1
|
Kacemi R, Campos MG. Bee Pollen Potential to Modulate Ferroptosis: Phytochemical Insights for Age-Related Diseases. Antioxidants (Basel) 2025; 14:265. [PMID: 40227202 PMCID: PMC11939620 DOI: 10.3390/antiox14030265] [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: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 04/15/2025] Open
Abstract
Bee pollen (BP) is one of the richest known natural resources of micronutrients and bioactive phytochemicals. Some captivating bioactivities of BP compounds, although being largely investigated for the latter as individual molecules, remain very scarcely investigated or completely uninvestigated in bee pollen as a whole product. Among the most intriguing of these bioactivities, we identified ferroptosis as a major one. Ferroptosis, a recently discovered form of cell death (connecting oxidative stress and inflammation), is a complex pathophysiological process and one of the most crucial and perplexing events in current challenging human diseases such as cancer, neurodegeneration, and general aging diseases. Many BP compounds were found to intricately modulate ferroptosis depending on the cellular context by inducing this cell death mechanism in malignant cells and preventing it in non-malignant cells. Since research in both fields, i.e., BP and ferroptosis, is still recent, we deemed it necessary to undertake this review to figure out the extent of BP potential in modulating ferroptosis mechanisms. Our research proved that a wide range of BP compounds (polyphenols, phenolamides, carotenoids, vitamins, minerals, and others) substantially modulate diverse ferroptosis mechanisms. Accordingly, these phytochemicals and nutrients showed interesting potential in preclinical studies to lead to ferroptosis-mediated outcomes in important pathophysiological processes, including many aging-related disorders. One of the most paramount challenges that remain to be resolved is to determine how different BP compounds act on ferroptosis in different biological and pathophysiological contexts, either through synergistic or antagonistic behaviors. We hope that our current work constitutes a valuable incentive for future investigations in this promising and very relevant research avenue.
Collapse
Affiliation(s)
- Rachid Kacemi
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria G. Campos
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra Chemistry Centre (CQC, FCT Unit 313) (FCTUC), University of Coimbra, Rua Larga, 3000-548 Coimbra, Portugal
| |
Collapse
|
2
|
Urano Y, Iwagaki A, Takeishi A, Uchiyama N, Noguchi N. Downregulation of the SREBP pathways and disruption of redox status by 25-hydroxycholesterol predispose cells to ferroptosis. Free Radic Biol Med 2025; 228:319-328. [PMID: 39778605 DOI: 10.1016/j.freeradbiomed.2025.01.010] [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: 11/22/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Enzymatically formed side-chain oxysterols function as signaling molecules regulating cholesterol homeostasis and act as intermediates in the biosynthesis of bile acids. In addition to these physiological functions, an imbalance in oxysterol homeostasis has been implicated in pathophysiology. Cholesterol 25-hydroxylase (CH25H) and its product 25-hydroxycholesterol (25-OHC), also formed by autoxidation, are associated with amyotrophic lateral sclerosis. However, the effects of 25-OHC on cell viability in glial cells remain unclear. This study demonstrates that 25-OHC induces ferroptosis, an iron-dependent programmed cell death, in mouse Schwann IMS32 cells. Mechanistically, 25-OHC suppressed the expression of selenoprotein glutathione peroxidase 4 (GPX4) at both the transcriptional and translational levels by inhibiting the processing of sterol regulatory element-binding proteins (SREBPs). In addition, 25-OHC upregulated the expression of NADH-cytochrome b5 reductase 1 (CYB5R1) and NADPH-cytochrome P450 reductase (POR), enzymes that promote lipid peroxidation. We further found that 25-OHC increases the expression of glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) and decreases glutathione levels. Importantly, non-cytotoxic concentrations of 25-OHC enhanced cellular sensitivity to ferroptosis inducers by downregulating GPX4 expression. These findings reveal a multifaceted approach whereby 25-OHC induces ferroptosis through SREBP pathway suppression and redox imbalance in mouse Schwann IMS32 cells.
Collapse
Affiliation(s)
- Yasuomi Urano
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan.
| | - Anan Iwagaki
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan
| | - Arisa Takeishi
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan
| | - Nazuna Uchiyama
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan
| | - Noriko Noguchi
- Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe, Kyoto, 610-0394, Japan.
| |
Collapse
|
3
|
Cuffaro F, Lamminpää I, Niccolai E, Amedei A. Nutritional and Microbiota-Based Approaches in Amyotrophic Lateral Sclerosis: From Prevention to Treatment. Nutrients 2024; 17:102. [PMID: 39796536 PMCID: PMC11722677 DOI: 10.3390/nu17010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
Metabolic alterations, including hypermetabolism, lipid imbalances, and glucose dysregulation, are pivotal contributors to the onset and progression of Amyotrophic Lateral Sclerosis (ALS). These changes exacerbate systemic energy deficits, heighten oxidative stress, and fuel neuroinflammation. Simultaneously, gastrointestinal dysfunction and gut microbiota (GM) dysbiosis intensify disease pathology by driving immune dysregulation, compromising the intestinal barrier, and altering gut-brain axis (GBA) signaling, and lastly advancing neurodegeneration. Therapeutic and preventive strategies focused on nutrition offer promising opportunities to address these interconnected pathophysiological mechanisms. Diets enriched with antioxidants, omega-3 fatty acids, and anti-inflammatory compounds-such as the Mediterranean diet-have shown potential in reducing oxidative stress and systemic inflammation. Additionally, microbiota-targeted approaches, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation, are emerging as innovative tools to restore microbial balance, strengthen gut integrity, and optimize GBA function. This review highlights the critical need for personalized strategies integrating immunonutrition and microbiota modulation to slow ALS progression, improve quality of life, and develop preventive measures for neurodegenerative and neuroinflammatory diseases. Future research should prioritize comprehensive dietary and microbiota-based interventions to uncover their therapeutic potential and establish evidence-based guidelines for managing ALS and related disorders.
Collapse
Affiliation(s)
- Francesca Cuffaro
- Division of Interdisciplinary Internal Medicine, Careggi University Hospital of Florence, 50134 Florence, Italy;
| | - Ingrid Lamminpää
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (I.L.); (A.A.)
| | - Elena Niccolai
- Division of Interdisciplinary Internal Medicine, Careggi University Hospital of Florence, 50134 Florence, Italy;
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (I.L.); (A.A.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (I.L.); (A.A.)
- Laboratorio Congiunto MIA-LAB (Microbiome-Immunity Axis Research for a Circular Health), University of Florence, 50134 Firenze, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50139 Florence, Italy
| |
Collapse
|
4
|
Kacemi R, Campos MG. Bee Pollen as a Source of Biopharmaceuticals for Neurodegeneration and Cancer Research: A Scoping Review and Translational Prospects. Molecules 2024; 29:5893. [PMID: 39769981 PMCID: PMC11677910 DOI: 10.3390/molecules29245893] [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: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Bee Pollen (BP) has many advantageous properties relying on its multitargeting potential, a new tendency in managing many challenging illnesses. In cancer and neurodegeneration, the multiple effects of BP could be of unequaled importance and need further investigation. Although still limited, available data interestingly spotlights some floral sources with promising activities in line with this investigation. Adopting scoping review methodology, we have identified many crucial bioactivities that are widely recognized to individual BP compounds but remain completely untapped in this valuable bee cocktail. A wide range of these compounds have been recently found to be endowed with great potential in modulating pivotal processes in neurodegeneration and cancer pathophysiology. In addition, some ubiquitous BP compounds have only been recently isolated, while the number of studied BPs remains extremely limited compared to the endless pool of plant species worldwide. We have also elucidated that clinical profits from these promising perspectives are still impeded by challenging hurdles such as limited bioavailability of the studied phytocompounds, diversity and lack of phytochemical standardization of BP, and the difficulty of selective targeting in some pathophysiological mechanisms. We finally present interesting insights to guide future research and pave the way for urgently needed and simplified clinical investigations.
Collapse
Affiliation(s)
- Rachid Kacemi
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
| | - Maria G. Campos
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- Coimbra Chemistry Centre (CQC, FCT Unit 313) (FCTUC), University of Coimbra, Rua Larga, 3004-531 Coimbra, Portugal
| |
Collapse
|
5
|
Papadimitriou‐Tsantarliotou A, Avgeros C, Konstantinidou M, Vizirianakis IS. Analyzing the role of ferroptosis in ribosome-related bone marrow failure disorders: From pathophysiology to potential pharmacological exploitation. IUBMB Life 2024; 76:1011-1034. [PMID: 39052023 PMCID: PMC11580388 DOI: 10.1002/iub.2897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/04/2024] [Indexed: 07/27/2024]
Abstract
Within the last decade, the scientific community has witnessed the importance of ferroptosis as a novel cascade of molecular events leading to cellular decisions of death distinct from apoptosis and other known forms of cell death. Notably, such non- apoptotic and iron-dependent regulated cell death has been found to be intricately linked to several physiological processes as well as to the pathogenesis of various diseases. To this end, recent data support the notion that a potential molecular connection between ferroptosis and inherited bone marrow failure (IBMF) in individuals with ribosomopathies may exist. In this review, we suggest that in ribosome-related IBMFs the identified mutations in ribosomal proteins lead to changes in the ribosome composition of the hematopoietic progenitors, changes that seem to affect ribosomal function, thus enhancing the expression of some mRNAs subgroups while reducing the expression of others. These events lead to an imbalance inside the cell as some molecular pathways are promoted while others are inhibited. This disturbance is accompanied by ROS production and lipid peroxidation, while an additional finding in most of them is iron accumulation. Once lipid peroxidation and iron accumulation are the two main characteristics of ferroptosis, it is possible that this mechanism plays a key role in the manifestation of IBMF in this type of disease. If this molecular mechanism is further confirmed, new pharmacological targets such as ferroptosis inhibitors that are already exploited for the treatment of other diseases, could be utilized to improve the treatment of ribosomopathies.
Collapse
Affiliation(s)
| | - Chrysostomos Avgeros
- Laboratory of Pharmacology, School of PharmacyAristotle University of ThessalonikiThessalonikiGreece
| | - Maria Konstantinidou
- Laboratory of Pharmacology, School of PharmacyAristotle University of ThessalonikiThessalonikiGreece
| | - Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of PharmacyAristotle University of ThessalonikiThessalonikiGreece
- Department of Health Sciences, School of Life and Health SciencesUniversity of NicosiaNicosiaCyprus
| |
Collapse
|
6
|
Hayakawa-Yano Y, Furukawa T, Matsuo T, Ogasawara T, Nogami M, Yokoyama K, Yugami M, Shinozaki M, Nakamoto C, Sakimura K, Koyama A, Ogi K, Onodera O, Takebayashi H, Okano H, Yano M. Qki5 safeguards spinal motor neuron function by defining the motor neuron-specific transcriptome via pre-mRNA processing. Proc Natl Acad Sci U S A 2024; 121:e2401531121. [PMID: 39226364 PMCID: PMC11406248 DOI: 10.1073/pnas.2401531121] [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: 01/24/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Many RNA-binding proteins (RBPs) are linked to the dysregulation of RNA metabolism in motor neuron diseases (MNDs). However, the molecular mechanisms underlying MN vulnerability have yet to be elucidated. Here, we found that such an RBP, Quaking5 (Qki5), contributes to formation of the MN-specific transcriptome profile, termed "MN-ness," through the posttranscriptional network and maintenance of the mature MNs. Immunohistochemical analysis and single-cell RNA sequencing (scRNA-seq) revealed that Qki5 is predominantly expressed in MNs, but not in other neuronal populations of the spinal cord. Furthermore, comprehensive RNA sequencing (RNA-seq) analyses revealed that Qki5-dependent RNA regulation plays a pivotal role in generating the MN-specific transcriptome through pre-messenger ribonucleic acid (mRNA) splicing for the synapse-related molecules and c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) signaling pathways. Indeed, MN-specific ablation of the Qki5 caused neurodegeneration in postnatal mice and loss of Qki5 function resulted in the aberrant activation of stress-responsive JNK/SAPK pathway both in vitro and in vivo. These data suggested that Qki5 plays a crucial biological role in RNA regulation and safeguarding of MNs and might be associated with pathogenesis of MNDs.
Collapse
Affiliation(s)
- Yoshika Hayakawa-Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
- Keio University Regenerative Medicine Research Center, Kawasaki, Kanagawa 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takako Furukawa
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Tsuyoshi Matsuo
- The Shonan Incubation Laboratory, Shonan Research Center, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Takahisa Ogasawara
- The Shonan Incubation Laboratory, Shonan Research Center, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Masahiro Nogami
- The Shonan Incubation Laboratory, Shonan Research Center, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazumasa Yokoyama
- The Shonan Incubation Laboratory, Shonan Research Center, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Masato Yugami
- The Shonan Incubation Laboratory, Shonan Research Center, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Munehisa Shinozaki
- Keio University Regenerative Medicine Research Center, Kawasaki, Kanagawa 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Chihiro Nakamoto
- Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Akihide Koyama
- Division of Legal Medicine, Department of Community Preventive Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Kazuhiro Ogi
- The Shonan Incubation Laboratory, Shonan Research Center, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8585, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Hideyuki Okano
- Keio University Regenerative Medicine Research Center, Kawasaki, Kanagawa 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masato Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
- Keio University Regenerative Medicine Research Center, Kawasaki, Kanagawa 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
7
|
Pinilla-González V, Montecinos-Barrientos B, Martin-Kommer C, Chichiarelli S, Saso L, Rodrigo R. Exploring antioxidant strategies in the pathogenesis of ALS. Open Life Sci 2024; 19:20220842. [PMID: 38585631 PMCID: PMC10997151 DOI: 10.1515/biol-2022-0842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 04/09/2024] Open
Abstract
The central nervous system is essential for maintaining homeostasis and controlling the body's physiological functions. However, its biochemical characteristics make it highly vulnerable to oxidative damage, which is a common factor in neurodegenerative diseases like amyotrophic lateral sclerosis (ALS). ALS is a leading cause of motor neuron disease, characterized by a rapidly progressing and incurable condition. ALS often results in death from respiratory failure within 3-5 years from the onset of the first symptoms, underscoring the urgent need to address this medical challenge. The aim of this study is to present available data supporting the role of oxidative stress in the mechanisms underlying ALS and to discuss potential antioxidant therapies currently in development. These therapies aim to improve the quality of life and life expectancy for patients affected by this devastating disease.
Collapse
Affiliation(s)
- Víctor Pinilla-González
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago8380000, Chile
| | | | - Clemente Martin-Kommer
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago8380000, Chile
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, P.le Aldo Moro 5, 00185Rome, Italy
| | - Ramón Rodrigo
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago8380000, Chile
| |
Collapse
|
8
|
Dar NJ, John U, Bano N, Khan S, Bhat SA. Oxytosis/Ferroptosis in Neurodegeneration: the Underlying Role of Master Regulator Glutathione Peroxidase 4 (GPX4). Mol Neurobiol 2024; 61:1507-1526. [PMID: 37725216 DOI: 10.1007/s12035-023-03646-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023]
Abstract
Oxytosis/ferroptosis is an iron-dependent oxidative form of cell death triggered by lethal accumulation of phospholipid hydroperoxides (PLOOHs) in membranes. Failure of the intricate PLOOH repair system is a principle cause of ferroptotic cell death. Glutathione peroxidase 4 (GPX4) is distinctly vital for converting PLOOHs in membranes to non-toxic alcohols. As such, GPX4 is known as the master regulator of oxytosis/ferroptosis. Ferroptosis has been implicated in a number of disorders such as neurodegenerative diseases (amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), etc.), ischemia/reperfusion injury, and kidney degeneration. Reduced function of GPX4 is frequently observed in degenerative disorders. In this study, we examine how diminished GPX4 function may be a critical event in triggering oxytosis/ferroptosis to perpetuate or initiate the neurodegenerative diseases and assess the possible therapeutic importance of oxytosis/ferroptosis in neurodegenerative disorders. These discoveries are important for advancing our understanding of neurodegenerative diseases because oxytosis/ferroptosis may provide a new target to slow the course of the disease.
Collapse
Affiliation(s)
- Nawab John Dar
- School of Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| | - Urmilla John
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India
- School of Studies in Zoology, Jiwaji University, Gwalior, India
| | - Nargis Bano
- Faculty of Life Sciences, Department of Zoology, Aligarh Muslim University, Aligarh, U.P, India
| | - Sameera Khan
- Faculty of Life Sciences, Department of Zoology, Aligarh Muslim University, Aligarh, U.P, India
| | - Shahnawaz Ali Bhat
- Faculty of Life Sciences, Department of Zoology, Aligarh Muslim University, Aligarh, U.P, India.
| |
Collapse
|
9
|
Wang Y, Li H, He Q, Zou R, Cai J, Zhang L. Ferroptosis: underlying mechanisms and involvement in neurodegenerative diseases. Apoptosis 2024; 29:3-21. [PMID: 37848673 DOI: 10.1007/s10495-023-01902-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2023] [Indexed: 10/19/2023]
Abstract
Ferroptosis, a mode of cell death that was recently identified in 2012, is driven by iron-dependent lipid peroxidation and distinct from other mechanisms of cell death such as autophagy and apoptosis. Ferroptosis has the unique features of disruptions in iron equilibrium, iron-induced lipid peroxidation, and the accumulation of glutamate-induced cellular toxicity. The regulation of ferroptosis mainly involves the iron, lipid, and amino acid metabolic pathways, which are controlled by system Xc-, voltage-dependent anion channels, p53 and other pathways. Neurodegenerative diseases involve gradual neuronal loss predominantly within the central nervous system and are categorized into both sporadic and rare hereditary disorders. These diseases result in the progressive decline of specific neuron populations and their interconnections. Recent investigations have revealed a strong correlation between the manifestation and progression of neurodegenerative diseases and ferroptosis. The pharmacological modulation of ferroptosis, whether by induction or inhibition, exhibits promising prospects for therapeutic interventions for these diseases. This review aims to examine the literature on ferroptosis and its implications in various neurodegenerative diseases. We hope to offer novel insights into the potential therapies targeting ferroptosis in central nervous system neurodegenerative diseases. However, there are still limitations of this review. First, despite our efforts to maintain objectivity during our analysis, this review does not cover all the studies on ferroptosis and neurodegenerative diseases. Second, cell death in neurodegenerative diseases is not solely caused by ferroptosis. Future research should focus on the interplay of different cell death mechanisms to better elucidate the specific disease pathogenesis.
Collapse
Affiliation(s)
- Yi Wang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - HongJing Li
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - QianXiong He
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rong Zou
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - JinRui Cai
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Lin Zhang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.
- Key Laboratory of Tibetan Medicine Research, Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences, Northwest Institute of Plateau Biology, Xining, 810008, Qinghai, China.
| |
Collapse
|
10
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
11
|
Zhang JB, Jia X, Cao Q, Chen YT, Tong J, Lu GD, Li DJ, Han T, Zhuang CL, Wang P. Ferroptosis-Regulated Cell Death as a Therapeutic Strategy for Neurodegenerative Diseases: Current Status and Future Prospects. ACS Chem Neurosci 2023; 14:2995-3012. [PMID: 37579022 DOI: 10.1021/acschemneuro.3c00406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
Ferroptosis is increasingly being recognized as a key element in the pathogenesis of diverse diseases. Recent studies have highlighted the intricate links between iron metabolism and neurodegenerative disorders. Emerging evidence suggests that iron homeostasis, oxidative stress, and neuroinflammation all contribute to the regulation of both ferroptosis and neuronal health. However, the precise molecular mechanisms underlying the involvement of ferroptosis in the pathological processes of neurodegeneration and its impact on neuronal dysfunction remain incompletely understood. In our Review, we provide a comprehensive analysis and summary of the potential molecular mechanisms underlying ferroptosis in neurodegenerative diseases, aiming to elucidate the disease progression of neurodegeneration. Additionally, we discuss potential therapeutic agents that modulate ferroptosis with the goal of identifying novel drug molecules for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Jia-Bao Zhang
- Department of Pharmacology, College of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
- National Experimental Teaching Demonstration Center of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Xiuqin Jia
- Department of Radiology, Beijing Chao Yang Hospital, Capital Medical University, Chaoyang District, Beijing 100020, China
| | - Qi Cao
- Department of Pharmacology, College of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
- National Experimental Teaching Demonstration Center of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Yi-Ting Chen
- Department of Pharmacology, College of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Guo-Dong Lu
- Department of Pharmacology, College of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ting Han
- Department of Pharmacology, College of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Chun-Lin Zhuang
- Department of Pharmacology, College of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Pei Wang
- Department of Pharmacology, College of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
- National Experimental Teaching Demonstration Center of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
12
|
Wang Y, Wu S, Li Q, Sun H, Wang H. Pharmacological Inhibition of Ferroptosis as a Therapeutic Target for Neurodegenerative Diseases and Strokes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300325. [PMID: 37341302 PMCID: PMC10460905 DOI: 10.1002/advs.202300325] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/23/2023] [Indexed: 06/22/2023]
Abstract
Emerging evidence suggests that ferroptosis, a unique regulated cell death modality that is morphologically and mechanistically different from other forms of cell death, plays a vital role in the pathophysiological process of neurodegenerative diseases, and strokes. Accumulating evidence supports ferroptosis as a critical factor of neurodegenerative diseases and strokes, and pharmacological inhibition of ferroptosis as a therapeutic target for these diseases. In this review article, the core mechanisms of ferroptosis are overviewed and the roles of ferroptosis in neurodegenerative diseases and strokes are described. Finally, the emerging findings in treating neurodegenerative diseases and strokes through pharmacological inhibition of ferroptosis are described. This review demonstrates that pharmacological inhibition of ferroptosis by bioactive small-molecule compounds (ferroptosis inhibitors) could be effective for treatments of these diseases, and highlights a potential promising therapeutic avenue that could be used to prevent neurodegenerative diseases and strokes. This review article will shed light on developing novel therapeutic regimens by pharmacological inhibition of ferroptosis to slow down the progression of these diseases in the future.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care MedicineAerospace Center HospitalPeking University Aerospace School of Clinical MedicineBeijing100049P. R. China
| | - Shuang Wu
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhan430000P. R. China
| | - Qiang Li
- Department of NeurologyThe Affiliated Hospital of Chifeng UniversityChifeng024005P. R. China
| | - Huiyan Sun
- Chifeng University Health Science CenterChifeng024000P. R. China
| | - Hongquan Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060P. R. China
| |
Collapse
|
13
|
Costa I, Barbosa DJ, Silva V, Benfeito S, Borges F, Remião F, Silva R. Research Models to Study Ferroptosis's Impact in Neurodegenerative Diseases. Pharmaceutics 2023; 15:pharmaceutics15051369. [PMID: 37242612 DOI: 10.3390/pharmaceutics15051369] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Ferroptosis is a type of regulated cell death promoted by the appearance of oxidative perturbations in the intracellular microenvironment constitutively controlled by glutathione peroxidase 4 (GPX4). It is characterized by increased production of reactive oxygen species, intracellular iron accumulation, lipid peroxidation, inhibition of system Xc-, glutathione depletion, and decreased GPX4 activity. Several pieces of evidence support the involvement of ferroptosis in distinct neurodegenerative diseases. In vitro and in vivo models allow a reliable transition to clinical studies. Several in vitro models, including differentiated SH-SY5Y and PC12 cells, among others, have been used to investigate the pathophysiological mechanisms of distinct neurodegenerative diseases, including ferroptosis. In addition, they can be useful in the development of potential ferroptosis inhibitors that can be used as disease-modifying drugs for the treatment of such diseases. On the other hand, in vivo models based on the manipulation of rodents and invertebrate animals, such as Drosophila melanogaster, Caenorhabditis elegans, and zebrafish, have been increasingly used for research in neurodegeneration. This work provides an up-to-date review of the main in vitro and in vivo models that can be used to evaluate ferroptosis in the most prevalent neurodegenerative diseases, and to explore potential new drug targets and novel drug candidates for effective disease-modifying therapies.
Collapse
Affiliation(s)
- Inês Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Vera Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
14
|
Costa I, Barbosa DJ, Benfeito S, Silva V, Chavarria D, Borges F, Remião F, Silva R. Molecular mechanisms of ferroptosis and their involvement in brain diseases. Pharmacol Ther 2023; 244:108373. [PMID: 36894028 DOI: 10.1016/j.pharmthera.2023.108373] [Citation(s) in RCA: 166] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Ferroptosis is a type of regulated cell death characterized by intracellular accumulation of iron and reactive oxygen species, inhibition of system Xc-, glutathione depletion, nicotinamide adenine dinucleotide phosphate oxidation and lipid peroxidation. Since its discovery and characterization in 2012, many efforts have been made to reveal the underlying mechanisms, modulating compounds, and its involvement in disease pathways. Ferroptosis inducers include erastin, sorafenib, sulfasalazine and glutamate, which, by inhibiting system Xc-, prevent the import of cysteine into the cells. RSL3, statins, Ml162 and Ml210 induce ferroptosis by inhibiting glutathione peroxidase 4 (GPX4), which is responsible for preventing the formation of lipid peroxides, and FIN56 and withaferin trigger GPX4 degradation. On the other side, ferroptosis inhibitors include ferrostatin-1, liproxstatin-1, α-tocopherol, zileuton, FSP1, CoQ10 and BH4, which interrupt the lipid peroxidation cascade. Additionally, deferoxamine, deferiprone and N-acetylcysteine, by targeting other cellular pathways, have also been classified as ferroptosis inhibitors. Increased evidence has established the involvement of ferroptosis in distinct brain diseases, including Alzheimer's, Parkinson's and Huntington's diseases, amyotrophic lateral sclerosis, multiple sclerosis, and Friedreich's ataxia. Thus, a deep understanding of how ferroptosis contributes to these diseases, and how it can be modulated, can open a new window of opportunities for novel therapeutic strategies and targets. Other studies have shown a sensitivity of cancer cells with mutated RAS to ferroptosis induction and that chemotherapeutic agents and ferroptosis inducers synergize in tumor treatment. Thus, it is tempting to consider that ferroptosis may arise as a target mechanistic pathway for the treatment of brain tumors. Therefore, this work provides an up-to-date review on the molecular and cellular mechanisms of ferroptosis and their involvement in brain diseases. In addition, information on the main ferroptosis inducers and inhibitors and their molecular targets is also provided.
Collapse
Affiliation(s)
- Inês Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- TOXRUN - Toxicology Research Unit, Department of Sciences, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal.
| | - Vera Silva
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
15
|
Martinez AM, Kim A, Flores CA, Rahman DF, Yang WS. Mouse embryonic stem cell-derived motor neurons are susceptible to ferroptosis. FEBS Open Bio 2023; 13:419-433. [PMID: 36595221 PMCID: PMC9989922 DOI: 10.1002/2211-5463.13545] [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: 09/23/2022] [Revised: 12/15/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
Ferroptosis is a regulated form of cell death driven by the lethal accumulation of lipid peroxides in cell membranes. Several regulators of ferroptosis have been identified using cancer cell lines. However, the cellular pathways of ferroptosis in neurons remain poorly characterized. In this study, we used a mouse embryonic stem cell-derived motor neuron model to investigate how motor neurons respond to ferroptosis inducers. Pharmacological and genetic inhibition of glutathione peroxidase 4 (GPx4) induced ferroptosis in motor neurons, while system xc - inhibition by erastin had no effect. RNA-seq analysis showed that the expression levels of several genes were altered during RSL3-induced ferroptosis. Subsequent bioinformatic analysis revealed alterations in several biological pathways during ferroptosis, including synaptogenesis and calcium signaling. Finally, we found that edaravone, an FDA-approved drug for treating amyotrophic lateral sclerosis (ALS) disease, rescued motor neurons from RSL3-induced ferroptosis. Our data highlight the crucial role of GPx4 in ferroptosis regulation and demonstrate that stem cell-derived motor neuron culture is a valuable model to study ferroptosis at the single-cell level in a neuronal context.
Collapse
Affiliation(s)
- Alejandra M Martinez
- Department of Biological Sciences, St. John's University, New York City, New York, USA
| | - Ahryun Kim
- Department of Biological Sciences, St. John's University, New York City, New York, USA
| | | | - Daoud F Rahman
- Department of Biological Sciences, St. John's University, New York City, New York, USA
| | - Wan Seok Yang
- Department of Biological Sciences, St. John's University, New York City, New York, USA
| |
Collapse
|
16
|
Torres J, Costa I, Peixoto AF, Silva R, Sousa Lobo JM, Silva AC. Intranasal Lipid Nanoparticles Containing Bioactive Compounds Obtained from Marine Sources to Manage Neurodegenerative Diseases. Pharmaceuticals (Basel) 2023; 16:311. [PMID: 37259454 PMCID: PMC9966140 DOI: 10.3390/ph16020311] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 01/22/2025] Open
Abstract
Marine sources contain several bioactive compounds with high therapeutic potential, such as remarkable antioxidant activity that can reduce oxidative stress related to the pathogenesis of neurodegenerative diseases. Indeed, there has been a growing interest in these natural sources, especially those resulting from the processing of marine organisms (i.e., marine bio-waste), to obtain natural antioxidants as an alternative to synthetic antioxidants in a sustainable approach to promote circularity by recovering and creating value from these bio-wastes. However, despite their expected potential to prevent, delay, or treat neurodegenerative diseases, antioxidant compounds may have difficulty reaching the brain due to the need to cross the blood-brain barrier (BBB). In this regard, alternative delivery systems administered by different routes have been proposed, including intranasal administration of lipid nanoparticles, such as solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC), which have shown promising results. Intranasal administration shows several advantages, including the fact that molecules do not need to cross the BBB to reach the central nervous system (CNS), as they can be transported directly from the nasal cavity to the brain (i.e., nose-to-brain transport). The benefits of using SLN and NLC for intranasal delivery of natural bioactive compounds for the treatment of neurodegenerative diseases have shown relevant outcomes through in vitro and in vivo studies. Noteworthy, for bioactive compounds obtained from marine bio-waste, few studies have been reported, showing the open potential of this research area. This review updates the state of the art of using SLN and NLC to transport bioactive compounds from different sources, in particular, those obtained from marine bio-waste, and their potential application in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Joana Torres
- UCIBIO, REQUIMTE, Laboratory of Pharmaceutical Technology/Centre of Research in Pharmaceutical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Inês Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-3131 Porto, Portugal
| | - Andreia F. Peixoto
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-3131 Porto, Portugal
| | - José Manuel Sousa Lobo
- UCIBIO, REQUIMTE, Laboratory of Pharmaceutical Technology/Centre of Research in Pharmaceutical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana Catarina Silva
- UCIBIO, REQUIMTE, Laboratory of Pharmaceutical Technology/Centre of Research in Pharmaceutical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- FP-I3ID (Instituto de Investigação, Inovação e Desenvolvimento), FP-BHS (Biomedical and Health Sciences Research Unit), Faculty of Health Sciences, University Fernando Pessoa, 4200-150 Porto, Portugal
| |
Collapse
|
17
|
Wang D, Liang W, Huo D, Wang H, Wang Y, Cong C, Zhang C, Yan S, Gao M, Su X, Tan X, Zhang W, Han L, Zhang D, Feng H. SPY1 inhibits neuronal ferroptosis in amyotrophic lateral sclerosis by reducing lipid peroxidation through regulation of GCH1 and TFR1. Cell Death Differ 2023; 30:369-382. [PMID: 36443440 PMCID: PMC9950139 DOI: 10.1038/s41418-022-01089-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 11/29/2022] Open
Abstract
Ferroptosis is an iron-dependent cell death with the accumulation of lipid peroxidation and dysfunction of antioxidant systems. As the critical regulator, glutathione peroxidase 4 (GPX4) has been demonstrated to be down-regulated in amyotrophic lateral sclerosis (ALS). However, the mechanism of ferroptosis in ALS remains unclear. In this research, bioinformatics analysis revealed a high correlation between ALS, ferroptosis, and Speedy/RINGO cell cycle regulator family member A (SPY1). Lipid peroxidation of ferroptosis in hSOD1G93A cells and mice was generated by TFR1-imported excess free iron, decreased GSH, mitochondrial membrane dysfunction, upregulated ALOX15, and inactivation of GCH1, GPX4. SPY1 is a "cyclin-like" protein that has been proved to enhance the viability of hSOD1G93A cells by inhibiting DNA damage. In our study, the decreased expression of SPY1 in ALS was resulted from unprecedented ubiquitination degradation mediated by MDM2 (a nuclear-localized E3 ubiquitin ligase). Further, SPY1 was identified as a novel ferroptosis suppressor via alleviating lipid peroxidation produced by dysregulated GCH1/BH4 axis (a resistance axis of ferroptosis) and transferrin receptor protein 1 (TFR1)-induced iron. Additionally, neuron-specific overexpression of SPY1 significantly delayed the occurrence and prolonged the survival in ALS transgenic mice through the above two pathways. These results suggest that SPY1 is a novel target for both ferroptosis and ALS.
Collapse
Affiliation(s)
- Di Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Weiwei Liang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Di Huo
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Hongyong Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Ying Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Chaohua Cong
- Department of Neurology, Shanghai JiaoTong University School of Medicine, Shanghai No. 9 People's Hospital, Shanghai, PR China
| | - Chunting Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei City, Anhui Province, PR China
| | - Shi Yan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Ming Gao
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang Province, PR China
| | - Xiaoli Su
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Xingli Tan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Wenmo Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Ling Han
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Dongmei Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Honglin Feng
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China.
| |
Collapse
|
18
|
Fu X, He Y, Xie Y, Lu Z. A conjoint analysis of bulk RNA-seq and single-nucleus RNA-seq for revealing the role of ferroptosis and iron metabolism in ALS. Front Neurosci 2023; 17:1113216. [PMID: 36937665 PMCID: PMC10017473 DOI: 10.3389/fnins.2023.1113216] [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: 12/01/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive and selective degeneration of motor neurons in the motor cortex of brain and spinal cord. Ferroptosis is a newly discovered form of cell death and reported to mediate selective motor neuron death in the mouse model of ALS. The growing awareness of ferroptosis and iron metabolism dysfunction in ALS prompted us to investigate the expression pattern of ferroptosis and iron metabolism-related genes (FIRGs) in ALS. Here, we performed a conjoint analysis of bulk-RNA sequence and single-nucleus RNA sequence data using the datasets from Gene Expression Omnibus (GEO) to reveal the role of FIRGs in ALS, especially in selective motor neuron death of ALS. We first investigated the differentially expressed genes (DEGs) between ALS and non-neurological controls. Weighted gene co-expression network analysis constructed the gene co-expression network and identified three modules closely associated with ALS. Fifteen FIRGs was identified as target genes based on least absolute shrinkage and selection operator regression analysis as follows: ACSL4, ANO6, ATP6V0E1, B2M, CD44, CHMP5, CYBB, CYBRD1, HIF1A, MOSPD1, NCF2, SDCBP, STEAP2, TMEM14C, ULK1. These genes could differentiate ALS patients from non-neurological controls (p < 2.2e-16) and had a valid value in predicting and diagnosing ALS (AUC = 0.881 in primary dataset and AUC = 0.768 in validation dataset). Then we performed the functional enrichment analysis of DEGs between ALS cases, the most significantly influenced by target genes, and non-neurological controls. The result indicated that the most significantly influenced functions in ALS pathogenesis by these identified FIRGs are synapse pathways, calcium signaling pathway, cAMP signaling pathway, and phagosome and several immune pathways. At last, the analysis of single- nuclear seq found that CHMP5, one of the 15 FIRGs identified by bulk single-nucleus RNA-seq data, was expressed significantly higher in ALS than pathologically normal (PN), specifically in excitatory neuron populations with layer 2 and layer 3 markers (Ex L2_L3), layer 3 and layer 5 markers (Ex L3_L5). Taken together, our study indicates the positive correlation between FIRGs and ALS, presents potential markers for ALS diagnosis and provides new research directions of CHMP5 function in selective motor neuron death in ALS.
Collapse
Affiliation(s)
- Xiujuan Fu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yizi He
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongzhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yongzhi Xie,
| | - Zuneng Lu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Zuneng Lu,
| |
Collapse
|
19
|
Soares P, Silva C, Chavarria D, Silva FSG, Oliveira PJ, Borges F. Drug discovery and amyotrophic lateral sclerosis: Emerging challenges and therapeutic opportunities. Ageing Res Rev 2023; 83:101790. [PMID: 36402404 DOI: 10.1016/j.arr.2022.101790] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the degeneration of upper and lower motor neurons (MNs) leading to paralysis and, ultimately, death by respiratory failure 3-5 years after diagnosis. Edaravone and Riluzole, the only drugs currently approved for ALS treatment, only provide mild symptomatic relief to patients. Extraordinary progress in understanding the biology of ALS provided new grounds for drug discovery. Over the last two decades, mitochondria and oxidative stress (OS), iron metabolism and ferroptosis, and the major regulators of hypoxia and inflammation - HIF and NF-κB - emerged as promising targets for ALS therapeutic intervention. In this review, we focused our attention on these targets to outline and discuss current advances in ALS drug development. Based on the challenges and the roadblocks, we believe that the rational design of multi-target ligands able to modulate the complex network of events behind the disease can provide effective therapies in a foreseeable future.
Collapse
Affiliation(s)
- Pedro Soares
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| | - Catia Silva
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Filomena S G Silva
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paulo J Oliveira
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; IIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
20
|
Nogami M, Sano O, Adachi-Tominari K, Hayakawa-Yano Y, Furukawa T, Iwata H, Ogi K, Okano H, Yano M. DNA damage stress-induced translocation of mutant FUS proteins into cytosolic granules and screening for translocation inhibitors. Front Mol Neurosci 2022; 15:953365. [PMID: 36606141 PMCID: PMC9808394 DOI: 10.3389/fnmol.2022.953365] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Fused in sarcoma/translated in liposarcoma (FUS) is an RNA-binding protein, and its mutations are associated with neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), through the DNA damage stress response, aberrant stress granule (SG) formation, etc. We previously reported that translocation of endogenous FUS into SGs was achieved by cotreatment with a DNA double-strand break inducer and an inhibitor of DNA-PK activity. In the present study, we investigated cytoplasmic SG formation using various fluorescent protein-tagged mutant FUS proteins in a human astrocytoma cell (U251) model. While the synergistic enhancement of the migration of fluorescent protein-tagged wild-type FUS to cytoplasmic SGs upon DNA damage induction was observed when DNA-PK activity was suppressed, the fluorescent protein-tagged FUSP525L mutant showed cytoplasmic localization. It migrated to cytoplasmic SGs upon DNA damage induction alone, and DNA-PK inhibition also showed a synergistic effect. Furthermore, analysis of 12 sites of DNA-PK-regulated phosphorylation in the N-terminal LC region of FUS revealed that hyperphosphorylation of FUS mitigated the mislocalization of FUS into cytoplasmic SGs. By using this cell model, we performed screening of a compound library to identify compounds that inhibit the migration of FUS to cytoplasmic SGs but do not affect the localization of the SG marker molecule G3BP1 to cytoplasmic SGs. Finally, we successfully identified 23 compounds that inhibit FUS-containing SG formation without changing normal SG formation. Highlights Characterization of DNA-PK-dependent FUS stress granule localization.A compound library was screened to identify compounds that inhibit the formation of FUS-containing stress granules.
Collapse
Affiliation(s)
- Masahiro Nogami
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan,Shonan Incubation Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan,*Correspondence: Masahiro Nogami,
| | - Osamu Sano
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Keiko Adachi-Tominari
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yoshika Hayakawa-Yano
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan,Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takako Furukawa
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hidehisa Iwata
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kazuhiro Ogi
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan,Shonan Incubation Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Masato Yano
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan,Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan,Masato Yano,
| |
Collapse
|
21
|
Yerton M, Winter A, Kostov A, Lieberman C, Gelevski D, Weber H, Doyle M, Kane G, Parikh N, Burke KM, Rohrer M, Stirrat T, Bruno M, Hochman A, Luppino S, Scalia J, Skoniecki D, D'Agostino D, Sinani E, Yu H, Sherman AV, Babu S, Berry JD, Midei MG, Milner PG, Cudkowicz ME, Paganoni S. An expanded access protocol of RT001 in amyotrophic lateral sclerosis-Initial experience with a lipid peroxidation inhibitor. Muscle Nerve 2022; 66:421-425. [PMID: 35765222 PMCID: PMC9796343 DOI: 10.1002/mus.27672] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 01/01/2023]
Abstract
INTRODUCTION/AIMS Lipid peroxidation is thought to play a biologically important role in motor neuron death in amyotrophic lateral sclerosis (ALS). 11,11 Di-deuterated linoleic ethyl ester (RT001) prevents lipid peroxidation in cellular and mitochondrial membranes. Herein we report on the use of RT001 under expanded access (EA). METHODS We provided RT001 to patients with ALS via EA at a single site. The starting dose was 2.88 g/day, which was increased to to 8.64 g/day as tolerated. Participants were not eligible for alternative clinical trials. Participants were followed for adverse events and pharmacokinetic (PK) parameters were measured approximately 3 months after RT001 initiation. RESULTS Sixteen participants received RT001 (5.6 ± 1.6 g/day; dose range, 1.92 to 8.64 g/day) for a mean period of 10.8 ± 7.1 months. After 3 months of treatment, PK studies showed that RT001 was absorbed, metabolized, and incorporated into red blood cell membranes at concentrations expected to be therapeutic based on in vitro models. The most common adverse events were gastrointestinal, including diarrhea, which occurred in 25% of the participants, and were considered possibly related to RT001. One participant (6%) discontinued due to an adverse event. Ten serious adverse events occurred: these events were recognized complications of ALS and none were attributed to treatment with RT001. DISCUSSION RT001 was administered safely to a small group of people living with ALS in the context of an EA protocol. Currently, there is an ongoing randomized, double-blind, controlled study of RT001 in ALS.
Collapse
Affiliation(s)
- Megan Yerton
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Allison Winter
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Anthony Kostov
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Cassandra Lieberman
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Dario Gelevski
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Harli Weber
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Michael Doyle
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Geli Kane
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Neil Parikh
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Katherine M. Burke
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Margot Rohrer
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Taylor Stirrat
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Margaret Bruno
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Alison Hochman
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Sarah Luppino
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Jennifer Scalia
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Debra Skoniecki
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Derek D'Agostino
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Ervin Sinani
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Hong Yu
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Alexander V. Sherman
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Suma Babu
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - James D. Berry
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | | | | | - Merit E. Cudkowicz
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts
| | - Sabrina Paganoni
- Sean M. Healey and AMG Center for ALS & the Neurological Clinical Research Institute, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusetts,Spaulding Rehabilitation Hospital, Harvard Medical SchoolBostonMassachusetts
| |
Collapse
|
22
|
Zhang L, Liu J, Dai Z, Wang J, Wu M, Su R, Zhang D. Crosstalk between regulated necrosis and micronutrition, bridged by reactive oxygen species. Front Nutr 2022; 9:1003340. [PMID: 36211509 PMCID: PMC9543034 DOI: 10.3389/fnut.2022.1003340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/26/2022] [Indexed: 11/15/2022] Open
Abstract
The discovery of regulated necrosis revitalizes the understanding of necrosis from a passive and accidental cell death to a highly coordinated and genetically regulated cell death routine. Since the emergence of RIPK1 (receptor-interacting protein kinase 1)-RIPK3-MLKL (mixed lineage kinase domain-like) axis-mediated necroptosis, various other forms of regulated necrosis, including ferroptosis and pyroptosis, have been described, which enrich the understanding of pathophysiological nature of diseases and provide novel therapeutics. Micronutrients, vitamins, and minerals, position centrally in metabolism, which are required to maintain cellular homeostasis and functions. A steady supply of micronutrients benefits health, whereas either deficiency or excessive amounts of micronutrients are considered harmful and clinically associated with certain diseases, such as cardiovascular disease and neurodegenerative disease. Recent advance reveals that micronutrients are actively involved in the signaling pathways of regulated necrosis. For example, iron-mediated oxidative stress leads to lipid peroxidation, which triggers ferroptotic cell death in cancer cells. In this review, we illustrate the crosstalk between micronutrients and regulated necrosis, and unravel the important roles of micronutrients in the process of regulated necrosis. Meanwhile, we analyze the perspective mechanism of each micronutrient in regulated necrosis, with a particular focus on reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Lei Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Jinting Liu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Ziyan Dai
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Jia Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Mengyang Wu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Ruicong Su
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Di Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- *Correspondence: Di Zhang,
| |
Collapse
|
23
|
Sun Y, Xia X, Basnet D, Zheng JC, Huang J, Liu J. Mechanisms of Ferroptosis and Emerging Links to the Pathology of Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:904152. [PMID: 35837484 PMCID: PMC9273851 DOI: 10.3389/fnagi.2022.904152] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/03/2022] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative diseases are a diverse class of diseases attributed to chronic progressive neuronal degeneration and synaptic loss in the brain and/or spinal cord, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis and multiple sclerosis. The pathogenesis of neurodegenerative diseases is complex and diverse, often involving mitochondrial dysfunction, neuroinflammation, and epigenetic changes. However, the pathogenesis of neurodegenerative diseases has not been fully elucidated. Recently, accumulating evidence revealed that ferroptosis, a newly discovered iron-dependent and lipid peroxidation-driven type of programmed cell death, provides another explanation for the occurrence and progression of neurodegenerative diseases. Here, we provide an overview of the process and regulation mechanisms of ferroptosis, and summarize current research progresses that support the contribution of ferroptosis to the pathogenesis of neurodegenerative diseases. A comprehensive understanding of the emerging roles of ferroptosis in neurodegenerative diseases will shed light on the development of novel therapeutic technologies and strategies for slowing down the progression of these diseases.
Collapse
Affiliation(s)
- Yiyan Sun
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaohuan Xia
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Diksha Basnet
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jialin C. Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
- *Correspondence: Jialin C. Zheng,
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Jian Huang,
| | - Jianhui Liu
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Jianhui Liu,
| |
Collapse
|
24
|
Ferroptosis mediates selective motor neuron death in amyotrophic lateral sclerosis. Cell Death Differ 2022; 29:1187-1198. [PMID: 34857917 PMCID: PMC9177596 DOI: 10.1038/s41418-021-00910-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/29/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is caused by selective degeneration of motor neurons in the brain and spinal cord; however, the primary cell death pathway(s) mediating motor neuron demise remain elusive. We recently established that necroptosis, an inflammatory form of regulated cell death, was dispensable for motor neuron death in a mouse model of ALS, implicating other forms of cell death. Here, we confirm these findings in ALS patients, showing a lack of expression of key necroptotic effector proteins in spinal cords. Rather, we uncover evidence for ferroptosis, a recently discovered iron-dependent form of regulated cell death, in ALS. Depletion of glutathione peroxidase 4 (GPX4), an anti-oxidant enzyme and central repressor of ferroptosis, occurred in post-mortem spinal cords of both sporadic and familial ALS patients. GPX4 depletion was also an early and universal feature of spinal cords and brains of transgenic mutant superoxide dismutase 1 (SOD1G93A), TDP-43 and C9orf72 mouse models of ALS. GPX4 depletion and ferroptosis were linked to impaired NRF2 signalling and dysregulation of glutathione synthesis and iron-binding proteins. Novel BAC transgenic mice overexpressing human GPX4 exhibited high GPX4 expression localised to spinal motor neurons. Human GPX4 overexpression in SOD1G93A mice significantly delayed disease onset, improved locomotor function and prolonged lifespan, which was attributed to attenuated lipid peroxidation and motor neuron preservation. Our study discovers a new role for ferroptosis in mediating motor neuron death in ALS, supporting the use of anti-ferroptotic therapeutic strategies, such as GPX4 pathway induction and upregulation, for ALS treatment.
Collapse
|
25
|
Shadfar S, Brocardo M, Atkin JD. The Complex Mechanisms by Which Neurons Die Following DNA Damage in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23052484. [PMID: 35269632 PMCID: PMC8910227 DOI: 10.3390/ijms23052484] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/12/2022] [Accepted: 02/17/2022] [Indexed: 01/18/2023] Open
Abstract
Human cells are exposed to numerous exogenous and endogenous insults every day. Unlike other molecules, DNA cannot be replaced by resynthesis, hence damage to DNA can have major consequences for the cell. The DNA damage response contains overlapping signalling networks that repair DNA and hence maintain genomic integrity, and aberrant DNA damage responses are increasingly described in neurodegenerative diseases. Furthermore, DNA repair declines during aging, which is the biggest risk factor for these conditions. If unrepaired, the accumulation of DNA damage results in death to eliminate cells with defective genomes. This is particularly important for postmitotic neurons because they have a limited capacity to proliferate, thus they must be maintained for life. Neuronal death is thus an important process in neurodegenerative disorders. In addition, the inability of neurons to divide renders them susceptible to senescence or re-entry to the cell cycle. The field of cell death has expanded significantly in recent years, and many new mechanisms have been described in various cell types, including neurons. Several of these mechanisms are linked to DNA damage. In this review, we provide an overview of the cell death pathways induced by DNA damage that are relevant to neurons and discuss the possible involvement of these mechanisms in neurodegenerative conditions.
Collapse
Affiliation(s)
- Sina Shadfar
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (S.S.); (M.B.)
| | - Mariana Brocardo
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (S.S.); (M.B.)
| | - Julie D. Atkin
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (S.S.); (M.B.)
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086, Australia
- Correspondence:
| |
Collapse
|
26
|
Arslanbaeva L, Bisaglia M. Activation of the Nrf2 Pathway as a Therapeutic Strategy for ALS Treatment. Molecules 2022; 27:1471. [PMID: 35268572 PMCID: PMC8911691 DOI: 10.3390/molecules27051471] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic lateral sclerosis is a progressive and fatal disease that causes motoneurons degeneration and functional impairment of voluntary muscles, with limited and poorly efficient therapies. Alterations in the Nrf2-ARE pathway are associated with ALS pathology and result in aberrant oxidative stress, making the stimulation of the Nrf2-mediated antioxidant response a promising therapeutic strategy in ALS to reduce oxidative stress. In this review, we first introduce the involvement of the Nrf2 pathway in the pathogenesis of ALS and the role played by astrocytes in modulating such a protective pathway. We then describe the currently developed activators of Nrf2, used in both preclinical animal models and clinical studies, taking into consideration their potentialities as well as the possible limitations associated with their use.
Collapse
Affiliation(s)
| | - Marco Bisaglia
- Department of Biology, University of Padua, 35131 Padua, Italy
- Center Study for Neurodegeneration (CESNE), University of Padua, 35131 Padua, Italy
| |
Collapse
|
27
|
Gleitze S, Paula-Lima A, Núñez MT, Hidalgo C. The calcium-iron connection in ferroptosis-mediated neuronal death. Free Radic Biol Med 2021; 175:28-41. [PMID: 34461261 DOI: 10.1016/j.freeradbiomed.2021.08.231] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022]
Abstract
Iron, through its participation in oxidation/reduction processes, is essential for the physiological function of biological systems. In the brain, iron is involved in the development of normal cognitive functions, and its lack during development causes irreversible cognitive damage. Yet, deregulation of iron homeostasis provokes neuronal damage and death. Ferroptosis, a newly described iron-dependent cell death pathway, differs at the morphological, biochemical, and genetic levels from other cell death types. Ferroptosis is characterized by iron-mediated lipid peroxidation, depletion of the endogenous antioxidant glutathione and altered mitochondrial morphology. Although iron promotes the emergence of Ca2+ signals via activation of redox-sensitive Ca2+ channels, the role of Ca2+ signaling in ferroptosis has not been established. The early dysregulation of the cellular redox state observed in ferroptosis is likely to disturb Ca2+ homeostasis and signaling, facilitating ferroptotic neuronal death. This review presents an overview of the role of iron and ferroptosis in neuronal function, emphasizing the possible involvement of Ca2+ signaling in these processes. We propose, accordingly, that the iron-ferroptosis-Ca2+ association orchestrates the progression of cognitive dysfunctions and memory loss that occurs in neurodegenerative diseases. Therefore, to prevent iron dyshomeostasis and ferroptosis, we suggest the use of drugs that target the abnormal Ca2+ signaling caused by excessive iron levels as therapy for neurological disorders.
Collapse
Affiliation(s)
- Silvia Gleitze
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrea Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Marco T Núñez
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Physiology and Biophysics Program, Institute of Biomedical Sciences and Center for Exercise, Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|