for glutathione-S-transferase (GST) mediated 4-HNE detoxification. Furthermore, RU486 induced RLIP76 and MRP1 export 4-HNE conjugate contributes to its anti-ferroptosis activity. Interestingly, RU486 induced GSH/GSTs/RLIP76&MRP1 anti-ferroptosis pathway acts independent of classic anti-ferroptosis systems: including xCT/GSH/GPX4, FSP1, DHODH, GCH1, SCD1 and FTH1. Moreover, NRF2 was identified to be important for RU486's anti-ferroptosis activity by inducing downstream gene expression. Importantly, in mouse model, RU486 showed strong protection effect on acetaminophen (APAP)-induced acute liver injury, evidenced by decreased ALT, AST level and histological recovery after APAP treatment. Interestingly, RU486 also decreased oxidative markers, including 4-HNE and MDA, and induced NRF2 activation as well as GSTs, MRP1 expression. Together, these data suggest NRF2/GSH/GST/RLIP76&MRP1 mediated detoxification pathway as an important independent anti-ferroptosis pathway act both in vitro and in vivo.
Collapse
Affiliation(s)
- Yanyun Shi
- GuiZhou University Medical College, Guiyang, 550025, China
| | - Nahua Xu
- Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Baiping Liu
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, 401120, China
| | - Yanni Ma
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xuemei Fu
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, 401120, China
| | - Yingying Shang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qilin Huang
- GuiZhou University Medical College, Guiyang, 550025, China; Department of Neurosurgery, Guiqian International General Hospital, Changpo Road, Wudang District, Guiyang, 550000, China.
| | - Qi Yao
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Hui Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, 401120, China; Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
8
|
Kline GM, Madrazo N, Cole CM, Pannikkat M, Bollong MJ, Rosarda JD, Kelly JW, Wiseman RL. Metabolically activated proteostasis regulators that protect against erastin-induced ferroptosis. RSC Chem Biol 2024; 5:866-876. [PMID: 39211477 PMCID: PMC11353103 DOI: 10.1039/d4cb00027g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
We previously showed that the proteostasis regulator compound AA147 (N-(2-hydroxy-5-methylphenyl)benzenepropanamide) potently protects against neurotoxic insults, such as glutamate-induced oxytosis. Though AA147 is a selective activator of the ATF6 arm of the unfolded protein response in non-neuronal cells, AA147-dependent protection against glutamate toxicity in cells of neuronal origin is primarily mediated through activation of the NRF2 oxidative stress response. AA147 activates NRF2 through a mechanism involving metabolic activation of AA147 by endoplasmic reticulum (ER) oxidases, affording an AA147-based quinone methide that covalently targets the NRF2 repressor protein KEAP1. Previous results show that the 2-amino-p-cresol A-ring of AA147 is required for NRF2 activation, while the phenyl B-ring of AA147 is amenable to modification. Here we explore whether the protease-sensitive amide linker between the A- and B-rings of this molecule can be modified to retain NRF2 activation. We show that replacement of the amide linker of AA147 with a carbamate linker retains NRF2 activation in neuronal cells and improves protection against neurotoxic insults, including glutamate-induced oxytosis and erastin-induced ferroptosis. Moreover, we demonstrate that inclusion of this carbamate linker facilitates identification of next-generation AA147 analogs with improved cellular tolerance and activity in disease-relevant assays.
Collapse
Affiliation(s)
- Gabriel M Kline
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Nicole Madrazo
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - Christian M Cole
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Meera Pannikkat
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - Michael J Bollong
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Jessica D Rosarda
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences Bethesda MD 20814 USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - R Luke Wiseman
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
| |
Collapse
|
9
|
Kong L, Liu Y, Li J, Wang Y, Ji P, Shi Q, Han M, Xu H, Li W, Li W. Ginsenoside Rg1 alleviates chronic inflammation-induced neuronal ferroptosis and cognitive impairments via regulation of AIM2 - Nrf2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118205. [PMID: 38641079 DOI: 10.1016/j.jep.2024.118205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng is a valuable herb in traditional Chinese medicine. Modern research has shown that it has various benefits, including tonifying vital energy, nourishing and strengthening the body, calming the mind, improving cognitive function, regulating fluids, and returning blood pressure, etc. Rg1 is a primary active component of ginseng. It protects hippocampal neurons, improves synaptic plasticity, enhances cognitive function, and boosts immunity. Furthermore, it exhibits anti-aging and anti-fatigue properties and holds great potential for preventing and managing neurodegenerative diseases (NDDs). AIM OF THE STUDY The objective of this study was to examine the role of Rg1 in treating chronic inflammatory NDDs and its molecular mechanisms. MATERIALS AND METHODS In vivo, we investigated the protective effects of Rg1 against chronic neuroinflammation and cognitive deficits in mice induced by 200 μg/kg lipopolysaccharide (LPS) for 21 days using behavioral tests, pathological sections, Western blot, qPCR and immunostaining. In vitro experiments involved the stimulation of HT22 cells with 10 μg/ml of LPS, verification of the therapeutic effect of Rg1, and elucidation of its potential mechanism of action using H2DCFDA staining, BODIPY™ 581/591 C11, JC-1 staining, Western blot, and immunostaining. RESULTS Firstly, it was found that Rg1 significantly improved chronic LPS-induced behavioral and cognitive dysfunction in mice. Further studies showed that Rg1 significantly attenuated LPS-induced neuronal damage by reducing levels of IL-6, IL-1β and ROS, and inhibiting AIM2 inflammasome. Furthermore, chronic LPS exposure induced the onset of neuronal ferroptosis by increasing the lipid peroxidation product MDA and regulating the ferroptosis-associated proteins Gpx4, xCT, FSP1, DMT1 and TfR, which were reversed by Rg1 treatment. Additionally, Rg1 was found to activate Nrf2 and its downstream antioxidant enzymes, such as HO1 and NQO1, both in vivo and in vitro. In vitro studies also showed that the Nrf2 inhibitor ML385 could inhibit the anti-inflammatory, antioxidant, and anti-ferroptosis effects of Rg1. CONCLUSIONS This study demonstrated that Rg1 administration ameliorated chronic LPS-induced cognitive deficits and neuronal ferroptosis in mice by inhibiting neuroinflammation and oxidative stress. The underlying mechanisms may be related to the inhibition of AIM2 inflammasome and activation of Nrf2 signaling. These findings provide valuable insights into the treatment of chronic neuroinflammation and associated NDDs.
Collapse
Affiliation(s)
- Liangliang Kong
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Yan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Jingwei Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Yanyan Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Pengmin Ji
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Qifeng Shi
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Min Han
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Hanyang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Weiping Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Weizu Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
10
|
Deng JL, Wang GY, Zhai YJ, Feng XY, Deng L, Han WB, Tang JJ. Herpotrichone A Exerts Neuroprotective Effects by Relieving Ferroptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:17356-17367. [PMID: 39042602 DOI: 10.1021/acs.jafc.4c02418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Inhibition of oxidative stress and ferroptosis is currently considered to be a promising therapeutic approach for neurodegenerative diseases. Herpotrichones, a class of compounds derived from insect symbionts, have shown potential for neuroprotective activity with low toxicity. However, the specific mechanisms through which herpotrichones exert their neuroprotective effects remain to be fully elucidated. In this study, the natural [4 + 2] adducts herpotrichone A (He-A) and its new analogues were isolated from the isopod-associated fungus Herpotrichia sp. SF09 and exhibited significantly protective effects in H2O2-, 6-OHDA-, and RSL3-stimulated PC12 cells and LPS-stimulated BV-2 cells. Moreover, He-A was able to relieve ferroptotic cell death in RSL3-stimulated PC12 cells and 6-OHDA-induced zebrafish larvae. Interestingly, He-A can activate antioxidant elements and modulate the SLC7A11 pathway without capturing oxidic free radical and chelating iron. These findings highlight He-A as a novel hit that protects against ferroptosis-like neuronal damage in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jia-Le Deng
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, No.3 Taicheng Road, Yangling, Shaanxi 712100, China
| | - Guo-Yan Wang
- College of Animal Science and Technology, Northwest A&F University, No.3 Taicheng Road, Yangling, Shaanxi 712100, China
| | - Yi-Jie Zhai
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, No.3 Taicheng Road, Yangling, Shaanxi 712100, China
| | - Xu-Yao Feng
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, No.3 Taicheng Road, Yangling, Shaanxi 712100, China
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, No.3 Taicheng Road, Yangling, Shaanxi 712100, China
| | - Wen-Bo Han
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, No.3 Taicheng Road, Yangling, Shaanxi 712100, China
| | - Jiang-Jiang Tang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, No.3 Taicheng Road, Yangling, Shaanxi 712100, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen Virtual University Park Building, High-Tech Industrial Park, Shenzhen, Guangdong 518000, China
| |
Collapse
|
11
|
Wang Y, Lan X, Liu N, Ma L, DU J, Wei W, Hai D, Wu J, Yu J, Liu Y. Traditional Chinese medicines derived natural inhibitors of ferroptosis on ischemic stroke. Chin J Nat Med 2024; 22:746-755. [PMID: 39197964 DOI: 10.1016/s1875-5364(24)60603-5] [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: 01/17/2024] [Indexed: 09/01/2024]
Abstract
Ischemic stroke (IS) is a globally prevalent cerebrovascular disorder resulting from cerebral vessel occlusion, leading to significant morbidity and mortality. The intricate pathological mechanisms underlying IS complicate the development of effective therapeutic interventions. Ferroptosis, a form of programmed cell death (PCD) characterized by iron overload and accumulation of lipid peroxidation products, has been increasingly recognized as a key contributor to IS pathology. Traditional Chinese medicines (TCMs) have long been utilized in the management of IS, prompting extensive research into their potential as sources of natural ferroptosis inhibitors. This review investigates the critical role of ferroptosis in IS and provides a comprehensive analysis of current research on natural ferroptosis inhibitors identified in TCMs, aiming to lay a theoretical groundwork for the development of innovative anti-IS therapies.
Collapse
Affiliation(s)
- Yongliang Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Xiaobing Lan
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Ning Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China; Ningxia Characteristic Traditional Chinese Medicine Modern Engineering Research Center, Ningxia Medical University, Yinchuan 750000, China
| | - Lin Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China; Ningxia Key Laboratory of Drug Development and Generic Drug Research, Ningxia Medical University, Yinchuan 750000, China
| | - Juan DU
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Wei Wei
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Dongmei Hai
- Ningxia Key Laboratory of Drug Development and Generic Drug Research, Ningxia Medical University, Yinchuan 750000, China; Ningxia Characteristic Traditional Chinese Medicine Modern Engineering Research Center, Ningxia Medical University, Yinchuan 750000, China
| | - Jing Wu
- Ningxia Key Laboratory of Drug Development and Generic Drug Research, Ningxia Medical University, Yinchuan 750000, China; College of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750000, China.
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China; Ningxia Key Laboratory of Drug Development and Generic Drug Research, Ningxia Medical University, Yinchuan 750000, China; Ningxia Characteristic Traditional Chinese Medicine Modern Engineering Research Center, Ningxia Medical University, Yinchuan 750000, China.
| | - Yue Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China.
| |
Collapse
|
12
|
Moorthy H, Ramesh M, Padhi D, Baruah P, Govindaraju T. Polycatechols inhibit ferroptosis and modulate tau liquid-liquid phase separation to mitigate Alzheimer's disease. MATERIALS HORIZONS 2024; 11:3082-3089. [PMID: 38647314 DOI: 10.1039/d4mh00023d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that affects learning, memory, and cognition. Current treatments targeting amyloid-β (Aβ) and tau have shown limited effectiveness, necessitating further research on the aggregation and toxicity mechanisms. One of these mechanisms involves the liquid-liquid phase separation (LLPS) of tau, contributing to the formation of pathogenic tau aggregates, although their conformational details remain elusive. Another mechanism is ferroptosis, a type of iron-dependent lipid peroxidation-mediated cell death, which has been implicated in AD. There is a lack of therapeutic strategies that simultaneously target amyloid toxicity and ferroptosis. This study aims to explore the potential of polycatechols, PDP and PLDP, consisting of dopamine and L-Dopa, respectively, as multifunctional agents to modulate the pathological nexus between ferroptosis and AD. Polycatechols were found to sequester the labile iron pool (LIP), inhibit Aβ and tau aggregation, scavenge free radicals, protect mitochondria, and prevent ferroptosis, thereby rescuing neuronal cell death. Interestingly, PLDP promotes tau LLPS, and modulates their intermolecular interactions to inhibit the formation of toxic tau aggregates, offering a conceptually innovative approach to tackle tauopathies. This is a first-of-its-kind polymer-based integrative approach that inhibits ferroptosis, counteracts amyloid toxicity, and modulates tau LLPS to mitigate the multifaceted toxicity of AD.
Collapse
Affiliation(s)
- Hariharan Moorthy
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India.
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India.
| | - Dikshaa Padhi
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India.
| | - Prayasee Baruah
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India.
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India.
| |
Collapse
|
13
|
Lyamzaev KG, Huan H, Panteleeva AA, Simonyan RA, Avetisyan AV, Chernyak BV. Exogenous Iron Induces Mitochondrial Lipid Peroxidation, Lipofuscin Accumulation, and Ferroptosis in H9c2 Cardiomyocytes. Biomolecules 2024; 14:730. [PMID: 38927133 PMCID: PMC11201805 DOI: 10.3390/biom14060730] [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: 04/25/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Lipid peroxidation plays an important role in various pathologies and aging, at least partially mediated by ferroptosis. The role of mitochondrial lipid peroxidation during ferroptosis remains poorly understood. We show that supplementation of exogenous iron in the form of ferric ammonium citrate at submillimolar doses induces production of reactive oxygen species (ROS) and lipid peroxidation in mitochondria that precede ferroptosis in H9c2 cardiomyocytes. The mitochondria-targeted antioxidant SkQ1 and the redox mediator methylene blue, which inhibits the production of ROS in complex I of the mitochondrial electron transport chain, prevent both mitochondrial lipid peroxidation and ferroptosis. SkQ1 and methylene blue also prevented accumulation of lipofuscin observed after 24 h incubation of cardiomyocytes with ferric ammonium citrate. Using isolated cardiac mitochondria as an in vitro ferroptosis model, it was shown that rotenone (complex I inhibitor) in the presence of ferrous iron stimulates lipid peroxidation and lipofuscin accumulation. Our data indicate that ROS generated in complex I stimulate mitochondrial lipid peroxidation, lipofuscin accumulation, and ferroptosis induced by exogenous iron.
Collapse
Affiliation(s)
- Konstantin G. Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
- The Russian Clinical Research Center for Gerontology, Ministry of Healthcare of the Russian Federation, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - He Huan
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Alisa A. Panteleeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Ruben A. Simonyan
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Armine V. Avetisyan
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| | - Boris V. Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (H.H.); (A.A.P.); (R.A.S.); (A.V.A.)
| |
Collapse
|
14
|
Deng L, Tian W, Luo L. Application of natural products in regulating ferroptosis in human diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155384. [PMID: 38547620 DOI: 10.1016/j.phymed.2024.155384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/05/2024] [Accepted: 01/23/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Ferroptosis is a type of cell death caused by excessive iron-induced peroxidation. It has been found to be involved in a variety of diseases, and natural products can be used to target ferroptosis in treatments. Natural products are biologically active compounds extracted or synthesized from nature. It is an important resource for the discovery of skeletons with a high degree of structural diversity and a wide range of bioactivities, which can be developed directly or used as a starting point for the optimization of new drugs. PURPOSE In this review, we aim to discuss the interactions between natural products and ferroptosis in the treatment of human diseases. METHODS Literature was searched in Pubmed, Science Direct, and Web of Science databases for the 11-year period from 2012 to 2023 using the search terms "natural products", "ferroptosis", "human disease", "neurodegenerative disease", "cardiovascular disease", and "cancer". RESULTS In this research, the roles of natural products and ferroptosis were investigated. We suggest that natural products, such as terpenoids, flavonoids, polyphenols, alkaloids, and saponins, can be used in therapeutic applications for human diseases, as well as in ferroptosis. Additionally, the main mechanisms of ferroptosis were summarized and discussed. Furthermore, we propose that natural products can be utilized to enhance the sensitivity of cancer cells to ferroptosis, thus helping to overcome drug resistance and inhibit metastasis. Moreover, natural products have the potential to modulate the expression levels of ferroptosis-related factors. Finally, the future directions of this field were highlighted. CONCLUSION The potential of natural products which focus on ferroptosis to treat human illnesses, particularly cancer, is very encouraging for human wellbeing.
Collapse
Affiliation(s)
- Liyan Deng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Wen Tian
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, Guangdong, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, Guangdong, China
| |
Collapse
|
15
|
Bell HN, Stockwell BR, Zou W. Ironing out the role of ferroptosis in immunity. Immunity 2024; 57:941-956. [PMID: 38749397 PMCID: PMC11101142 DOI: 10.1016/j.immuni.2024.03.019] [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: 01/04/2024] [Revised: 02/20/2024] [Accepted: 03/26/2024] [Indexed: 05/19/2024]
Abstract
Ferroptosis is a type of regulated cell death that drives the pathophysiology of many diseases. Oxidative stress is detectable in many types of regulated cell death, but only ferroptosis involves lipid peroxidation and iron dependency. Ferroptosis originates and propagates from several organelles, including the mitochondria, endoplasmic reticulum, Golgi, and lysosomes. Recent data have revealed that immune cells can both induce and undergo ferroptosis. A mechanistic understanding of how ferroptosis regulates immunity is critical to understanding how ferroptosis controls immune responses and how this is dysregulated in disease. Translationally, more work is needed to produce ferroptosis-modulating immunotherapeutics. This review focuses on the role of ferroptosis in immune-related diseases, including infection, autoimmune diseases, and cancer. We discuss how ferroptosis is regulated in immunity, how this regulation contributes to disease pathogenesis, and how targeting ferroptosis may lead to novel therapies.
Collapse
Affiliation(s)
- Hannah N Bell
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan School of Medicine, Rogel Cancer Center, Ann Arbor, MI, USA; Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA.
| | - Brent R Stockwell
- Department of Biological Sciences, Department of Chemistry, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan School of Medicine, Rogel Cancer Center, Ann Arbor, MI, USA; Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Abstract
Regulated cell death mediated by dedicated molecular machines, known as programmed cell death, plays important roles in health and disease. Apoptosis, necroptosis and pyroptosis are three such programmed cell death modalities. The caspase family of cysteine proteases serve as key regulators of programmed cell death. During apoptosis, a cascade of caspase activation mediates signal transduction and cellular destruction, whereas pyroptosis occurs when activated caspases cleave gasdermins, which can then form pores in the plasma membrane. Necroptosis, a form of caspase-independent programmed necrosis mediated by RIPK3 and MLKL, is inhibited by caspase-8-mediated cleavage of RIPK1. Disruption of cellular homeostatic mechanisms that are essential for cell survival, such as normal ionic and redox balance and lysosomal flux, can also induce cell death without invoking programmed cell death mechanisms. Excitotoxicity, ferroptosis and lysosomal cell death are examples of such cell death modes. In this Review, we provide an overview of the major cell death mechanisms, highlighting the latest insights into their complex regulation and execution, and their relevance to human diseases.
Collapse
Affiliation(s)
- Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| | - Dimitry Ofengeim
- Sanofi, Rare and Neurological Diseases Research, Cambridge, MA, USA.
| |
Collapse
|
17
|
Yang W, Wang Y, Li H, Liao F, Peng Y, Lu A, Tan L, Qu H, Long L, Fu C. Enhanced TfR1 Recognition of Myocardial Injury after Acute Myocardial Infarction with Cardiac Fibrosis via Pre-Degrading Excess Fibrotic Collagen. BIOLOGY 2024; 13:213. [PMID: 38666825 PMCID: PMC11048469 DOI: 10.3390/biology13040213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024]
Abstract
The fibrosis process after myocardial infarction (MI) results in a decline in cardiac function due to fibrotic collagen deposition and contrast agents' metabolic disorders, posing a significant challenge to conventional imaging strategies in making heart damage clear in the fibrosis microenvironment. To address this issue, we developed an imaging strategy. Specifically, we pretreated myocardial fibrotic collagen with collagenase I combined with human serum albumin (HSA-C) and subsequently visualized the site of cardiac injury by near-infrared (NIR) fluorescence imaging using an optical contrast agent (CI, CRT-indocyanine green) targeting transferrin receptor 1 peptides (CRT). The key point of this strategy is that pretreatment with HSA-C can reduce background signal interference in the fibrotic tissue while enhancing CI uptake at the heart lesion site, making the boundary between the injured heart tissue and the normal myocardium clearer. Our results showed that compared to that in the untargeted group, the normalized fluorescence intensity of cardiac damage detected by NIR in the targeted group increased 1.28-fold. The normalized fluorescence intensity increased 1.21-fold in the pretreatment group of the targeted groups. These data demonstrate the feasibility of applying pretreated fibrotic collagen and NIR contrast agents targeting TfR1 to identify ferroptosis at sites of cardiac injury, and its clinical value in the management of patients with MI needs further study.
Collapse
Affiliation(s)
- Wenwen Yang
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yueqi Wang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Hongzheng Li
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Feifei Liao
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuxuan Peng
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Aimei Lu
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Ling Tan
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Hua Qu
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Linzi Long
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Changgeng Fu
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
18
|
Lin S, Zheng Y, Chen M, Xu L, Huang H. The interactions between ineffective erythropoiesis and ferroptosis in β-thalassemia. Front Physiol 2024; 15:1346173. [PMID: 38468700 PMCID: PMC10925657 DOI: 10.3389/fphys.2024.1346173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
In Guangxi, Hainan, and Fujian Province in southern China, β-thalassemia is a frequent monogenic hereditary disorder that is primarily defined by hemolytic anemia brought on by inefficient erythropoiesis. It has been found that ineffective erythropoiesis in β-thalassemia is closely associated with a high accumulation of Reactive oxygen species, a product of oxidative stress, in erythroid cells. During recent years, ferroptosis is an iron-dependent lipid peroxidation that involves abnormalities in lipid and iron metabolism as well as reactive oxygen species homeostasis. It is a recently identified kind of programmed cell death. β-thalassemia patients experience increased iron release from reticuloendothelial cells and intestinal absorption of iron, ultimately resulting in iron overload. Additionally, the secretion of Hepcidin is inhibited in these patients. What counts is both ineffective erythropoiesis and ferroptosis in β-thalassemia are intricately linked to the iron metabolism and Reactive oxygen species homeostasis. Consequently, to shed further light on the pathophysiology of β-thalassemia and propose fresh ideas for its therapy, this paper reviews ferroptosis, ineffective erythropoiesis, and the way they interact.
Collapse
Affiliation(s)
- Siyang Lin
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
| | - Yanping Zheng
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Meihuan Chen
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| | - Liangpu Xu
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| | - Hailong Huang
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| |
Collapse
|
19
|
Carvalho FV, Landis HE, Getachew B, Silva VDA, Ribeiro PR, Aschner M, Tizabi Y. Iron toxicity, ferroptosis and microbiota in Parkinson's disease: Implications for novel targets. ADVANCES IN NEUROTOXICOLOGY 2024; 11:105-132. [PMID: 38770370 PMCID: PMC11105119 DOI: 10.1016/bs.ant.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Parkinson's Disease (PD) is a progressive neurodegenerative disease characterized by loss of dopaminergic neurons in substantia nigra pars compacta (SNpc). Iron (Fe)-dependent programmed cell death known as ferroptosis, plays a crucial role in the etiology and progression of PD. Since SNpc is particularly vulnerable to Fe toxicity, a central role for ferroptosis in the etiology and progression of PD is envisioned. Ferroptosis, characterized by reactive oxygen species (ROS)-dependent accumulation of lipid peroxides, is tightly regulated by a variety of intracellular metabolic processes. Moreover, the recently characterized bi-directional interactions between ferroptosis and the gut microbiota, not only provides another window into the mechanistic underpinnings of PD but could also suggest novel interventions in this devastating disease. Here, following a brief discussion of PD, we focus on how our expanding knowledge of Fe-induced ferroptosis and its interaction with the gut microbiota may contribute to the pathophysiology of PD and how this knowledge may be exploited to provide novel interventions in PD.
Collapse
Affiliation(s)
| | - Harold E. Landis
- Integrative Medicine Fellow, University of Arizona, Tucson, AZ, United States
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | | | - Paulo R. Ribeiro
- Metabolomics Research Group, Institute of Chemistry, Federal University of Bahia, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
20
|
Campos J, Gleitze S, Hidalgo C, Núñez MT. IP 3R-Mediated Calcium Release Promotes Ferroptotic Death in SH-SY5Y Neuroblastoma Cells. Antioxidants (Basel) 2024; 13:196. [PMID: 38397794 PMCID: PMC10886377 DOI: 10.3390/antiox13020196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Ferroptosis is an iron-dependent cell death pathway that involves the depletion of intracellular glutathione (GSH) levels and iron-mediated lipid peroxidation. Ferroptosis is experimentally caused by the inhibition of the cystine/glutamate antiporter xCT, which depletes cells of GSH, or by inhibition of glutathione peroxidase 4 (GPx4), a key regulator of lipid peroxidation. The events that occur between GPx4 inhibition and the execution of ferroptotic cell death are currently a matter of active research. Previous work has shown that calcium release from the endoplasmic reticulum (ER) mediated by ryanodine receptor (RyR) channels contributes to ferroptosis-induced cell death in primary hippocampal neurons. Here, we used SH-SY5Y neuroblastoma cells, which do not express RyR channels, to test if calcium release mediated by the inositol 1,4,5-trisphosphate receptor (IP3R) channel plays a role in this process. We show that treatment with RAS Selective Lethal Compound 3 (RSL3), a GPx4 inhibitor, enhanced reactive oxygen species (ROS) generation, increased cytoplasmic and mitochondrial calcium levels, increased lipid peroxidation, and caused cell death. The RSL3-induced calcium signals were inhibited by Xestospongin B, a specific inhibitor of the ER-resident IP3R calcium channel, by decreasing IP3R levels with carbachol and by IP3R1 knockdown, which also prevented the changes in cell morphology toward roundness induced by RSL3. Intracellular calcium chelation by incubation with BAPTA-AM inhibited RSL3-induced calcium signals, which were not affected by extracellular calcium depletion. We propose that GPx4 inhibition activates IP3R-mediated calcium release in SH-SY5Y cells, leading to increased cytoplasmic and mitochondrial calcium levels, which, in turn, stimulate ROS production and induce lipid peroxidation and cell death in a noxious positive feedback cycle.
Collapse
Affiliation(s)
- Joaquín Campos
- Chica and Heinz Schaller Foundation, Institute for Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Silvia Gleitze
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (S.G.); (C.H.)
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (S.G.); (C.H.)
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
- Physiology and Biophysics Program, Institute of Biomedical Sciences and Center for Exercise, Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
| | - Marco T. Núñez
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago 7800024, Chile
| |
Collapse
|
21
|
Claudio-Ares O, Luciano-Rodríguez J, Del Valle-González YL, Schiavone-Chamorro SL, Pastor AJ, Rivera-Reyes JO, Metzler CL, Domínguez-Orona LM, Vargas-Pérez BL, Skouta R, Tinoco AD. Exploring the Use of Intracellular Chelation and Non-Iron Metals to Program Ferroptosis for Anticancer Application. INORGANICS 2024; 12:26. [PMID: 39380574 PMCID: PMC11460773 DOI: 10.3390/inorganics12010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
The discovery of regulated cell death (RCD) revolutionized chemotherapy. With caspase-dependent apoptosis initially being thought to be the only form of RCD, many drug development strategies aimed to synthesize compounds that turn on this kind of cell death. While yielding a variety of drugs, this approach is limited, given the acquired resistance of cancers to these drugs and the lack of specificity of the drugs for targeting cancer cells alone. The discovery of non-apoptotic forms of RCD is leading to new avenues for drug design. Evidence shows that ferroptosis, a relatively recently discovered iron-based cell death pathway, has therapeutic potential for anticancer application. Recent studies point to the interrelationship between iron and other essential metals, copper and zinc, and the disturbance of their respective homeostasis as critical to the onset of ferroptosis. Other studies reveal that several coordination complexes of non-iron metals have the capacity to induce ferroptosis. This collective knowledge will be assessed to determine how chelation approaches and coordination chemistry can be engineered to program ferroptosis in chemotherapy.
Collapse
Affiliation(s)
- Oscar Claudio-Ares
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | | | | | | | - Alex J. Pastor
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | - Javier O. Rivera-Reyes
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | - Carmen L. Metzler
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | | | - Brenda Lee Vargas-Pérez
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | - Rachid Skouta
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003-9248, USA
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003-9248, USA
| | - Arthur D. Tinoco
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| |
Collapse
|
22
|
Marmolejo-Garza A, Krabbendam IE, Luu MDA, Brouwer F, Trombetta-Lima M, Unal O, O'Connor SJ, Majerníková N, Elzinga CRS, Mammucari C, Schmidt M, Madesh M, Boddeke E, Dolga AM. Negative modulation of mitochondrial calcium uniporter complex protects neurons against ferroptosis. Cell Death Dis 2023; 14:772. [PMID: 38007529 PMCID: PMC10676387 DOI: 10.1038/s41419-023-06290-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
Ferroptosis is an iron- and reactive oxygen species (ROS)-dependent form of regulated cell death, that has been implicated in Alzheimer's disease and Parkinson's disease. Inhibition of cystine/glutamate antiporter could lead to mitochondrial fragmentation, mitochondrial calcium ([Ca2+]m) overload, increased mitochondrial ROS production, disruption of the mitochondrial membrane potential (ΔΨm), and ferroptotic cell death. The observation that mitochondrial dysfunction is a characteristic of ferroptosis makes preservation of mitochondrial function a potential therapeutic option for diseases associated with ferroptotic cell death. Mitochondrial calcium levels are controlled via the mitochondrial calcium uniporter (MCU), the main entry point of Ca2+ into the mitochondrial matrix. Therefore, we have hypothesized that negative modulation of MCU complex may confer protection against ferroptosis. Here we evaluated whether the known negative modulators of MCU complex, ruthenium red (RR), its derivative Ru265, mitoxantrone (MX), and MCU-i4 can prevent mitochondrial dysfunction and ferroptotic cell death. These compounds mediated protection in HT22 cells, in human dopaminergic neurons and mouse primary cortical neurons against ferroptotic cell death. Depletion of MICU1, a [Ca2+]m gatekeeper, demonstrated that MICU is protective against ferroptosis. Taken together, our results reveal that negative modulation of MCU complex represents a therapeutic option to prevent degenerative conditions, in which ferroptosis is central to the progression of these pathologies.
Collapse
Affiliation(s)
- Alejandro Marmolejo-Garza
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge E Krabbendam
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Minh Danh Anh Luu
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Famke Brouwer
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Osman Unal
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Shane J O'Connor
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Naďa Majerníková
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Carolina R S Elzinga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy
| | - Martina Schmidt
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Muniswamy Madesh
- Department of Medicine/Cardiology, Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Erik Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
23
|
Fahrer J, Wittmann S, Wolf AC, Kostka T. Heme Oxygenase-1 and Its Role in Colorectal Cancer. Antioxidants (Basel) 2023; 12:1989. [PMID: 38001842 PMCID: PMC10669411 DOI: 10.3390/antiox12111989] [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: 09/15/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an enzyme located at the endoplasmic reticulum, which is responsible for the degradation of cellular heme into ferrous iron, carbon monoxide and biliverdin-IXa. In addition to this main function, the enzyme is involved in many other homeostatic, toxic and cancer-related mechanisms. In this review, we first summarize the importance of HO-1 in physiology and pathophysiology with a focus on the digestive system. We then detail its structure and function, followed by a section on the regulatory mechanisms that control HO-1 expression and activity. Moreover, HO-2 as important further HO isoform is discussed, highlighting the similarities and differences with regard to HO-1. Subsequently, we describe the direct and indirect cytoprotective functions of HO-1 and its breakdown products carbon monoxide and biliverdin-IXa, but also highlight possible pro-inflammatory effects. Finally, we address the role of HO-1 in cancer with a particular focus on colorectal cancer. Here, relevant pathways and mechanisms are presented, through which HO-1 impacts tumor induction and tumor progression. These include oxidative stress and DNA damage, ferroptosis, cell cycle progression and apoptosis as well as migration, proliferation, and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Jörg Fahrer
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, Erwin-Schrödinger Strasse 52, D-67663 Kaiserslautern, Germany; (S.W.); (A.-C.W.)
| | | | | | - Tina Kostka
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, Erwin-Schrödinger Strasse 52, D-67663 Kaiserslautern, Germany; (S.W.); (A.-C.W.)
| |
Collapse
|