1
|
Naderi S, Khodagholi F, Janahmadi M, Motamedi F, Torabi A, Batool Z, Heydarabadi MF, Pourbadie HG. Ferroptosis and cognitive impairment: Unraveling the link and potential therapeutic targets. Neuropharmacology 2025; 263:110210. [PMID: 39521042 DOI: 10.1016/j.neuropharm.2024.110210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases, share key characteristics, notably cognitive impairment and significant cell death in specific brain regions. Cognition, a complex mental process allowing individuals to perceive time and place, is disrupted in these conditions. This consistent disruption suggests the possibility of a shared underlying mechanism across all neurodegenerative diseases. One potential common factor is the activation of pathways leading to cell death. Despite significant progress in understanding cell death pathways, no definitive treatments have emerged. This has shifted focus towards less-explored mechanisms like ferroptosis, which holds potential due to its involvement in oxidative stress and iron metabolism. Unlike apoptosis or necrosis, ferroptosis offers a novel therapeutic avenue due to its distinct biochemical and genetic underpinnings, making it a promising target in neurodegenerative disease treatment. Ferroptosis is distinguished from other cellular death mechanisms, by distinctive characteristics such as an imbalance of iron hemostasis, peroxidation of lipids in the plasma membrane, and dysregulated glutathione metabolism. In this review, we discuss the potential role of ferroptosis in cognitive impairment. We then summarize the evidence linking ferroptosis biomarkers to cognitive impairment brought on by neurodegeneration while highlighting recent advancements in our understanding of the molecular and genetic mechanisms behind the condition. Finally, we discuss the prospective therapeutic implications of targeting ferroptosis for the treatment of cognitive abnormalities associated with neurodegeneration, including natural and synthetic substances that suppress ferroptosis via a variety of mechanisms. Promising therapeutic candidates, including antioxidants and iron chelators, are being explored to inhibit ferroptosis and mitigate cognitive decline.
Collapse
Affiliation(s)
- Soudabeh Naderi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Torabi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Hamid Gholami Pourbadie
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
da Costa Caiado MJ, Dolga AM, den Dunnen WFA. Iron(ing) out parkinsonisms: The interplay of proteinopathy and ferroptosis in Parkinson's disease and tau-related parkinsonisms. Redox Biol 2024; 79:103478. [PMID: 39721496 DOI: 10.1016/j.redox.2024.103478] [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: 11/01/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Parkinsonian syndromes are characterised by similar motor-related symptomology resulting from dopaminergic neuron damage. While Parkinson's disease (PD) is the most prevalent parkinsonism, we also focus on two other variants, Progressive supranuclear palsy (PSP) and Corticobasal degeneration (CBD). Due to the clinical similarities of these parkinsonisms, and since definite diagnoses are only possible post-mortem, effective therapies and novel biomarkers of disease are scarce. Thus, we explore the current findings relating to the relationship of parkinsonism proteinopathy (α-synuclein in PD, and tau in PSP/CBD) paralleled to a specific form of cell death, ferroptosis. Ferroptosis is characterised by iron-induced lipid peroxidation and several markers of this pathway have been identified to control intracellular iron fluctuations. However, in parkinsonism, these mechanisms are thought to become dysfunctional. Although both proteinopathies have been linked to ferroptosis, much less is known about ferroptotic cell death and tau in the context of PSP/CBD. Interestingly, clinical trials targeting iron have recently shown conflicting results which begs to question the complexity of the ferroptotic pathway and alludes to the need for exploring other ferroptosis-related machinery as possible therapeutic targets. Overall, we address the literature gap in parkinsonism proteinopathy and ferroptosis, and its relevance to understanding disease pathophysiology and aetiology.
Collapse
Affiliation(s)
- Maria João da Costa Caiado
- Graduate School of Medical Sciences (GSMS) and Research School of Behavioural and Cognitive Neurosciences (BCN), University of Groningen, 9713 GZ, Groningen, the Netherlands; Department of Pathology and Medical Biology, University Medical Centre Groningen (UMCG), Hanzeplein 1, 9713 GZ, Groningen, the Netherlands; Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, the Netherlands.
| | - Amalia M Dolga
- Department of Pathology and Medical Biology, University Medical Centre Groningen (UMCG), Hanzeplein 1, 9713 GZ, Groningen, the Netherlands; Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, the Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University Medical Centre Groningen (UMCG), Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
3
|
Sabogal-Guaqueta AM, Mitchell-Garcia T, Hunneman J, Voshart D, Thiruvalluvan A, Foijer F, Kruyt F, Trombetta-Lima M, Eggen BJL, Boddeke E, Barazzuol L, Dolga AM. Brain organoid models for studying the function of iPSC-derived microglia in neurodegeneration and brain tumours. Neurobiol Dis 2024; 203:106742. [PMID: 39581553 DOI: 10.1016/j.nbd.2024.106742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Microglia represent the main resident immune cells of the brain. The interplay between microglia and other cells in the central nervous system, such as neurons or other glial cells, influences the function and ability of microglia to respond to various stimuli. These cellular communications, when disrupted, can affect the structure and function of the brain, and the initiation and progression of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease, as well as the progression of other brain diseases like glioblastoma. Due to the difficult access to patient brain tissue and the differences reported in the murine models, the available models to study the role of microglia in disease progression are limited. Pluripotent stem cell technology has facilitated the generation of highly complex models, allowing the study of control and patient-derived microglia in vitro. Moreover, the ability to generate brain organoids that can mimic the 3D tissue environment and intercellular interactions in the brain provide powerful tools to study cellular pathways under homeostatic conditions and various disease pathologies. In this review, we summarise the most recent developments in modelling degenerative diseases and glioblastoma, with a focus on brain organoids with integrated microglia. We provide an overview of the most relevant research on intercellular interactions of microglia to evaluate their potential to study brain pathologies.
Collapse
Affiliation(s)
- Angelica Maria Sabogal-Guaqueta
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Teresa Mitchell-Garcia
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Jasmijn Hunneman
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Daniëlle Voshart
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Arun Thiruvalluvan
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frank Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands; Faculty of Science and Engineering, Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section of Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Erik Boddeke
- Department of Biomedical Sciences, Section of Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Lara Barazzuol
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands; Department Pathology and Medical biology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
4
|
Majerníková N, Marmolejo-Garza A, Salinas CS, Luu MDA, Zhang Y, Trombetta-Lima M, Tomin T, Birner-Gruenberger R, Lehtonen Š, Koistinaho J, Wolters JC, Ayton S, den Dunnen WFA, Dolga AM. The link between amyloid β and ferroptosis pathway in Alzheimer's disease progression. Cell Death Dis 2024; 15:782. [PMID: 39468028 PMCID: PMC11519607 DOI: 10.1038/s41419-024-07152-0] [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: 06/03/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024]
Abstract
Alzheimer's disease (AD) affects millions of people worldwide and represents the most prevalent form of dementia. Treatment strategies aiming to interfere with the formation of amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs), the two major AD hallmarks, have shown modest or no effect. Recent evidence suggests that ferroptosis, a type of programmed cell death caused by iron accumulation and lipid peroxidation, contributes to AD pathogenesis. The existing link between ferroptosis and AD has been largely based on cell culture and animal studies, while evidence from human brain tissue is limited. Here we evaluate if Aβ is associated with ferroptosis pathways in post-mortem human brain tissue and whether ferroptosis inhibition could attenuate Aβ-related effects in human brain organoids. Performing positive pixel density scoring on immunohistochemically stained post-mortem Brodmann Area 17 sections revealed that the progression of AD pathology was accompanied by decreased expression of nuclear receptor co-activator 4 and glutathione peroxidase 4 in the grey matter. Differentiating between white and grey matter, allowed for a more precise understanding of the disease's impact on different brain regions. In addition, ferroptosis inhibition prevented Aβ pathology, decreased lipid peroxidation and restored iron storage in human AD iPSCs-derived brain cortical organoids at day 50 of differentiation. Differential gene expression analysis of RNAseq of AD organoids compared to isogenic controls indicated activation of the ferroptotic pathway. This was also supported by results from untargeted proteomic analysis revealing significant changes between AD and isogenic brain organoids. Determining the causality between the development of Aβ plaques and the deregulation of molecular pathways involved in ferroptosis is crucial for developing potential therapeutic interventions.
Collapse
Affiliation(s)
- Naďa Majerníková
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, Research Institute Brain and Cognition, Molecular Neuroscience and Aging Research, Research School of Behavioural and Cognitive Neuroscience, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Alejandro Marmolejo-Garza
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells and Systems, Molecular Neurobiology Section, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Casandra Salinas Salinas
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
| | - Minh D A Luu
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
| | - Yuequ Zhang
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
| | - Šárka Lehtonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jari Koistinaho
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Justina C Wolters
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Scott Ayton
- The Florey Neuroscience Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, Research Institute Brain and Cognition, Molecular Neuroscience and Aging Research, Research School of Behavioural and Cognitive Neuroscience, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands.
- Department of Pathology and Medical Biology, Research Institute Brain and Cognition, Molecular Neuroscience and Aging Research, Research School of Behavioural and Cognitive Neuroscience, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
5
|
Li S, Yang J. Pathogenesis of Alzheimer's disease and therapeutic strategies involving traditional Chinese medicine. RSC Med Chem 2024; 15:d4md00660g. [PMID: 39430949 PMCID: PMC11484936 DOI: 10.1039/d4md00660g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent degenerative disorder affecting the central nervous system of the elderly. Patients primarily manifest cognitive decline and non-cognitive neuro-psychiatric symptoms. Currently, western medications for AD primarily include cholinesterase inhibitors and glutamate receptor inhibitors, which have limited efficacy and accompanied by significant toxic side effects. Given the intricate pathogenesis of AD, the use of single-target inhibitors is limited. In recent years, as research on AD has progressed, traditional Chinese medicine (TCM) and its active ingredients have increasingly played a crucial role in clinical treatment. Numerous studies demonstrate that TCM and its active ingredients can exert anti-Alzheimer's effects by modulating pathological protein production and deposition, inhibiting tau protein hyperphosphorylation, apoptosis, inflammation, and oxidative stress, while enhancing the central cholinergic system, protecting neurons and synapses, and optimizing energy metabolism. This article summarizes extracts from TCM and briefly elucidates their pharmacological mechanisms against AD, aiming to provide a foundation for further research into the specific mechanisms of TCM in the prevention and treatment of the disease, as well as the identification of efficacious active ingredients.
Collapse
Affiliation(s)
- Shutang Li
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine Qingdao 266041 China
| | - Jinfei Yang
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine Qingdao 266041 China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences Qingdao 266113 China
| |
Collapse
|
6
|
Marmolejo-Garza A, Chatre L, Croteau DL, Herron-Bedoya A, Luu MDA, Bernay B, Pontin J, Bohr VA, Boddeke E, Dolga AM. Nicotinamide riboside modulates the reactive species interactome, bioenergetic status and proteomic landscape in a brain-region-specific manner. Neurobiol Dis 2024; 200:106645. [PMID: 39179121 DOI: 10.1016/j.nbd.2024.106645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
Nicotinamide riboside (NR), a precursor of nicotinamide adenine dinucleotide (NAD+), has robust cognitive benefits and alleviates neuroinflammation in Alzheimer's Disease (AD) mouse models without decreasing beta-amyloid plaque pathology. Such effects may be mediated by the reactive species interactome (RSI), at the metabolome level. In this study, we employed in vitro and in vivo models of oxidative stress, aging and AD to profile the effects of NR on neuronal survival, RSI, and the whole proteome characterization of cortex and hippocampus. RSI analysis yielded a complex modulation upon NR treatment. We constructed protein co-expression networks and correlated them to NR treatment and all measured reactive species. We observed brain-area specific effects of NR on co-expressed protein modules of oxidative phosphorylation, fatty acid oxidation, and neurotransmitter regulation pathways, which correlated with RSI components. The current study contributes to the understanding of modulation of the metabolome, specifically after NR treatment in AD and how it may play disease-modifying roles.
Collapse
Affiliation(s)
- Alejandro Marmolejo-Garza
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713, AV, Groningen, the Netherlands; Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Laurent Chatre
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP CYCERON, F-14000 Caen, France
| | - Deborah L Croteau
- Section on DNA repair, National Institute on Aging, 251 Bayview Blvd, Baltimore, MD, USA; Laboratory of Genetics and Genomics, Computational Biology and Genomics Core, National Institute on Aging, 251 Bayview Blvd, Baltimore, USA
| | - Alejandro Herron-Bedoya
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713, AV, Groningen, the Netherlands
| | - Minh Danh Anh Luu
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713, AV, Groningen, the Netherlands
| | - Benoit Bernay
- Université de Caen Normandie, US EMerode, Plateform Proteogen, F-14000 Caen, France
| | - Julien Pontin
- Université de Caen Normandie, US EMerode, Plateform Proteogen, F-14000 Caen, France
| | - Vilhelm A Bohr
- Section on DNA repair, National Institute on Aging, 251 Bayview Blvd, Baltimore, MD, USA; Center for Healthy Aging, Department of Cellular and Molecular Medicine, SUND, University of Copenhagen, 2200, Copenhagen N, Denmark; Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Erik Boddeke
- Department of Biomedical Sciences of Cells & Systems, section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713, AV, Groningen, the Netherlands.
| |
Collapse
|
7
|
Chen D, Zhao C, Zhang J, Knol CWJ, Osipyan A, Majerníková N, Chen T, Xiao Z, Adriana J, Griffith AJ, Gamez AS, van der Wouden PE, Coppes RP, Dolga AM, Haisma HJ, Dekker FJ. Small Molecule MIF Modulation Enhances Ferroptosis by Impairing DNA Repair Mechanisms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403963. [PMID: 38924362 PMCID: PMC11348242 DOI: 10.1002/advs.202403963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Ferroptosis is a form of regulated cell death that can be modulated by small molecules and has the potential for the development of therapeutics for oncology. Although excessive lipid peroxidation is the defining hallmark of ferroptosis, DNA damage may also play a significant role. In this study, a potential mechanistic role for MIF in homologous recombination (HR) DNA repair is identified. The inhibition or genetic depletion of MIF or other HR proteins, such as breast cancer type 1 susceptibility protein (BRCA1), is demonstrated to significantly enhance the sensitivity of cells to ferroptosis. The interference with HR results in the translocation of the tumor suppressor protein p53 to the mitochondria, which in turn stimulates the production of reactive oxygen species. Taken together, the findings demonstrate that MIF-directed small molecules enhance ferroptosis via a putative MIF-BRCA1-RAD51 axis in HR, which causes resistance to ferroptosis. This suggests a potential novel druggable route to enhance ferroptosis by targeted anticancer therapeutics in the future.
Collapse
Affiliation(s)
- Deng Chen
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Chunlong Zhao
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Jianqiu Zhang
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Catharina W. J. Knol
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Angelina Osipyan
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Nad'a Majerníková
- Research School of Behavioural and Cognitive NeuroscienceUniversity of GroningenGroningen9713 AVThe Netherlands
- Department of Pathology and Medical BiologyUniversity Medical Centre GroningenUniversity of GroningenGroningen9713 GZThe Netherlands
- Department of Molecular PharmacologyGroningen Research Institute of PharmacyUniversity of GroningenGroningen9713 AVThe Netherlands
| | - Tingting Chen
- Department of Molecular PharmacologyGroningen Research Institute of PharmacyUniversity of GroningenGroningen9713 AVThe Netherlands
| | - Zhangping Xiao
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Jeaunice Adriana
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Andrew J. Griffith
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Abel Soto Gamez
- Department of Biomedical Sciences of Cell & SystemsSection Molecular Cell BiologyUniversity Medical Center GroningenUniversity of GroningenGroningen9712 CPThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenHanzeplein 1Groningen9713 GZNetherlands
| | - Petra E. van der Wouden
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Robert P. Coppes
- Department of Biomedical Sciences of Cell & SystemsSection Molecular Cell BiologyUniversity Medical Center GroningenUniversity of GroningenGroningen9712 CPThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenHanzeplein 1Groningen9713 GZNetherlands
| | - Amalia M. Dolga
- Research School of Behavioural and Cognitive NeuroscienceUniversity of GroningenGroningen9713 AVThe Netherlands
- Department of Molecular PharmacologyGroningen Research Institute of PharmacyUniversity of GroningenGroningen9713 AVThe Netherlands
| | - Hidde J. Haisma
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Frank J. Dekker
- Department of Chemical and Pharmaceutical BiologyGroningen Research Institute of Pharmacy (GRIP)University of GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| |
Collapse
|
8
|
Renner N, Schöb F, Pape R, Suciu I, Spreng AS, Ückert AK, Cöllen E, Bovio F, Chilian B, Bauer J, Röpcke S, Bergemann J, Leist M, Schildknecht S. Modeling ferroptosis in human dopaminergic neurons: Pitfalls and opportunities for neurodegeneration research. Redox Biol 2024; 73:103165. [PMID: 38688061 PMCID: PMC11070765 DOI: 10.1016/j.redox.2024.103165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/17/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
The activation of ferroptosis is being pursued in cancer research as a strategy to target apoptosis-resistant cells. By contrast, in various diseases that affect the cardiovascular system, kidneys, liver, and central and peripheral nervous systems, attention is directed toward interventions that prevent ferroptotic cell death. Mechanistic insights into both research areas stem largely from studies using cellular in vitro models. However, intervention strategies that show promise in cellular test systems often fail in clinical trials, which raises concerns regarding the predictive validity of the utilized in vitro models. In this study, the human LUHMES cell line, which serves as a model for human dopaminergic neurons, was used to characterize factors influencing the activation of ferroptosis. Erastin and RSL-3 induced cell death that was distinct from apoptosis. Parameters such as the differentiation state of LUHMES cells, cell density, and the number and timing of medium changes were identified as determinants of sensitivity to ferroptosis activation. In differentiated LUHMES cells, interventions at mechanistically divergent sites (iron chelation, coenzyme Q10, peroxidase mimics, or inhibition of 12/15-lipoxygenase) provide almost complete protection from ferroptosis. LUHMES cells allowed the experimental modulation of intracellular iron concentrations and demonstrated a correlation between intracellular iron levels, the rate of lipid peroxidation, as well as the sensitivity of the cells to ferroptotic cell death. These findings underscore the importance of understanding the various factors that influence ferroptosis activation and highlight the need for well-characterized in vitro models to enhance the reliability and predictive value of observations in ferroptosis research, particularly when translating findings into in vivo contexts.
Collapse
Affiliation(s)
- Nadine Renner
- Albstadt-Sigmaringen University, Faculty of Life Sciences, 72488, Sigmaringen, Germany
| | - Franziska Schöb
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Regina Pape
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Anna-Sophie Spreng
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Anna-Katharina Ückert
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Eike Cöllen
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Federica Bovio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milano, Italy
| | - Bruno Chilian
- TRI Thinking Research Instruments GmbH, Große Freiheit 77, 22767, Hamburg, Germany
| | - Johannes Bauer
- TRI Thinking Research Instruments GmbH, Große Freiheit 77, 22767, Hamburg, Germany
| | - Stefan Röpcke
- Stemick GmbH, Byk-Gulden Str. 2, 78467, Konstanz, Germany
| | - Jörg Bergemann
- Albstadt-Sigmaringen University, Faculty of Life Sciences, 72488, Sigmaringen, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Stefan Schildknecht
- Albstadt-Sigmaringen University, Faculty of Life Sciences, 72488, Sigmaringen, Germany.
| |
Collapse
|
9
|
Langner M, Fröbel D, Helm J, Chavakis T, Peitzsch M, Bechmann N. Accurate redox state indication by in situ derivatization with N-ethylmaleimide - Profiling of transsulfuration and glutathione pathway metabolites by UPLC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1236:124062. [PMID: 38432191 DOI: 10.1016/j.jchromb.2024.124062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Reduced and oxidized glutathione play an important role for the intracellular detoxification of reactive oxygen species. The iron-dependent formation of such reactive oxygen species in conjunction with the inhibition of the redox-balancing enzyme glutathione peroxidase 4 underlie an imbalance in the cellular redox state, thereby resulting in a non-apoptotic form of cell death, defined as ferroptosis, which is relevant in several pathologies. METHODS Here we present a rapid ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) based method providing the accurate quantification of 12 glutathione pathway metabolites after in situ derivatization with N-Ethylmaleimide (NEM). The method was validated regards linearity, recovery and accuracy as well as precision. The assay includes glutathione and its oxidized form glutathione disulfide. Furthermore, the related precursors cysteine, cystine, glutamic acid, γ-glutamylcysteine and cysteinylglycine, biomarkers of protein crosslinking such as cystathionine and lanthionine, as well as metabolites of the transsulfuration pathway, methionine, homocysteine and serine are simultaneously determined. RESULTS Twelve glutathione pathway metabolites were simultaneously analyzed in four different human cell line extracts within a total LC run time of 5.5 min. Interday coefficients of variation (1.7 % to 12.0 %), the mean observed accuracy (100.0 % ± 5.2 %), linear quantification ranges over three orders of magnitude for all analytes and sufficient metabolite stability after NEM-derivatization demonstrate method reliability. Immediate derivatization with NEM at cell harvesting prevents autooxidation of glutathione, ensures accurate results for the GSH/GSSG redox ratio and thereby allows interpretation of cellular redox state. CONCLUSION The described UPLC-MS/MS method provides a sensitive and selective tool for a fast and simultaneous analysis of glutathione pathway metabolites, its direct precursors and related compounds. Assay performance characteristics demonstrate the suitability of the method for applications in different cell cultures. Therefore, by providing glutathione related functional metabolic readouts, the method enables investigations in mechanisms of ferroptosis and alterations in oxidative stress levels in several pathophysiologies.
Collapse
Affiliation(s)
- Mathias Langner
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Dennis Fröbel
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Jana Helm
- Department of Medicine III, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| |
Collapse
|
10
|
Hacioglu C, Kar F, Ozbayer C, Gundogdu AC. Ex vivo investigation of betaine and boric acid function as preprotective agents on rat synaptosomes to be treated with Aβ (1-42). ENVIRONMENTAL TOXICOLOGY 2024; 39:2138-2149. [PMID: 38108610 DOI: 10.1002/tox.24098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 08/31/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Recent evidence suggests that ferroptosis, an iron-dependent cell death process, may be involved in Alzheimer's disease (AD) pathology. The study evaluated the therapeutic potential of betaine and boric acid (BA) pretreatment administered to rats for 21 days in AD. Then, the rats were sacrificed, and morphological and biochemical analyses were performed in brain tissues. Next, an ex vivo AD model was created by applying amyloid-β (Aβ1-42) to synaptosomes isolated from the brain tissues. Synaptosomes were analyzed with micrograph images, and protein and mRNA levels of ferroptotic markers were determined. Betaine and BA pretreatments did not cause any morphological and biochemical differences in the brain tissue. However, Aβ (1-42) administration in synaptosomes increased the levels of acyl-CoA synthetase long chain family member-4 (ACSL4), transferrin receptor-1 protein (TfR1), malondialdehyde (MDA), and 8-hydroxydeoxyguanosine (8-OHdG) and decreased the levels glutathione peroxidase-4 (GPx4) and glutathione (GSH). Moreover, ACSL4, GPx4, and TfR1 mRNA and protein levels were similar to the ELISA results. In contrast, betaine and BA pretreatments decreased the levels of ACSL4, TfR1, MDA, and 8-OHdG in synaptosomes incubated with Aβ1-42, while promoting increased levels of GPx4 and GSH. In addition, betaine and BA pretreatments completely reversed ACSL4, GPx4, and TfR1 mRNA and protein levels. Therefore, betaine and BA pretreatments may contribute to the prevention of neurodegenerative damage by supporting antiferroptotic activities.
Collapse
Affiliation(s)
- Ceyhan Hacioglu
- Faculty of Pharmacy, Department of Biochemistry, Düzce University, Düzce, Turkey
- Faculty of Medicine, Department of Medical Biochemistry, Düzce University, Düzce, Turkey
| | - Fatih Kar
- Faculty of Medicine, Department of Medical Biochemistry, Kütahya Health Sciences University, Kütahya, Turkey
| | - Cansu Ozbayer
- Faculty of Medicine, Department of Medical Biology, Kütahya Health Sciences University, Kütahya, Turkey
| | - Ayse Cakir Gundogdu
- Faculty of Medicine Department of Histology and Embryology, Kütahya Health Sciences University, Kütahya, Turkey
| |
Collapse
|
11
|
Consoli V, Fallica AN, Sorrenti V, Pittalà V, Vanella L. Novel Insights on Ferroptosis Modulation as Potential Strategy for Cancer Treatment: When Nature Kills. Antioxid Redox Signal 2024; 40:40-85. [PMID: 37132605 PMCID: PMC10824235 DOI: 10.1089/ars.2022.0179] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
Significance: The multifactorial nature of the mechanisms implicated in cancer development still represents a major issue for the success of established antitumor therapies. The discovery of ferroptosis, a novel form of programmed cell death distinct from apoptosis, along with the identification of the molecular pathways activated during its execution, has led to the uncovering of novel molecules characterized by ferroptosis-inducing properties. Recent advances: As of today, the ferroptosis-inducing properties of compounds derived from natural sources have been investigated and interesting findings have been reported both in vitro and in vivo. Critical Issues: Despite the efforts made so far, only a limited number of synthetic compounds have been identified as ferroptosis inducers, and their utilization is still limited to basic research. In this review, we analyzed the most important biochemical pathways involved in ferroptosis execution, with particular attention to the newest literature findings on canonical and non-canonical hallmarks, together with mechanisms of action of natural compounds identified as novel ferroptosis inducers. Compounds have been classified based on their chemical structure, and modulation of ferroptosis-related biochemical pathways has been reported. Future Directions: The outcomes herein collected represent a fascinating starting point from which to take hints for future drug discovery studies aimed at identifying ferroptosis-inducing natural compounds for anticancer therapies. Antioxid. Redox Signal. 40, 40-85.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| |
Collapse
|
12
|
Currais A, Raschke W, Maher P. CMS121, a Novel Drug Candidate for the Treatment of Alzheimer's Disease and Age-Related Dementia. J Alzheimers Dis 2024; 101:S179-S192. [PMID: 39422940 DOI: 10.3233/jad-231062] [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] [Indexed: 10/19/2024]
Abstract
Old age is the major risk factor for sporadic Alzheimer's disease (AD). However, old age-related changes in brain physiology have generally not been taken into consideration in developing drug candidates for the treatment of AD. This is at least partly because the role of these age-related processes in the development and progression of AD are still not well understood. Nevertheless, we and others have described an association between the oxytosis/ferroptosis non-apoptotic regulated cell death pathway and aging. Based on this association, we incorporated protection against this pathway as part of a cell-based phenotypic screening approach to identify novel drug candidates for the treatment of AD. Using this approach, we identified the fisetin derivative CMS121 as a potent neuroprotective molecule that is able to maintain cognitive function in multiple pre-clinical models of AD. Furthermore, we identified a key target of CMS121 as fatty acid synthase, a protein which had not been previously considered in the context of AD. Herein, we provide a comprehensive description of the development of CMS121, its preclinical activities, and the results of the toxicology testing that led to its IND approval.
Collapse
Affiliation(s)
| | | | - Pamela Maher
- Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
13
|
Qiao O, Wang X, Wang Y, Li N, Gong Y. Ferroptosis in acute kidney injury following crush syndrome: A novel target for treatment. J Adv Res 2023; 54:211-222. [PMID: 36702249 PMCID: PMC10703611 DOI: 10.1016/j.jare.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/29/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Crush syndrome (CS) is a kind of traumatic and ischemic injury that seriously threatens life after prolonged compression. It is characterized by systemic inflammatory reaction, myoglobinuria, hyperkalemia and acute kidney injury (AKI). Especially AKI, it is the leading cause of death from CS. There are various cell death forms in AKI, among which ferroptosis is a typical form of cell death. However, the role of ferroptosis has not been fully revealed in CS-AKI. AIM OF REVIEW This review aimed to summarize the evidence of ferroptosis in CS-AKI and its related molecular mechanism, discuss the therapeutic significance of ferroptosis in CS-AKI, and open up new ideas for the treatment of CS-AKI. KEY SCIENTIFIC CONCEPTS OF REVIEW One of the main pathological manifestations of CS-AKI is renal tubular epithelial cell dysfunction and cell death, which has been attributed to massive deposition of myoglobin. Large amounts of myoglobin released from damaged muscle deposited in the renal tubules, impeding the normal renal tubules function and directly damaging the tubules with oxidative stress and elevated iron levels. Lipid peroxidation damage and iron overload are the distinguishing features of ferroptosis. Moreover, high levels of pro-inflammatory cytokines and damage-associated molecule pattern molecules (HMGB1, double-strand DNA, and macrophage extracellular trap) in renal tissue have been shown to promote ferroptosis. However, how ferroptosis occurs in CS-AKI and whether it can be a therapeutic target remains unclear. In our current work, we systematically reviewed the occurrence and underlying mechanism of ferroptosis in CS-AKI.
Collapse
Affiliation(s)
- Ou Qiao
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Xinyue Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yuru Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Ning Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China.
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China.
| |
Collapse
|
14
|
Manzano-Covarrubias AL, Yan H, Luu MDA, Gadjdjoe PS, Dolga AM, Schmidt M. Unravelling the signaling power of pollutants. Trends Pharmacol Sci 2023; 44:917-933. [PMID: 37783643 DOI: 10.1016/j.tips.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023]
Abstract
Exposure to environmental pollutants contributes to diverse pathologies, including pulmonary disease, lower respiratory infections, cancer, and stroke. Pollutants' entry can occur through inhalation, traversing endothelial and epithelial barriers, and crossing the blood-brain barrier, leading to a wide distribution throughout the human body via systemic circulation. Pollutants cause cellular damage by multiple mechanisms encompassing oxidative stress, mitochondrial dysfunction, (neuro)inflammation, and protein instability/proteotoxicity. Sensing pollutants has added a new dimension to disease progression and drug failure. Understanding the molecular pathways and potential receptor binding/signaling that underpin 'sensing' could contribute to ways to combat the detrimental effects of pollutants. We highlight key points of pollutant signaling, crosstalk with receptors acting as drug targets for chronic diseases, and discuss the potential for future therapeutics.
Collapse
Affiliation(s)
- Ana L Manzano-Covarrubias
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Minh D A Luu
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Phoeja S Gadjdjoe
- Department of Molecular Pharmacology, University of Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
15
|
Marmolejo-Garza A, Krabbendam IE, Luu MDA, Brouwer F, Trombetta-Lima M, Unal O, O'Connor SJ, Majerníková N, Elzinga CRS, Mammucari C, Schmidt M, Madesh M, Boddeke E, Dolga AM. Negative modulation of mitochondrial calcium uniporter complex protects neurons against ferroptosis. Cell Death Dis 2023; 14:772. [PMID: 38007529 PMCID: PMC10676387 DOI: 10.1038/s41419-023-06290-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
Ferroptosis is an iron- and reactive oxygen species (ROS)-dependent form of regulated cell death, that has been implicated in Alzheimer's disease and Parkinson's disease. Inhibition of cystine/glutamate antiporter could lead to mitochondrial fragmentation, mitochondrial calcium ([Ca2+]m) overload, increased mitochondrial ROS production, disruption of the mitochondrial membrane potential (ΔΨm), and ferroptotic cell death. The observation that mitochondrial dysfunction is a characteristic of ferroptosis makes preservation of mitochondrial function a potential therapeutic option for diseases associated with ferroptotic cell death. Mitochondrial calcium levels are controlled via the mitochondrial calcium uniporter (MCU), the main entry point of Ca2+ into the mitochondrial matrix. Therefore, we have hypothesized that negative modulation of MCU complex may confer protection against ferroptosis. Here we evaluated whether the known negative modulators of MCU complex, ruthenium red (RR), its derivative Ru265, mitoxantrone (MX), and MCU-i4 can prevent mitochondrial dysfunction and ferroptotic cell death. These compounds mediated protection in HT22 cells, in human dopaminergic neurons and mouse primary cortical neurons against ferroptotic cell death. Depletion of MICU1, a [Ca2+]m gatekeeper, demonstrated that MICU is protective against ferroptosis. Taken together, our results reveal that negative modulation of MCU complex represents a therapeutic option to prevent degenerative conditions, in which ferroptosis is central to the progression of these pathologies.
Collapse
Affiliation(s)
- Alejandro Marmolejo-Garza
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge E Krabbendam
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Minh Danh Anh Luu
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Famke Brouwer
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Osman Unal
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Shane J O'Connor
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Naďa Majerníková
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Carolina R S Elzinga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy
| | - Martina Schmidt
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Muniswamy Madesh
- Department of Medicine/Cardiology, Center for Mitochondrial Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Erik Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
16
|
Chen T, Majerníková N, Marmolejo-Garza A, Trombetta-Lima M, Sabogal-Guáqueta AM, Zhang Y, Ten Kate R, Zuidema M, Mulder PPMFA, den Dunnen W, Gosens R, Verpoorte E, Culmsee C, Eisel ULM, Dolga AM. Mitochondrial transplantation rescues neuronal cells from ferroptosis. Free Radic Biol Med 2023; 208:62-72. [PMID: 37536459 DOI: 10.1016/j.freeradbiomed.2023.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Ferroptosis is a type of oxidative cell death that can occur in neurodegenerative diseases and involves damage to mitochondria. Previous studies demonstrated that preventing mitochondrial dysfunction can rescue cells from ferroptotic cell death. However, the complexity of mitochondrial dysfunction and the timing of therapeutic interventions make it difficult to develop an effective treatment strategy against ferroptosis in neurodegeneration conditions. In this study, we explored the use of mitochondrial transplantation as a novel therapeutic approach for preventing ferroptotic neuronal cell death. Our data showed that isolated exogenous mitochondria were incorporated into both healthy and ferroptotic immortalized hippocampal HT-22 cells and primary cortical neurons (PCN). The mitochondrial incorporation was accompanied by increased metabolic activity and cell survival through attenuating lipid peroxidation and mitochondrial superoxide production. Further, the function of mitochondrial complexes I, III and V activities contributed to the neuroprotective activity of exogenous mitochondria. Similarly, we have also showed the internalization of exogenous mitochondria in mouse PCN; these internalized mitochondria were found to effectively preserve the neuronal networks when challenged with ferroptotic stimuli. The administration of exogenous mitochondria into the axonal compartment of a two-compartment microfluidic device induced mitochondrial transportation to the cell body, which prevented fragmentation of the neuronal network in ferroptotic PCN. These findings suggest that mitochondria transplantation may be a promising therapeutic approach for protecting neuronal cells from ferroptotic cell death.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Nad'a Majerníková
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands; Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Alejandro Marmolejo-Garza
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands; Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marina Trombetta-Lima
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands; Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Angélica María Sabogal-Guáqueta
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Yuequ Zhang
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Ruth Ten Kate
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Minte Zuidema
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Patty P M F A Mulder
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Wilfred den Dunnen
- Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Elisabeth Verpoorte
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
17
|
Filippenkov IB, Khrunin AV, Mozgovoy IV, Dergunova LV, Limborska SA. Are Ischemic Stroke and Alzheimer's Disease Genetically Consecutive Pathologies? Biomedicines 2023; 11:2727. [PMID: 37893101 PMCID: PMC10604604 DOI: 10.3390/biomedicines11102727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Complex diseases that affect the functioning of the central nervous system pose a major problem for modern society. Among these, ischemic stroke (IS) holds a special place as one of the most common causes of disability and mortality worldwide. Furthermore, Alzheimer's disease (AD) ranks first among neurodegenerative diseases, drastically reducing brain activity and overall life quality and duration. Recent studies have shown that AD and IS share several common risk and pathogenic factors, such as an overlapping genomic architecture and molecular signature. In this review, we will summarize the genomics and RNA biology studies of IS and AD, discussing the interconnected nature of these pathologies. Additionally, we highlight specific genomic points and RNA molecules that can serve as potential tools in predicting the risks of diseases and developing effective therapies in the future.
Collapse
Affiliation(s)
| | | | | | | | - Svetlana A. Limborska
- Laboratory of Human Molecular Genetics, National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, 123182 Moscow, Russia (A.V.K.); (I.V.M.); (L.V.D.)
| |
Collapse
|
18
|
Li N, Duan YH, Chen L, Zhang K. Iron metabolism: An emerging therapeutic target underlying the anti-Alzheimer's disease effect of ginseng. J Trace Elem Med Biol 2023; 79:127252. [PMID: 37418790 DOI: 10.1016/j.jtemb.2023.127252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 06/05/2023] [Accepted: 06/22/2023] [Indexed: 07/09/2023]
Abstract
Finding neuroprotective drugs with fewer side effects and more efficacy has become a major problem as the global prevalence of Alzheimer's disease (AD) rises. Natural drugs have risen to prominence as potential medication candidates. Ginseng has a long history of use in China, and it has a wide range of pharmacological actions that can help with neurological issues. Iron loaded in the brain has been linked to AD pathogenesis. We reviewed the regulation of iron metabolism and its studies in AD and explored how ginseng might regulate iron metabolism and prevent or treat AD. Researchers utilized network pharmacology analysis to identify key factive components of ginseng that protect against AD by regulating ferroptosis. Ginseng and its active ingredients may benefit AD by regulating iron metabolism and targeting ferroptosis genes to inhibit the ferroptosis process. The results present new ideas for ginseng pharmacological studies and initiatives for further research into AD-related drugs. To provide comprehensive information on the neuroprotective use of ginseng to modulate iron metabolism, reveal its potential to treat AD, and provide insights for future research opportunities.
Collapse
Affiliation(s)
- Nan Li
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Yu-Han Duan
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Lei Chen
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Kun Zhang
- Department of Medical Research Center, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
19
|
Zhang T, Luu MDA, Dolga AM, Eisel ULM, Schmidt M. The old second messenger cAMP teams up with novel cell death mechanisms: potential translational therapeutical benefit for Alzheimer's disease and Parkinson's disease. Front Physiol 2023; 14:1207280. [PMID: 37405135 PMCID: PMC10315612 DOI: 10.3389/fphys.2023.1207280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/07/2023] [Indexed: 07/06/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) represent the most prevalent neurodegenerative disorders severely impacting life expectancy and quality of life of millions of people worldwide. AD and PD exhibit both a very distinct pathophysiological disease pattern. Intriguingly, recent researches, however, implicate that overlapping mechanisms may underlie AD and PD. In AD and PD, novel cell death mechanisms, encompassing parthanatos, netosis, lysosome-dependent cell death, senescence and ferroptosis, apparently rely on the production of reactive oxygen species, and seem to be modulated by the well-known, "old" second messenger cAMP. Signaling of cAMP via PKA and Epac promotes parthanatos and induces lysosomal cell death, while signaling of cAMP via PKA inhibits netosis and cellular senescence. Additionally, PKA protects against ferroptosis, whereas Epac1 promotes ferroptosis. Here we review the most recent insights into the overlapping mechanisms between AD and PD, with a special focus on cAMP signaling and the pharmacology of cAMP signaling pathways.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Minh D. A. Luu
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Ulrich L. M. Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
20
|
Sun 孙意冉 Y, Yan C, He L, Xiang S, Wang P, Li Z, Chen Y, Zhao J, Yuan Y, Wang W, Zhang X, Su P, Su Y, Ma J, Xu J, Peng Q, Ma H, Xie Z, Zhang Z. Inhibition of ferroptosis through regulating neuronal calcium homeostasis: An emerging therapeutic target for Alzheimer's disease. Ageing Res Rev 2023; 87:101899. [PMID: 36871781 DOI: 10.1016/j.arr.2023.101899] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD), a chronic and progressive neurodegenerative disease, generates a serious threat to the health of the elderly. The AD brain is microscopically characterized by amyloid plaques and neurofibrillary tangles. There are still no effective therapeutic drugs to restrain the progression of AD though much attention has been paid to exploit AD treatments. Ferroptosis, a type of programmed cell death, has been reported to promote the pathological occurrence and development of AD, and inhibition of neuronal ferroptosis can effectively improve the cognitive impairment of AD. Studies have shown that calcium (Ca2+) dyshomeostasis is closely related to the pathology of AD, and can drive the occurrence of ferroptosis through several pathways, such as interacting with iron, and regulating the crosstalk between endoplasmic reticulum (ER) and mitochondria. This paper mainly reviews the roles of ferroptosis and Ca2+ in the pathology of AD, and highlights that restraining ferroptosis through maintaining the homeostasis of Ca2+ may be an innovative target for the treatment of AD.
Collapse
Affiliation(s)
- Yiran Sun 孙意冉
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Chenchen Yan
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Libo He
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China
| | - Shixie Xiang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Pan Wang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Zhonghua Li
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yuanzhao Chen
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jie Zhao
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ye Yuan
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Wang Wang
- School of basic medicine, Nanchang Medical College, Nanchang 330052, Jiangxi, China
| | - Xiaowei Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Pan Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yunfang Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jinlian Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jiangyan Xu
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Quekun Peng
- School of Biosciences and Technology, Chengdu Medical College, Chengdu 610500, China.
| | - Huifen Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhishen Xie
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhenqiang Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
21
|
Intranasal Lipid Nanoparticles Containing Bioactive Compounds Obtained from Marine Sources to Manage Neurodegenerative Diseases. Pharmaceuticals (Basel) 2023. [DOI: 10.3390/ph16020311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] 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
|
22
|
Coradduzza D, Congiargiu A, Chen Z, Zinellu A, Carru C, Medici S. Ferroptosis and Senescence: A Systematic Review. Int J Mol Sci 2023; 24:ijms24043658. [PMID: 36835065 PMCID: PMC9963234 DOI: 10.3390/ijms24043658] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Senescence is a cellular aging process in all multicellular organisms. It is characterized by a decay in cellular functions and proliferation, resulting in increased cellular damage and death. This condition plays an essential role in the aging process and significantly contributes to the development of age-related complications. On the other hand, ferroptosis is a systemic cell death pathway characterized by excessive iron accumulation followed by the generation of reactive oxygen species (ROS). Oxidative stress is a common trigger of this condition and may be induced by various factors such as toxins, drugs, and inflammation. Ferroptosis is linked to numerous disorders, including cardiovascular disease, neurodegeneration, and cancer. Senescence is believed to contribute to the decay in tissue and organ functions occurring with aging. It has also been linked to the development of age-related pathologies, such as cardiovascular diseases, diabetes, and cancer. In particular, senescent cells have been shown to produce inflammatory cytokines and other pro-inflammatory molecules that can contribute to these conditions. In turn, ferroptosis has been linked to the development of various health disorders, including neurodegeneration, cardiovascular disease, and cancer. Ferroptosis is known to play a role in the development of these pathologies by promoting the death of damaged or diseased cells and contributing to the inflammation often associated. Both senescence and ferroptosis are complex pathways that are still not fully understood. Further research is needed to thoroughly investigate the role of these processes in aging and disease, and to identify potential interventions to target such processes in order to prevent or treat age-related conditions. This systematic review aims to assess the potential mechanisms underlying the link connecting senescence, ferroptosis, aging, and disease, and whether they can be exploited to block or limit the decay of the physiological functions in elderly people for a healthy longevity.
Collapse
Affiliation(s)
| | | | - Zhichao Chen
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Control Quality Unit, Azienda-Ospedaliera Universitaria (AOU), 07100 Sassari, Italy
- Correspondence:
| | - Serenella Medici
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
23
|
Yang L, Nao J. Ferroptosis: a potential therapeutic target for Alzheimer's disease. Rev Neurosci 2022:revneuro-2022-0121. [PMID: 36514247 DOI: 10.1515/revneuro-2022-0121] [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: 10/03/2022] [Accepted: 10/30/2022] [Indexed: 12/15/2022]
Abstract
The most prevalent dementia-causing neurodegenerative condition is Alzheimer's disease (AD). The aberrant buildup of amyloid β and tau hyperphosphorylation are the two most well-known theories about the mechanisms underlying AD development. However, a significant number of pharmacological clinical studies conducted around the world based on the two aforementioned theories have not shown promising outcomes, and AD is still not effectively treated. Ferroptosis, a non-apoptotic programmed cell death defined by the buildup of deadly amounts of iron-dependent lipid peroxides, has received more attention in recent years. A wealth of data is emerging to support the role of iron in the pathophysiology of AD. Cell line and animal studies applying ferroptosis modulators to the treatment of AD have shown encouraging results. Based on these studies, we describe in this review the underlying mechanisms of ferroptosis; the role that ferroptosis plays in AD pathology; and summarise some of the research advances in the treatment of AD with ferroptosis modulators. We hope to contribute to the clinical management of AD.
Collapse
Affiliation(s)
- Lan Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
24
|
Blood Analytes as Biomarkers of Mechanisms Involved in Alzheimer’s Disease Progression. Int J Mol Sci 2022; 23:ijms232113289. [DOI: 10.3390/ijms232113289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia, but the pathogenetic factors are not yet well known, and the relationships between brain and systemic biochemical derangements and disease onset and progression are unclear. We aim to focus on blood biomarkers for an accurate prognosis of the disease. We used a dataset characterized by longitudinal findings collected over the past 10 years from 90 AD patients. The dataset included 277 observations (both clinical and biochemical ones, encompassing blood analytes encompassing routine profiles for different organs, together with immunoinflammatory and oxidative markers). Subjects were grouped into four severity classes according to the Clinical Dementia Rating (CDR) Scale: mild (CDR = 0.5 and CDR = 1), moderate (CDR = 2), severe (CDR = 3) and very severe (CDR = 4 and CDR = 5). Statistical models were used for the identification of potential blood markers of AD progression. Moreover, we employed the Pathfinder tool of the Reactome database to investigate the biological pathways in which the analytes of interest could be involved. Statistical results reveal an inverse significant relation between four analytes (high-density cholesterol, total cholesterol, iron and ferritin) with AD severity. In addition, the Reactome database suggests that such analytes could be involved in pathways that are altered in AD progression. Indeed, the identified blood markers include molecules that reflect the heterogeneous pathogenetic mechanisms of AD. The combination of such blood analytes might be an early indicator of AD progression and constitute useful therapeutic targets.
Collapse
|
25
|
Li Y, Zhao Y, Li X, Zhai L, Zheng H, Yan Y, Fu Q, Ma J, Fu H, Zhang Z, Li Z. Biological and therapeutic role of LSD1 in Alzheimer’s diseases. Front Pharmacol 2022; 13:1020556. [PMID: 36386192 PMCID: PMC9640401 DOI: 10.3389/fphar.2022.1020556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/13/2022] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) is a common chronic neurodegenerative disease characterized by cognitive learning and memory impairments, however, current treatments only provide symptomatic relief. Lysine-specific demethylase 1 (LSD1), regulating the homeostasis of histone methylation, plays an important role in the pathogenesis of many neurodegenerative disorders. LSD1 functions in regulating gene expression via transcriptional repression or activation, and is involved in initiation and progression of AD. Pharmacological inhibition of LSD1 has shown promising therapeutic benefits for AD treatment. In this review, we attempt to elaborate on the role of LSD1 in some aspects of AD including neuroinflammation, autophagy, neurotransmitters, ferroptosis, tau protein, as well as LSD1 inhibitors under clinical assessments for AD treatment.
Collapse
Affiliation(s)
- Yu Li
- Department of Pharmacy, Yellow River Central Hospital of Yellow River Conservancy Commission, Zhengzhou, China
| | - Yuanyuan Zhao
- Department of Pharmacy, Yellow River Central Hospital of Yellow River Conservancy Commission, Zhengzhou, China
| | - Xiaona Li
- Department of Pharmacy, Yellow River Central Hospital of Yellow River Conservancy Commission, Zhengzhou, China
| | - Liuqun Zhai
- Department of Pharmacy, Yellow River Central Hospital of Yellow River Conservancy Commission, Zhengzhou, China
| | - Hua Zheng
- Department of Pharmacy, Yellow River Central Hospital of Yellow River Conservancy Commission, Zhengzhou, China
| | - Ying Yan
- Department of Pharmacy, Yellow River Central Hospital of Yellow River Conservancy Commission, Zhengzhou, China
| | - Qiang Fu
- Department of Pharmacy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinlian Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Haier Fu
- Department of Pharmacy, Yellow River Central Hospital of Yellow River Conservancy Commission, Zhengzhou, China
- *Correspondence: Haier Fu, ; Zhenqiang Zhang, ; Zhonghua Li,
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- *Correspondence: Haier Fu, ; Zhenqiang Zhang, ; Zhonghua Li,
| | - Zhonghua Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- *Correspondence: Haier Fu, ; Zhenqiang Zhang, ; Zhonghua Li,
| |
Collapse
|
26
|
Edgerton-Fulton M, Ergul A. Vascular contributions to cognitive impairment/dementia in diabetes: role of endothelial cells and pericytes. Am J Physiol Cell Physiol 2022; 323:C1177-C1189. [PMID: 36036445 PMCID: PMC9576164 DOI: 10.1152/ajpcell.00072.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/19/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022]
Abstract
Vascular contributions to cognitive impairment/dementia (VCID) are a leading cause of dementia, a known neurodegenerative disorder characterized by progressive cognitive decline. Although diabetes increases the risks of stroke and the development of cerebrovascular disease, the cellular and vascular mechanisms that lead to VCID in diabetes are yet to be determined. A growing body of research has identified that cerebrovascular cells within the neurovascular complex display an array of cellular responses that impact their survival and reparative properties, which plays a significant role in VCID development. Specifically, endothelial cells and pericytes are the primary cell types that have gained much attention in dementia-related studies due to their molecular and phenotypic heterogeneity. In this review, we will discuss the various morphological subclasses of endothelial cells and pericytes as well as their relative distribution throughout the cerebrovasculature. Furthermore, the use of diabetic and stroke animal models in preclinical studies has provided more insight into the impact of sex differences on cerebral vascularization in progressive VCID. Understanding how cellular responses and sex differences contribute to endothelial cell and pericyte survival and function will set the stage for the development of potential preventive therapies for dementia-related disorders in diabetes.
Collapse
Affiliation(s)
- Mia Edgerton-Fulton
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Adviye Ergul
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
27
|
Deng Y, Feng Y, Lv Z, He J, Chen X, Wang C, Yuan M, Xu T, Gao W, Chen D, Zhu H, Hou D. Machine learning models identify ferroptosis-related genes as potential diagnostic biomarkers for Alzheimer’s disease. Front Aging Neurosci 2022; 14:994130. [PMID: 36262887 PMCID: PMC9575464 DOI: 10.3389/fnagi.2022.994130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a complex, and multifactorial neurodegenerative disease. Previous studies have revealed that oxidative stress, synaptic toxicity, autophagy, and neuroinflammation play crucial roles in the progress of AD, however, its pathogenesis is still unclear. Recent researches have indicated that ferroptosis, an iron-dependent programmed cell death, might be involved in the pathogenesis of AD. Therefore, we aim to screen correlative ferroptosis-related genes (FRGs) in the progress of AD to clarify insights into the diagnostic value. Interestingly, we identified eight FRGs were significantly differentially expressed in AD patients. 10,044 differentially expressed genes (DEGs) were finally identified by differential expression analysis. The following step was investigating the function of DEGs using gene set enrichment analysis (GSEA). Weight gene correlation analysis was performed to explore ten modules and 104 hub genes. Subsequently, based on machine learning algorithms, we constructed diagnostic classifiers to select characteristic genes. Through the multivariable logistic regression analysis, five features (RAF1, NFKBIA, MOV10L1, IQGAP1, FOXO1) were then validated, which composed a diagnostic model of AD. Thus, our findings not only developed genetic diagnostics strategy, but set a direction for further study of the disease pathogenesis and therapy targets.
Collapse
Affiliation(s)
- Yanyao Deng
- Department of Rehabilitation, The First Hospital of Changsha, Changsha, China
| | - Yanjin Feng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Lv
- Department of Neurosurgery, The First People’s Hospital of Chenzhou, Chenzhou, China
| | - Jinli He
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xun Chen
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chen Wang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mingyang Yuan
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ting Xu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenzhe Gao
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dongjie Chen
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongwei Zhu
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Deren Hou
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Deren Hou,
| |
Collapse
|
28
|
Wang X, Tian Y, Li C, Chen M. Exploring the key ferroptosis-related gene in the peripheral blood of patients with Alzheimer’s disease and its clinical significance. Front Aging Neurosci 2022; 14:970796. [PMID: 36118694 PMCID: PMC9475071 DOI: 10.3389/fnagi.2022.970796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction Alzheimer’s disease (AD) is the most common type of dementia, and there is growing evidence suggesting that ferroptosis is involved in its pathogenesis. In this study, we aimed to investigate the key ferroptosis-related genes in AD and identify a novel ferroptosis-related gene diagnosis model for patients with AD. Materials and methods We extracted the human blood and hippocampus gene expression data of five datasets (GSE63060, GSE63061, GSE97760, GSE48350, and GSE5281) in the Gene Expression Omnibus database as well as the ferroptosis-related genes from FerrDb. Differentially expressed ferroptosis-related genes were screened by random forest classifier, and were further used to construct a diagnostic model of AD using an artificial neural network. The patterns of immune infiltration in the peripheral immune system of AD were also investigated using the CIBERSORT algorithm. Results We first screened and identified 12 ferroptosis-related genes (ATG3, BNIP3, DDIT3, FH, GABARAPL1, MAPK14, SOCS1, SP1, STAT3, TNFAIP3, UBC, and ULK) via a random forest classifier, which was differentially expressed between the AD and normal control groups. Based on the 12 hub genes, we successfully constructed a satisfactory diagnostic model for differentiating AD patients from normal controls using an artificial neural network and validated its diagnostic efficacy in several external datasets. Further, the key ferroptosis-related genes were found to be strongly correlated to immune cells infiltration in AD. Conclusion We successfully identified 12 ferroptosis-related genes and established a novel diagnostic model of significant predictive value for AD. These results may help understand the role of ferroptosis in AD pathogenesis and provide promising therapeutic strategies for patients with AD.
Collapse
Affiliation(s)
- Xiaonan Wang
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yaotian Tian
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chunmei Li
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Chen
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Min Chen,
| |
Collapse
|
29
|
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: 47] [Impact Index Per Article: 15.7] [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
|
30
|
Sfera A, Thomas KG, Andronescu CV, Jafri N, Sfera DO, Sasannia S, Zapata-Martín del Campo CM, Maldonado JC. Bromodomains in Human-Immunodeficiency Virus-Associated Neurocognitive Disorders: A Model of Ferroptosis-Induced Neurodegeneration. Front Neurosci 2022; 16:904816. [PMID: 35645713 PMCID: PMC9134113 DOI: 10.3389/fnins.2022.904816] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) comprise a group of illnesses marked by memory and behavioral dysfunction that can occur in up to 50% of HIV patients despite adequate treatment with combination antiretroviral drugs. Iron dyshomeostasis exacerbates HIV-1 infection and plays a major role in Alzheimer's disease pathogenesis. In addition, persons living with HIV demonstrate a high prevalence of neurodegenerative disorders, indicating that HAND provides a unique opportunity to study ferroptosis in these conditions. Both HIV and combination antiretroviral drugs increase the risk of ferroptosis by augmenting ferritin autophagy at the lysosomal level. As many viruses and their proteins exit host cells through lysosomal exocytosis, ferroptosis-driving molecules, iron, cathepsin B and calcium may be released from these organelles. Neurons and glial cells are highly susceptible to ferroptosis and neurodegeneration that engenders white and gray matter damage. Moreover, iron-activated microglia can engage in the aberrant elimination of viable neurons and synapses, further contributing to ferroptosis-induced neurodegeneration. In this mini review, we take a closer look at the role of iron in the pathogenesis of HAND and neurodegenerative disorders. In addition, we describe an epigenetic compensatory system, comprised of bromodomain-containing protein 4 (BRD4) and microRNA-29, that may counteract ferroptosis by activating cystine/glutamate antiporter, while lowering ferritin autophagy and iron regulatory protein-2. We also discuss potential interventions for lysosomal fitness, including ferroptosis blockers, lysosomal acidification, and cathepsin B inhibitors to achieve desirable therapeutic effects of ferroptosis-induced neurodegeneration.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
- Department of Psychiatry, University of California, Riverside, Riverside, CA, United States
| | | | | | - Nyla Jafri
- Patton State Hospital, San Bernardino, CA, United States
| | - Dan O. Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | | | | | - Jose C. Maldonado
- Department of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| |
Collapse
|