1
|
Won JP, Lee HG, Yoon HJ, Seo HG. Biochanin A-mediated anti-ferroptosis is associated with reduction of septic kidney injury. Life Sci 2024; 358:123124. [PMID: 39396639 DOI: 10.1016/j.lfs.2024.123124] [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: 07/03/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
AIMS This study aimed to investigate the therapeutic potential of biochanin A in a sepsis associated- acute kidney injury (SA-AKI) mouse model induced by lipopolysaccharide (LPS). MAIN METHODS Male BALB/C mice (n = 7 per group) were injected with biochanin A (40 mg/kg, i.p.) or ferrostatin-1 (5 mg/kg, i.p.) in the presence or absence of LPS (10 mg/kg, i.p.). Survival rates were monitored twice a day for up to 2 weeks. Morphologic and functional changes in kidney tissue were assessed by H&E staining and by analyzing of levels of blood-urea nitrogen (BUN) and creatinine (CR) in serum, respectively. Kidney epithelial cell death was analyzed by TUNEL staining, Prussian blue staining, iron quantification, lipid peroxide quantification, and glutathione quantification. Anti-ferroptosis mechanism of biochanin A was analyzed by RNA sequencing in mouse embryonic fibroblast cells. KEY FINDINGS Biochanin A increased the survival rate of septic mice and inhibited the secretion of high mobility group box 1, an important inflammatory mediator in sepsis. Biochanin A inhibited LPS-induced kidney damage by suppressing dilatation and kidney tubular epithelial cell death. Furthermore, serum levels of BUN and CR were reduced in biochanin A-treated endotoxemic mice. Biochanin A inhibited the accumulation of iron and lipid peroxide and prevented glutathione depletion in the kidney tissue. Also, nine genes associated with the anti-ferroptosis effects of biochanin A were identified by RNA sequencing analysis. SIGNIFICANCE The present study suggests that biochanin A is an effective inhibitor of ferroptosis, representing a potential treatment or prophylactic for sepsis-related disorders such as SA-AKI.
Collapse
Affiliation(s)
- Jun Pil Won
- Department of Food Science and Biotechnology of Animal Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyuk Gyoon Lee
- Department of Food Science and Biotechnology of Animal Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Han Jun Yoon
- Department of Food Science and Biotechnology of Animal Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Han Geuk Seo
- Department of Food Science and Biotechnology of Animal Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
2
|
Tripathy RK, Frohock Z, Wang H, Cary GA, Keegan S, Carter GW, Li Y. An explainable graph neural network approach for effectively integrating multi-omics with prior knowledge to identify biomarkers from interacting biological domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609465. [PMID: 39253523 PMCID: PMC11383059 DOI: 10.1101/2024.08.23.609465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
The rapid growth of multi-omics datasets, in addition to the wealth of existing biological prior knowledge, necessitates the development of effective methods for their integration. Such methods are essential for building predictive models and identifying disease-related molecular markers. We propose a framework for supervised integration of multi-omics data with biological priors represented as knowledge graphs. Our framework leverages graph neural networks (GNNs) to model the relationships among features from high-dimensional 'omics data and set transformers to integrate low-dimensional representations of 'omics features. Furthermore, our framework incorporates explainability methods to elucidate important biomarkers and extract interaction relationships between biological quantities of interest. We demonstrate the effectiveness of our approach by applying it to Alzheimer's disease (AD) multi-omics data from the ROSMAP cohort, showing that the integration of transcriptomics and proteomics data with AD biological domain network priors improves the prediction accuracy of AD status and highlights functional AD biomarkers.
Collapse
Affiliation(s)
| | - Zachary Frohock
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hong Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | | | | | - Yi Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| |
Collapse
|
3
|
Padalko V, Posnik F, Adamczyk M. Mitochondrial Aconitase and Its Contribution to the Pathogenesis of Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9950. [PMID: 39337438 PMCID: PMC11431987 DOI: 10.3390/ijms25189950] [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: 08/06/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
This survey reviews modern ideas on the structure and functions of mitochondrial and cytosolic aconitase isoenzymes in eukaryotes. Cumulative experimental evidence about mitochondrial aconitases (Aco2) as one of the main targets of reactive oxygen and nitrogen species is generalized. The important role of Aco2 in maintenance of homeostasis of the intracellular iron pool and maintenance of the mitochondrial DNA is discussed. The role of Aco2 in the pathogenesis of some neurodegenerative diseases is highlighted. Inactivation or dysfunction of Aco2 as well as mutations found in the ACO2 gene appear to be significant factors in the development and promotion of various types of neurodegenerative diseases. A restoration of efficient mitochondrial functioning as a source of energy for the cell by targeting Aco2 seems to be one of the promising therapeutic directions to minimize progressive neurodegenerative disorders.
Collapse
Affiliation(s)
- Volodymyr Padalko
- Laboratory of Systems and Synthetic Biology, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
- School of Medicine, V. N. Karazin Kharkiv National University, 61022 Kharkiv, Ukraine
| | - Filip Posnik
- Laboratory of Systems and Synthetic Biology, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Malgorzata Adamczyk
- Laboratory of Systems and Synthetic Biology, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| |
Collapse
|
4
|
Li C, Cui K, Zhu X, Wang S, Yang Q, Fang G. 8-weeks aerobic exercise ameliorates cognitive deficit and mitigates ferroptosis triggered by iron overload in the prefrontal cortex of APP Swe/ PSEN 1dE9 mice through Xc -/GPx4 pathway. Front Neurosci 2024; 18:1453582. [PMID: 39315073 PMCID: PMC11417105 DOI: 10.3389/fnins.2024.1453582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Background Alzheimer's disease (AD) is a degenerative disorder of the central nervous system characterized by notable pathological features such as neurofibrillary tangles and amyloid beta deposition. Additionally, the significant iron accumulation in the brain is another important pathological hallmark of AD. Exercise can play a positive role in ameliorating AD, but the mechanism is unclear. The purpose of the study is to explore the effect of regular aerobic exercise iron homeostasis and lipid antioxidant pathway regarding ferroptosis in the prefrontal cortex (PFC) of APP Swe/PSEN 1dE9 (APP/PS1) mice. Methods Eighty 6-month-old C57BL/6 J and APP/PS1 mice were divided equally into 8-weeks aerobic exercise groups and sedentary groups. Subsequently, Y-maze, Morris water maze test, iron ion detection by probe, Western Blot, ELISA, RT-qPCR, HE, Nissle, Prussian Blue, IHC, IF, and FJ-C staining experiments were conducted to quantitatively assess the behavioral performance, iron levels, iron-metabolism-related proteins, lipid antioxidant-related proteins and morphology in each group of mice. Results In APP/PS1 mice, the increase in heme input proteins and heme oxygenase lead to the elevated levels of free iron in the PFC. The decrease in ferritin content by ferritin autophagy fails to meet the storage needs for excess free iron within the nerve cells. Ultimately, the increase of free ferrous iron triggers the Fenton reaction, may lead to ferroptosis and resulting in cognitive impairment in APP/PS1 mice. However, 8-weeks aerobic exercise induce upregulation of the Xc-/GPx4 pathway, which can reverse the lipid peroxidation process, thereby inhibiting ferroptosis in APP/PS1 mice. Conclusion 8 weeks aerobic exercise can improve learning and memory abilities in AD, upregulate GPx4/Xc- pathway in PFC to reduce ferroptosis induced by AD.
Collapse
Affiliation(s)
- Chaoyang Li
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Kaiyin Cui
- Sport Science School, Beijing Sport University, Beijing, China
| | - Xinyuan Zhu
- Department of Medical Supervision, China National Institute of Sports Medicine, Beijing, China
| | - Shufan Wang
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Qing Yang
- National Fitness and Scientific Exercise Research Center, China Institute of Sport Science, Beijing, China
| | - Guoliang Fang
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
5
|
Lin N, Gao XY, Li X, Chu WM. Involvement of ubiquitination in Alzheimer's disease. Front Neurol 2024; 15:1459678. [PMID: 39301473 PMCID: PMC11412110 DOI: 10.3389/fneur.2024.1459678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024] Open
Abstract
The hallmark pathological features of Alzheimer's disease (AD) consist of senile plaques, which are formed by extracellular β-amyloid (Aβ) deposition, and neurofibrillary tangles, which are formed by the hyperphosphorylation of intra-neuronal tau proteins. With the increase in clinical studies, the in vivo imbalance of iron homeostasis and the dysfunction of synaptic plasticity have been confirmed to be involved in AD pathogenesis. All of these mechanisms are constituted by the abnormal accumulation of misfolded or conformationally altered protein aggregates, which in turn drive AD progression. Proteostatic imbalance has emerged as a key mechanism in the pathogenesis of AD. Ubiquitination modification is a major pathway for maintaining protein homeostasis, and protein degradation is primarily carried out by the ubiquitin-proteasome system (UPS). In this review, we provide an overview of the ubiquitination modification processes and related protein ubiquitination degradation pathways in AD, focusing on the microtubule-associated protein Tau, amyloid precursor protein (APP), divalent metal transporter protein 1 (DMT1), and α-amino-3-hyroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors. We also discuss recent advances in ubiquitination-based targeted therapy for AD, with the aim of contributing new ideas to the development of novel therapeutic interventions for AD.
Collapse
Affiliation(s)
- Nan Lin
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xi-Yan Gao
- The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiao Li
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wen-Ming Chu
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Spence H, Mengoa-Fleming S, Sneddon AA, McNeil CJ, Waiter GD. Associations between sex, systemic iron and inflammatory status and subcortical brain iron. Eur J Neurosci 2024; 60:5069-5085. [PMID: 39113267 DOI: 10.1111/ejn.16467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
Brain iron increases in several neurodegenerative diseases are associated with disease progression. However, the causes of increased brain iron remain unclear. This study investigates relationships between subcortical iron, systemic iron and inflammatory status. Brain magnetic resonance imaging (MRI) scans and blood plasma samples were collected from cognitively healthy females (n = 176, mean age = 61.4 ± 4.5 years, age range = 28-72 years) and males (n = 152, mean age = 62.0 ± 5.1 years, age range = 32-74 years). Regional brain iron was quantified using quantitative susceptibility mapping. To assess systemic iron, haematocrit, ferritin and soluble transferrin receptor were measured, and total body iron index was calculated. To assess systemic inflammation, C-reactive protein (CRP), neutrophil:lymphocyte ratio (NLR), macrophage colony-stimulating factor 1 (MCSF), interleukin 6 (IL6) and interleukin 1β (IL1β) were measured. We demonstrated that iron levels in the right hippocampus were higher in males compared with females, while iron in the right caudate was higher in females compared with males. There were no significant associations observed between subcortical iron levels and blood markers of iron and inflammatory status indicating that such blood measures are not markers of brain iron. These results suggest that brain iron may be regulated independently of blood iron and so directly targeting global iron change in the treatment of neurodegenerative disease may have differential impacts on blood and brain iron.
Collapse
Affiliation(s)
- Holly Spence
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Stephanie Mengoa-Fleming
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Christopher J McNeil
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Gordon D Waiter
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
7
|
Petralla S, Saveleva L, Kanninen KM, Oster JS, Panayotova M, Fricker G, Puris E. Increased Expression of Transferrin Receptor 1 in the Brain Cortex of 5xFAD Mouse Model of Alzheimer's Disease Is Associated with Activation of HIF-1 Signaling Pathway. Mol Neurobiol 2024; 61:6383-6394. [PMID: 38296900 PMCID: PMC11339108 DOI: 10.1007/s12035-024-03990-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/24/2024] [Indexed: 02/02/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Despite intensive research efforts, there are currently no effective treatments to cure and prevent AD. There is growing evidence that dysregulation of iron homeostasis may contribute to the pathogenesis of AD. Given the important role of the transferrin receptor 1 (TfR1) in regulating iron distribution in the brain, as well as in the drug delivery, we investigated its expression in the brain cortex and isolated brain microvessels from female 8-month-old 5xFAD mice mimicking advanced stage of AD. Moreover, we explored the association between the TfR1 expression and the activation of the HIF-1 signaling pathway, as well as oxidative stress and inflammation in 5xFAD mice. Finally, we studied the impact of Aβ1-40 and Aβ1-42 on TfR1 expression in the brain endothelial cell line hCMEC/D3. In the present study, we revealed that an increase in TfR1 protein levels observed in the brain cortex of 5xFAD mice was associated with activation of the HIF-1 signaling pathway as well as accompanied by oxidative stress and inflammation. Interestingly, incubation of Aβ peptides in hCMEC/D3 cells did not affect the expression of TfR1, which supported our findings of unaltered TfR1 expression in the isolated brain microvessels in 5xFAD mice. In conclusion, the study provides important information about the expression of TfR1 in the 5xFAD mouse model and the potential role of HIF-1 signaling pathway in the regulation of TfR1 in AD, which could represent a promising strategy for the development of therapies for AD.
Collapse
Affiliation(s)
- Sabrina Petralla
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Julia S Oster
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Maria Panayotova
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany
| | - Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120, Heidelberg, Germany.
| |
Collapse
|
8
|
Faraji P, Kühn H, Ahmadian S. Multiple Roles of Apolipoprotein E4 in Oxidative Lipid Metabolism and Ferroptosis During the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2024; 74:62. [PMID: 38958788 PMCID: PMC11222241 DOI: 10.1007/s12031-024-02224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/14/2024] [Indexed: 07/04/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide and has a great socio-economic impact. Modified oxidative lipid metabolism and dysregulated iron homeostasis have been implicated in the pathogenesis of this disorder, but the detailed pathophysiological mechanisms still remain unclear. Apolipoprotein E (APOE) is a lipid-binding protein that occurs in large quantities in human blood plasma, and a polymorphism of the APOE gene locus has been identified as risk factors for AD. The human genome involves three major APOE alleles (APOE2, APOE3, APOE4), which encode for three subtly distinct apolipoprotein E isoforms (APOE2, APOE3, APOE4). The canonic function of these apolipoproteins is lipid transport in blood and brain, but APOE4 allele carriers have a much higher risk for AD. In fact, about 60% of clinically diagnosed AD patients carry at least one APOE4 allele in their genomes. Although the APOE4 protein has been implicated in pathophysiological key processes of AD, such as extracellular beta-amyloid (Aβ) aggregation, mitochondrial dysfunction, neuroinflammation, formation of neurofibrillary tangles, modified oxidative lipid metabolism, and ferroptotic cell death, the underlying molecular mechanisms are still not well understood. As for all mammalian cells, iron plays a crucial role in neuronal functions and dysregulation of iron homeostasis has also been implicated in the pathogenesis of AD. Imbalances in iron homeostasis and impairment of the hydroperoxy lipid-reducing capacity induce cellular dysfunction leading to neuronal ferroptosis. In this review, we summarize the current knowledge on APOE4-related oxidative lipid metabolism and the potential role of ferroptosis in the pathogenesis of AD. Pharmacological interference with these processes might offer innovative strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Parisa Faraji
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Hartmut Kühn
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
9
|
Kostenko A, Zuffa S, Zhi H, Mildau K, Raffatellu M, Dorrestein PC, Aron AT. Dietary iron intake has long-term effects on the fecal metabolome and microbiome. Metallomics 2024; 16:mfae033. [PMID: 38992131 PMCID: PMC11272056 DOI: 10.1093/mtomcs/mfae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024]
Abstract
Iron is essential for life, but its imbalances can lead to severe health implications. Iron deficiency is the most common nutrient disorder worldwide, and iron dysregulation in early life has been found to cause long-lasting behavioral, cognitive, and neural effects. However, little is known about the effects of dietary iron on gut microbiome function and metabolism. In this study, we sought to investigate the impact of dietary iron on the fecal metabolome and microbiome by using mice fed with three diets with different iron content: an iron deficient, an iron sufficient (standard), and an iron overload diet for 7 weeks. Additionally, we sought to understand whether any observed changes would persist past the 7-week period of diet intervention. To assess this, all feeding groups were switched to a standard diet, and this feeding continued for an additional 7 weeks. Analysis of the fecal metabolome revealed that iron overload and deficiency significantly alter levels of peptides, nucleic acids, and lipids, including di- and tri-peptides containing branched-chain amino acids, inosine and guanosine, and several microbial conjugated bile acids. The observed changes in the fecal metabolome persist long after the switch back to a standard diet, with the cecal gut microbiota composition and function of each group distinct after the 7-week standard diet wash-out. Our results highlight the enduring metabolic consequences of nutritional imbalances, mediated by both the host and gut microbiome, which persist after returning to the original standard diets.
Collapse
Affiliation(s)
- Anastasiia Kostenko
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Hui Zhi
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Kevin Mildau
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Manuela Raffatellu
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Chiba University, UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Allegra T Aron
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
10
|
Abdul-Rahman T, Awuah WA, Mikhailova T, Kalmanovich J, Mehta A, Ng JC, Coghlan MA, Zivcevska M, Tedeschi AJ, de Oliveira EC, Kumar A, Cantu-Herrera E, Lyndin M, Sikora K, Alexiou A, Bilgrami AL, Al-Ghamdi KM, Perveen A, Papadakis M, Ashraf GM. Antioxidant, anti-inflammatory and epigenetic potential of curcumin in Alzheimer's disease. Biofactors 2024; 50:693-708. [PMID: 38226733 DOI: 10.1002/biof.2039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
Alzheimer's disease (AD) constitutes a multifactorial neurodegenerative pathology characterized by cognitive deterioration, personality alterations, and behavioral shifts. The ongoing brain impairment process poses significant challenges for therapeutic interventions due to activating multiple neurotoxic pathways. Current pharmacological interventions have shown limited efficacy and are associated with significant side effects. Approaches focusing on the early interference with disease pathways, before activation of broad neurotoxic processes, could be promising to slow down symptomatic progression of the disease. Curcumin-an integral component of traditional medicine in numerous cultures worldwide-has garnered interest as a promising AD treatment. Current research indicates that curcumin may exhibit therapeutic potential in neurodegenerative pathologies, attributed to its potent anti-inflammatory and antioxidant properties. Additionally, curcumin and its derivatives have demonstrated an ability to modulate cellular pathways via epigenetic mechanisms. This article aims to raise awareness of the neuroprotective properties of curcuminoids that could provide therapeutic benefits in AD. The paper provides a comprehensive overview of the neuroprotective efficacy of curcumin against signaling pathways that could be involved in AD and summarizes recent evidence of the biological efficiency of curcumins in vivo.
Collapse
Affiliation(s)
- Toufik Abdul-Rahman
- Sumy State University, Sumy, Ukraine
- Toufik's World Medical Association, Ukraine
| | - Wireko Andrew Awuah
- Sumy State University, Sumy, Ukraine
- Toufik's World Medical Association, Ukraine
| | | | - Jacob Kalmanovich
- Drexel University College of Medicine, Philadelphia, Pennsylvania, United States
| | - Aashna Mehta
- University of Debrecen-Faculty of Medicine, Debrecen, Hungary
| | - Jyi Cheng Ng
- Faculty of Medicine and Health Sciences, University of Putra Malaysia, Serdang, Malaysia
| | - Megan Ariel Coghlan
- University of Louisville School of Medicine, Louisville, Kentucky, United States
| | - Marija Zivcevska
- Liberty University College of Osteopathic Medicine, Lynchburg, Virginia, United States
| | | | | | - Akinchita Kumar
- Lincoln Memorial University-DeBusk College of Osteopathic Medicine Harrogate, Harrogate, Tennessee, United States
| | - Emiliano Cantu-Herrera
- Department of Clinical Sciences, Division of Health Sciences, University of Monterrey, San Pedro Garza García, Nuevo León, Mexico
| | - Mykola Lyndin
- Sumy State University, Sumy, Ukraine
- Medical Faculty, Institute of Anatomy, University of Duisburg-Essen, Essen, Germany
| | | | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
- AFNP Med, Wien, Austria
| | - Anwar L Bilgrami
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, Uttar Pradesh, India
- Princess Dr. Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Wuppertal, Germany
| | - Ghulam Md Ashraf
- University of Sharjah, College of Health Sciences, and Research Institute for Medical and Health Sciences, Department of Medical Laboratory Sciences, Sharjah, United Arab Emirates
| |
Collapse
|
11
|
Thorwald M, Godoy-Lugo JA, Garcia G, Silva J, Kim M, Christensen A, Mack WJ, Head E, O'Day PA, Benayoun BA, Morgan TE, Pike CJ, Higuchi-Sanabria R, Forman HJ, Finch CE. Iron associated lipid peroxidation in Alzheimers disease is increased in lipid rafts with decreased ferroptosis suppressors, tested by chelation in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.28.534324. [PMID: 37034750 PMCID: PMC10081222 DOI: 10.1101/2023.03.28.534324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Iron-mediated cell death (ferroptosis) is a proposed mechanism of Alzheimers disease (AD) pathology. While iron is essential for basic biological functions, its reactivity generates oxidants which contribute to cell damage and death. To further resolve mechanisms of iron-mediated toxicity in AD, we analyzed postmortem human brain and ApoEFAD mice. AD brains had decreased antioxidant enzymes, including those mediated by glutathione (GSH). Subcellular analyses of AD brains showed greater oxidative damage and lower antioxidant enzymes in lipid rafts, the site of amyloid processing, than in the non-raft membrane fraction. ApoE4 carriers had lower lipid raft yield with greater membrane oxidation. The hypothesized role of iron to AD pathology was tested in ApoEFAD mice by iron chelation with deferoxamine, which decreased fibrillar amyloid and lipid peroxidation, together with increased GSH-mediated antioxidants. These novel molecular pathways in iron mediated damage during AD.
Collapse
|
12
|
Alster P, Otto‐Ślusarczyk D, Wiercińska‐Drapało A, Struga M, Madetko‐Alster N. The potential significance of hepcidin evaluation in progressive supranuclear palsy. Brain Behav 2024; 14:e3552. [PMID: 38953731 PMCID: PMC11191750 DOI: 10.1002/brb3.3552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 07/04/2024] Open
Abstract
INTRODUCTION Hepcidin is a peptide associated with controlling the distribution of iron in tissues. Growing interest is linked with its impact on neurodegenerative diseases, as disruption of the iron regulation may be considered an initiatory element of pathological protein accumulation. The possible impact of hepcidin was not previously sufficiently explored in progressive supranuclear palsy (PSP). METHODS Twelve patients with PSP-Richardson's syndrome (PSP-RS), 12 with PSP-Parkinsonism Predominant (PSP-P), and 12 controls were examined using Unified Parkinson's Disease Rating Scale-III part (UPDRS-III) in OFF stage and analyzed in the context of hepcidin levels in the serum. RESULTS The work revealed increased levels of hepcidin in PSP-RS when compared to PSP-P and controls. Moreover, hepcidin was found to be negatively correlated with UPDRS-III results in PSP-RS, whereas positively in PSP-P. CONCLUSION The work may suggest a possible impact of hepcidin in PSP, possibly differing depending on its subtype.
Collapse
Affiliation(s)
- Piotr Alster
- Department of NeurologyMedical University of WarsawWarsawPoland
| | | | - Alicja Wiercińska‐Drapało
- Department of Infectious and Tropical Diseases and HepatologyMedical University of WarsawWarsawPoland
| | - Marta Struga
- Department of BiochemistryMedical University of WarsawWarsawPoland
| | | |
Collapse
|
13
|
Wilcockson TDW, Roy S. Could Alcohol-Related Cognitive Decline Be the Result of Iron-Induced Neuroinflammation? Brain Sci 2024; 14:520. [PMID: 38928521 PMCID: PMC11201715 DOI: 10.3390/brainsci14060520] [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: 03/18/2024] [Revised: 04/20/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024] Open
Abstract
Excessive and prolonged alcohol use can have long-term severe neurological consequences. The mechanisms involved may be complicated; however, new evidence seems to indicate the involvement of iron accumulation and neuroinflammation. Prolonged alcohol consumption has been linked to the accumulation of iron in specific regions of the brain. Evidence suggests that excess iron in the brain can trigger microglia activation in response. This activation leads to the release of pro-inflammatory cytokines and reactive oxygen species, which can cause damage to neurons and surrounding brain tissue. Additionally, iron-induced oxidative stress and inflammation can disrupt the blood-brain barrier, allowing immune cells from the periphery to infiltrate the brain. This infiltration can lead to further neuroinflammatory responses. Inflammation in the brain subsequently disrupts neuronal networks, impairs synaptic plasticity, and accelerates neuronal cell death. Consequently, cognitive functions such as memory, attention, and decision-making are compromised. Additionally, chronic neuroinflammation can hasten the development and progression of neurodegenerative diseases, further exacerbating cognitive impairment. Therefore, alcohol could act as a trigger for iron-induced neuroinflammation and cognitive decline. Overall, the mechanisms at play here seem to strongly link alcohol with cognitive decline, with neuroinflammation resulting from alcohol-induced iron accumulation playing a pivotal role.
Collapse
Affiliation(s)
- Thomas D. W. Wilcockson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK
| | - Sankanika Roy
- Department of Neurology, Leicester Royal Infirmary, Leicester LE1 5WW, UK;
| |
Collapse
|
14
|
Wang H, Xu L, Tang X, Jiang Z, Feng X. Lipid peroxidation-induced ferroptosis as a therapeutic target for mitigating neuronal injury and inflammation in sepsis-associated encephalopathy: insights into the hippocampal PEBP-1/15-LOX/GPX4 pathway. Lipids Health Dis 2024; 23:128. [PMID: 38685023 PMCID: PMC11057122 DOI: 10.1186/s12944-024-02116-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/21/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) refers to the widespread impairment of brain function caused by noncentral nervous system infection mediated by sepsis. Lipid peroxidation-induced ferroptosis contributes to the occurrence and course of SAE. This study aimed to investigate the relationship between neuronal injury and lipid peroxidation-induced ferroptosis in SAE. METHODS Baseline data were collected from pediatric patients upon admission, and the expression levels of various markers related to lipid peroxidation and ferroptosis were monitored in the serum and peripheral blood mononuclear cells (PBMCs) of patients with SAE as well as SAE model mice. The hippocampal phosphatidylethanolamine-binding protein (PEBP)-1/15-lysine oxidase (LOX)/ glutathione peroxidase 4 (GPX4) pathway was assessed for its role on the inhibitory effect of ferroptosis in SAE treatment. RESULTS The results showed elevated levels of S100 calcium-binding protein beta (S-100β), glial fibrillary acidic protein, and malondialdehyde in the serum of SAE patients, while superoxide dismutase levels were reduced. Furthermore, analysis of PBMCs revealed increased transcription levels of PEBP1, LOX, and long-chain fatty acyl-CoA synthetase family member 4 (ACSL4) in SAE patients, while the transcription levels of GPX4 and cystine/glutamate transporter xCT (SLC7A11) were decreased. In comparison to the control group, the SAE mice exhibited increased expression of S-100β and neuron-specific enolase (NSE) in the hippocampus, whereas the expression of S-100β and NSE were reduced in deferoxamine (DFO) mice. Additionally, iron accumulation was observed in the hippocampus of SAE mice, while the iron ion levels were reduced in the DFO mice. Inhibition of ferroptosis alleviated the mitochondrial damage (as assessed by transmission electron microscopy, hippocampal mitochondrial ATP detection, and the JC-1 polymer-to-monomer ratio in the hippocampus) and the oxidative stress response induced by SAE as well as attenuated neuroinflammatory reactions. Further investigations revealed that the mechanism underlying the inhibitory effect of ferroptosis in SAE treatment is associated with the hippocampal PEBP-1/15-LOX/GPX4 pathway. CONCLUSION These results offer potential therapeutic targets for the management of neuronal injury in SAE and valuable insights into the potential mechanisms of ferroptosis in neurological disorders.
Collapse
Affiliation(s)
- Haosen Wang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou, 221002, Jiangsu, China
| | - Lixiao Xu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
| | - Xiaojuan Tang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
| | - Zhen Jiang
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou, 221002, Jiangsu, China
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China.
| |
Collapse
|
15
|
Everett J, Brooks J, Tjendana Tjhin V, Lermyte F, Hands-Portman I, Plascencia-Villa G, Perry G, Sadler PJ, O’Connor PB, Collingwood JF, Telling ND. Label-Free In Situ Chemical Characterization of Amyloid Plaques in Human Brain Tissues. ACS Chem Neurosci 2024; 15:1469-1483. [PMID: 38501754 PMCID: PMC10995949 DOI: 10.1021/acschemneuro.3c00756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/20/2024] Open
Abstract
The accumulation of amyloid plaques and increased brain redox burdens are neuropathological hallmarks of Alzheimer's disease. Altered metabolism of essential biometals is another feature of Alzheimer's, with amyloid plaques representing sites of disturbed metal homeostasis. Despite these observations, metal-targeting disease treatments have not been therapeutically effective to date. A better understanding of amyloid plaque composition and the role of the metals associated with them is critical. To establish this knowledge, the ability to resolve chemical variations at nanometer length scales relevant to biology is essential. Here, we present a methodology for the label-free, nanoscale chemical characterization of amyloid plaques within human Alzheimer's disease tissue using synchrotron X-ray spectromicroscopy. Our approach exploits a C-H carbon absorption feature, consistent with the presence of lipids, to visualize amyloid plaques selectively against the tissue background, allowing chemical analysis to be performed without the addition of amyloid dyes that alter the native sample chemistry. Using this approach, we show that amyloid plaques contain elevated levels of calcium, carbonates, and iron compared to the surrounding brain tissue. Chemical analysis of iron within plaques revealed the presence of chemically reduced, low-oxidation-state phases, including ferromagnetic metallic iron. The zero-oxidation state of ferromagnetic iron determines its high chemical reactivity and so may contribute to the redox burden in the Alzheimer's brain and thus drive neurodegeneration. Ferromagnetic metallic iron has no established physiological function in the brain and may represent a target for therapies designed to lower redox burdens in Alzheimer's disease. Additionally, ferromagnetic metallic iron has magnetic properties that are distinct from the iron oxide forms predominant in tissue, which might be exploitable for the in vivo detection of amyloid pathologies using magnetically sensitive imaging. We anticipate that this label-free X-ray imaging approach will provide further insights into the chemical composition of amyloid plaques, facilitating better understanding of how plaques influence the course of Alzheimer's disease.
Collapse
Affiliation(s)
- James Everett
- School
of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Thornburrow Drive,Stoke-on-Trent,Staffordshire ST4 7QB, U.K.
- School
of Engineering, University of Warwick, Library Road,Coventry CV4 7AL, U.K.
| | - Jake Brooks
- School
of Engineering, University of Warwick, Library Road,Coventry CV4 7AL, U.K.
| | - Vindy Tjendana Tjhin
- School
of Engineering, University of Warwick, Library Road,Coventry CV4 7AL, U.K.
| | - Frederik Lermyte
- School
of Engineering, University of Warwick, Library Road,Coventry CV4 7AL, U.K.
- Department
of Chemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, 64287 Darmstadt, Germany
| | - Ian Hands-Portman
- School
of Life Sciences, University of Warwick, Gibbet Hill Campus,Coventry CV4 7AL, U.K.
| | - Germán Plascencia-Villa
- Department
of Developmental and Regenerative Biology, The University of Texas at San Antonio (UTSA), San Antonio, Texas 78249, United States
| | - George Perry
- Department
of Developmental and Regenerative Biology, The University of Texas at San Antonio (UTSA), San Antonio, Texas 78249, United States
| | - Peter J. Sadler
- Department
of Chemistry, University of Warwick, Library Road,Coventry CV4 7AL, U.K.
| | - Peter B. O’Connor
- Department
of Chemistry, University of Warwick, Library Road,Coventry CV4 7AL, U.K.
| | | | - Neil D. Telling
- School
of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Thornburrow Drive,Stoke-on-Trent,Staffordshire ST4 7QB, U.K.
| |
Collapse
|
16
|
Liu S, Wang A, Zhou D, Zhai X, Ding L, Tian L, Zhang Y, Wang J, Xin L. PM 2.5 induce neurotoxicity via iron overload and redox imbalance mediated-ferroptosis in HT22 cells. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART A, TOXIC/HAZARDOUS SUBSTANCES & ENVIRONMENTAL ENGINEERING 2024; 59:55-63. [PMID: 38532551 DOI: 10.1080/10934529.2024.2331938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/05/2024] [Indexed: 03/28/2024]
Abstract
PM2.5 is an important risk factor for the development and progression of cognitive impairment-related diseases. Ferroptosis, a new form of cell death driven by iron overload and lipid peroxidation, is proposed to have significant implications. To verify the possible role of ferroptosis in PM2.5-induced neurotoxicity, we investigated the cytotoxicity, intracellular iron content, iron metabolism-related genes, oxidative stress indices and indicators involving in Nrf2 and ferroptosis signaling pathways. Neurotoxicity biomarkers as well as the ferroptotic cell morphological changes were determined by Western Blot and TEM analysis. Our results revealed that PM2.5 induced cytotoxicity, lipid peroxidation, as indicated by MDA content, and neurotoxicity via Aβ deposition in a dose-related manner. Decreased cell viability and excessive iron accumulation in HT-22 cells can be partially blocked by ferroptosis inhibitors. Interestingly, GPX activity, Nrf2, and its regulated ferroptotic-related proteins (i.e. GPX4 and HO-1) were significantly up-regulated by PM2.5. Moreover, gene expression of DMT1, TfR1, IRP2 and FPN1 involved in iron homeostasis and NCOA4-dependent ferritinophagy were activated after PM2.5 exposure. The results demonstrated that PM2.5 triggered ferritinophagy-dependent ferroptotic cell death due to iron overload and redox imbalance. Activation of Nrf2 signaling pathways may confer a protective mechanism for PM2.5-induced oxidative stress and ferroptosis.
Collapse
Affiliation(s)
- Shuhui Liu
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Aiqing Wang
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Danhong Zhou
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Xuedi Zhai
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Occupational Disease Prevention and Control, Yancheng Center for Disease Prevention and Control, Yancheng, China
| | - Ling Ding
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Liang Tian
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yidan Zhang
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Jianshu Wang
- Department of Environmental Hygiene, Suzhou Center for Disease Prevention and Control, Suzhou, China
| | - Lili Xin
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Imtiaz A, Shimonaka S, Uddin MN, Elahi M, Ishiguro K, Hasegawa M, Hattori N, Motoi Y. Selection of lansoprazole from an FDA-approved drug library to inhibit the Alzheimer's disease seed-dependent formation of tau aggregates. Front Aging Neurosci 2024; 16:1368291. [PMID: 38633982 PMCID: PMC11022852 DOI: 10.3389/fnagi.2024.1368291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
The efficacy of current treatments is still insufficient for Alzheimer's disease (AD), the most common cause of Dementia. Out of the two pathological hallmarks of AD amyloid-β plaques and neurofibrillary tangles, comprising of tau protein, tau pathology strongly correlates with the symptoms of AD. Previously, screening for inhibitors of tau aggregation that target recombinant tau aggregates have been attempted. Since a recent cryo-EM analysis revealed distinct differences in the folding patterns of heparin-induced recombinant tau filaments and AD tau filaments, this study focused on AD seed-dependent tau aggregation in drug repositioning for AD. We screened 763 compounds from an FDA-approved drug library using an AD seed-induced tau aggregation in SH-SY5Y cell-based assay. In the first screening, 180 compounds were selected, 72 of which were excluded based on the results of lactate dehydrogenase assay. In the third screening with evaluations of soluble and insoluble tau, 38 compounds were selected. In the fourth screening with 3 different AD seeds, 4 compounds, lansoprazole, calcipotriene, desogestrel, and pentamidine isethionate, were selected. After AD seed-induced real-time quaking-induced conversion, lansoprazole was selected as the most suitable drug for repositioning. The intranasal administration of lansoprazole for 4 months to AD seed-injected mice improved locomotor activity and reduced both the amount of insoluble tau and the extent of phosphorylated tau-positive areas. Alanine replacement of the predicted binding site to an AD filament indicated the involvement of Q351, H362, and K369 in lansoprazole and C-shaped tau filaments. These results suggest the potential of lansoprazole as a candidate for drug repositioning to an inhibitor of tau aggregate formation in AD.
Collapse
Affiliation(s)
- Ahmed Imtiaz
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Neurology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shotaro Shimonaka
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Mohammad Nasir Uddin
- Department of Biochemistry & Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science & Technology University, Tangail, Bangladesh
| | - Montasir Elahi
- Center for Birth Defect Research, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Koichi Ishiguro
- Department of Neurology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Masato Hasegawa
- Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Yumiko Motoi
- Medical Center for Dementia, Juntendo University Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
18
|
Wang N, Que H, Luo Q, Zheng W, Li H, Wang Q, Gu J. Mechanisms of ferroptosis in nonalcoholic fatty liver disease and therapeutic effects of traditional Chinese medicine: a review. Front Med (Lausanne) 2024; 11:1356225. [PMID: 38590315 PMCID: PMC10999571 DOI: 10.3389/fmed.2024.1356225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/22/2024] [Indexed: 04/10/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive accumulation of fat in hepatocytes (nonalcoholic fatty liver (NAFL)), and lobular inflammation and hepatocyte damage (which characterize nonalcoholic steatohepatitis (NASH) are found in most patients). A subset of patients will gradually develop liver fibrosis, cirrhosis, and eventually hepatocellular carcinoma, which is a deadly disease that threatens human life worldwide. Ferroptosis, a novel nonapoptotic form of programmed cell death (PCD) characterized by iron-dependent accumulation of reactive oxygen radicals and lipid peroxides, is closely related to NAFLD. Traditional Chinese medicine (TCM) has unique advantages in the prevention and treatment of NAFLD due to its multicomponent, multipathway and multitarget characteristics. In this review, we discuss the effect of TCM on NAFLD by regulating ferroptosis, in order to provide reference for the further development and application of therapeutic drugs to treat NAFLD.
Collapse
Affiliation(s)
- Nan Wang
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Hanyun Que
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Qiulin Luo
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Wenxin Zheng
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Hong Li
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Qin Wang
- College of Pharmacy, Southwest Minzu University, Chengdu, China
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| |
Collapse
|
19
|
Miskovic R, Ljubicic J, Bonaci-Nikolic B, Petkovic A, Markovic V, Rankovic I, Djordjevic J, Stankovic A, Klaassen K, Pavlovic S, Stojanovic M. Case report: Rapidly progressive neurocognitive disorder with a fatal outcome in a patient with PU.1 mutated agammaglobulinemia. Front Immunol 2024; 15:1324679. [PMID: 38500873 PMCID: PMC10945545 DOI: 10.3389/fimmu.2024.1324679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Introduction PU.1-mutated agammaglobulinemia (PU.MA) represents a recently described autosomal-dominant form of agammaglobulinemia caused by mutation of the SPI1 gene. This gene codes for PU.1 pioneer transcription factor important for the maturation of monocytes, B lymphocytes, and conventional dendritic cells. Only six cases with PU.MA, presenting with chronic sinopulmonary and systemic enteroviral infections, have been previously described. Accumulating literature evidence suggests a possible relationship between SPI1 mutation, microglial phagocytic dysfunction, and the development of Alzheimer's disease (AD). Case description We present a Caucasian female patient born from a non-consanguineous marriage, who was diagnosed with agammaglobulinemia at the age of 15 years when the immunoglobulin replacement therapy was started. During the following seventeen years, she was treated for recurrent respiratory and intestinal infections. At the age of 33 years, the diagnosis of celiac-like disease was established. Five years later progressive cognitive deterioration, unstable gait, speech disturbances, and behavioral changes developed. Comprehensive microbiological investigations were negative, excluding possible infective etiology. Brain MRI, 18FDG-PET-CT, and neuropsychological testing were suggestive for a diagnosis of a frontal variant of AD. Clinical exome sequencing revealed the presence of a novel frameshift heterozygous variant c.441dup in exon 4 of the SPI1 gene. Despite intensive therapy, the patient passed away a few months after the onset of the first neurological symptoms. Conclusion We describe the first case of PU.MA patient presenting with a rapidly progressive neurocognitive deterioration. The possible role of microglial dysfunction in patients with SPI1 mutation could explain their susceptibility to neurodegenerative diseases thus highlighting the importance of genetic testing in patients with inborn errors of immunity. Since PU.MA represents a newly described form of agammaglobulinemia, our case expands the spectrum of manifestations associated with SPI1 mutation.
Collapse
Affiliation(s)
- Rada Miskovic
- Clinic of Allergy and Immunology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Ljubicic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Branka Bonaci-Nikolic
- Clinic of Allergy and Immunology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ana Petkovic
- Diagnostic Department, Center of Sterotaxic Radiosurgery, Clinic of Neurosurgery, University Clinical Center of Serbia, Belgrade, Serbia
| | - Vladana Markovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Neurology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Ivan Rankovic
- Department of Gastroenterology and Liver Unit, Royal Cornwall Hospitals NHS Trust, University of Exeter, Truro, United Kingdom
| | - Jelena Djordjevic
- Clinic of Neurology and Psychiatry for Children and Youth, Belgrade, Serbia
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Ana Stankovic
- Center for Radiology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Kristel Klaassen
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Sonja Pavlovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Maja Stojanovic
- Clinic of Allergy and Immunology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
20
|
Alrouji M, Alhumaydhi FA, Venkatesan K, Sharaf SE, Shahwan M, Shamsi A. Evaluation of binding mechanism of dietary phytochemical, capsaicin, with human transferrin: targeting neurodegenerative diseases therapeutics. Front Pharmacol 2024; 15:1348128. [PMID: 38495092 PMCID: PMC10943693 DOI: 10.3389/fphar.2024.1348128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/16/2024] [Indexed: 03/19/2024] Open
Abstract
Human transferrin (htf) plays a crucial role in regulating the balance of iron within brain cells; any disruption directly contributes to the development of Neurodegenerative Diseases (NDs) and other related pathologies, especially Alzheimer's Disease (AD). In recent times, a transition towards natural compounds is evident to treat diseases and this shift is mainly attributed to their broad therapeutic potential along with minimal side effects. Capsaicin, a natural compound abundantly found in red and chili peppers, possess neuroprotective potential. The current work targets to decipher the interaction mechanism of capsaicin with htf using experimental and computational approaches. Molecular docking analysis revealed that capsaicin occupies the iron binding pocket of htf, with good binding affinity. Further, the binding mechanism was investigated atomistically using Molecular dynamic (MD) simulation approach. The results revealed no significant alterations in the structure of htf implying the stability of the complex. In silico observations were validated by fluorescence binding assay. Capsaicin binds to htf with a binding constant (K) of 3.99 × 106 M-1, implying the stability of the htf-capsaicin complex. This study lays a platform for potential applications of capsaicin in treatment of NDs in terms of iron homeostasis.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Kumar Venkatesan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sharaf E. Sharaf
- Pharmaceutical Sciences Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Anas Shamsi
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
21
|
Xiang Y, Song X, Long D. Ferroptosis regulation through Nrf2 and implications for neurodegenerative diseases. Arch Toxicol 2024; 98:579-615. [PMID: 38265475 PMCID: PMC10861688 DOI: 10.1007/s00204-023-03660-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024]
Abstract
This article provides an overview of the background knowledge of ferroptosis in the nervous system, as well as the key role of nuclear factor E2-related factor 2 (Nrf2) in regulating ferroptosis. The article takes Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) as the starting point to explore the close association between Nrf2 and ferroptosis, which is of clear and significant importance for understanding the mechanism of neurodegenerative diseases (NDs) based on oxidative stress (OS). Accumulating evidence links ferroptosis to the pathogenesis of NDs. As the disease progresses, damage to the antioxidant system, excessive OS, and altered Nrf2 expression levels, especially the inhibition of ferroptosis by lipid peroxidation inhibitors and adaptive enhancement of Nrf2 signaling, demonstrate the potential clinical significance of Nrf2 in detecting and identifying ferroptosis, as well as targeted therapy for neuronal loss and mitochondrial dysfunction. These findings provide new insights and possibilities for the treatment and prevention of NDs.
Collapse
Affiliation(s)
- Yao Xiang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
| | - Xiaohua Song
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China
| | - Dingxin Long
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China.
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang Medical School, University of South China, Hengyang, 421001, People's Republic of China.
| |
Collapse
|
22
|
Zeng W, Cai J, Zhang L, Peng Q. Iron Deposition in Parkinson's Disease: A Mini-Review. Cell Mol Neurobiol 2024; 44:26. [PMID: 38393383 PMCID: PMC10891198 DOI: 10.1007/s10571-024-01459-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
Iron deposition is crucial pathological changes observed in patients with Parkinson's disease (PD). Recently, scientists have actively explored therapeutic approaches targeting iron deposition in PD. However, several clinical studies have failed to yield consistent results. In this review, we provide an overview of iron deposition in PD, from both basic research and clinical perspectives. PD patients exhibit abnormalities in various iron metabolism-related proteins, leading to disruptions in iron distribution, transport, storage, and circulation, ultimately resulting in iron deposition. Excess iron can induce oxidative stress and iron-related cell death, and exacerbate mitochondrial dysfunction, contributing to the progression of PD pathology. Magnetic resonance imaging studies have indicated that the characteristics of iron deposition in the brains of PD patients vary. Iron deposition correlates with the clinical symptoms of PD, and patients with different disease courses and clinical presentations display distinct patterns of iron deposition. These iron deposition patterns may contribute to PD diagnosis. Iron deposition is a promising target for PD treatment. However, further research is required to elucidate the underlying mechanisms and their impacts on PD.
Collapse
Affiliation(s)
- Weiqi Zeng
- Department of Neurology, The First People's Hospital of Foshan, Foshan, China
| | - Jin Cai
- Department of Cardiology, The Second Hospital of Zhangzhou, Zhangzhou, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qiwei Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Kang T, Han Z, Zhu L, Cao B. TFR1 knockdown alleviates iron overload and mitochondrial dysfunction during neural differentiation of Alzheimer's disease-derived induced pluripotent stem cells by interacting with GSK3B. Eur J Med Res 2024; 29:101. [PMID: 38321571 PMCID: PMC10845644 DOI: 10.1186/s40001-024-01677-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Iron metabolism disorders are implicated in the pathogenesis of Alzheimer's disease (AD). It was previously reported that transferrin receptor (TFR1) expression was upregulated in AD mouse model. However, the precise biological functions of TFR1 in AD progression remains unclear. Herein, we observed a gradual increase in TFR1 protein expression during the differentiation of AD patient-derived induced pluripotent stem cells (AD-iPS). TFR1 knockdown inhibited the protein expression of ferritin and ferritin heavy chain 1 (FTH1), enhanced the expression of ferroportin 1 (FPN1), and decreased intracellular levels of total iron, labile iron, and reactive oxygen species (ROS). Moreover, TFR1 knockdown improved mitochondrial membrane potential (MMP), increased adenosine triphosphate (ATP) content, downregulated mitochondrial fission proteins, and upregulated mitochondrial fusion proteins. TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS, while TFR1 overexpression showed the opposite results. Additionally, TFR1interacted with glycogen synthase kinase 3 beta (GSK3B) and promoted GSK3B expression. GSK3B overexpression reversed the inhibitory effects of TFR1 knockdown on iron overload and mitochondrial dysfunction in AD-iPS differentiated neural cells. In conclusion, TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS by promoting GSK3B expression. Our findings provide a potential therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Tao Kang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Zheng Han
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Lijuan Zhu
- Department of Anesthesia, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Bingqing Cao
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| |
Collapse
|
24
|
Yeerlan J, He B, Hu X, Zhang L. Global Research Trends and Hotspots for Ferroptosis, Necroptosis, and Pyroptosis in Alzheimer's Disease from the Past to 2023: A Combined Bibliometric Review. J Alzheimers Dis Rep 2024; 8:129-142. [PMID: 38312529 PMCID: PMC10836606 DOI: 10.3233/adr-230092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024] Open
Abstract
Background Alzheimer's disease (AD) is a genetically intricate neurodegenerative disorder. Studies on "Ferroptosis in AD", "Pyroptosis in AD", and "Necroptosis in AD" are becoming more prevalent and there is increasing evidence that they are closely related to AD. However, there has not yet been a thorough bibliometrics-based investigation on this subject. Objective This study uses a bibliometric approach to visualize and analyze the literature within the field of three distinct types of cell death in AD and explores the current research hotspots and prospective research directions. Methods We collected relevant articles from the Web of Science and used CiteSpace, VOS viewer, and Pajek to perform a visual analysis. Results A total of 123, 95, and 84 articles were published in "Ferroptosis in AD", "Pyroptosis in AD", and "Necroptosis in AD", respectively. Based on keywords analysis, we can observe that "oxidative stress" and "lipid peroxidation", "cell death" and "activation", and "Nlrp3 inflammasome" and "activation" were the three most prominent words in the field of "Ferroptosis in AD", "Pyroptosis in AD", and "Necroptosis in AD", respectively. Focusing on the breakout words in the keyword analysis, we reviewed the mechanisms of ferroptosis, pyroptosis, and necroptosis in AD. By mapping the time zones of the keywords, we speculated on the evolutionary trends of ferroptosis, pyrotosis, and necroptosis in AD. Conclusions Our findings can help researchers grasp the research status of three types of cell death in AD and determine new directions for future research as soon as possible.
Collapse
Affiliation(s)
| | - Binhong He
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Xianliang Hu
- Chengdu Eighth People’s Hospital, Geriatric Hospital of Chengdu Medical College, Chengdu, China
| | - LuShun Zhang
- Department of Neurobiology, Department of Pathology and Pathophysiology, Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China
| |
Collapse
|
25
|
Nelson D, Thompson KJ, Wang L, Wang Z, Eberts P, Azarin SM, Kalari KR, Kandimalla KK. Pericyte Control of Gene Expression in the Blood-Brain Barrier Endothelium: Implications for Alzheimer's Disease. J Alzheimers Dis 2024; 99:S281-S297. [PMID: 38393902 DOI: 10.3233/jad-230907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Background A strong body of evidence suggests that cerebrovascular pathologies augment the onset and progression of Alzheimer's disease (AD). One distinctive aspect of this cerebrovascular dysfunction is the degeneration of brain pericytes-often overlooked supporting cells of blood-brain barrier endothelium. Objective The current study investigates the influence of pericytes on gene and protein expressions in the blood-brain barrier endothelium, which is expected to facilitate the identification of pathophysiological pathways that are triggered by pericyte loss and lead to blood-brain barrier dysfunction in AD. Methods Bioinformatics analysis was conducted on the RNA-Seq expression counts matrix (GSE144474), which compared solo-cultured human blood-brain barrier endothelial cells against endothelial cells co-cultured with human brain pericytes in a non-contact model. We constructed a similar cell culture model to verify protein expression using western blots. Results The insulin resistance and ferroptosis pathways were found to be enriched. Western blots of the insulin receptor and heme oxygenase expressions were consistent with those observed in RNA-Seq data. Additionally, we observed more than 5-fold upregulation of several genes associated with neuroprotection, including insulin-like growth factor 2 and brain-derived neurotrophic factor. Conclusions Results suggest that pericyte influence on blood-brain barrier endothelial gene expression confers protection from insulin resistance, iron accumulation, oxidative stress, and amyloid deposition. Since these are conditions associated with AD pathophysiology, they imply mechanisms by which pericyte degeneration could contribute to disease progression.
Collapse
Affiliation(s)
- Doug Nelson
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Kevin J Thompson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Lushan Wang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Zengtao Wang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Paulina Eberts
- Department of Chemical Engineering and Materials Science, College of Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, College of Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Krishna R Kalari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
26
|
Sande R, Godad A, Doshi G. Zebrafish Experimental Animal Models for AD: A Comprehensive Review. Curr Rev Clin Exp Pharmacol 2024; 19:295-311. [PMID: 38284707 DOI: 10.2174/0127724328279684240104094257] [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: 08/29/2023] [Revised: 11/26/2023] [Accepted: 12/06/2023] [Indexed: 01/30/2024]
Abstract
AD disease (AD) is a multifaceted and intricate neurodegenerative disorder characterized by intracellular neurofibrillary tangle (NFT) formation and the excessive production and deposition of Aβ senile plaques. While transgenic AD models have been found instrumental in unravelling AD pathogenesis, they involve cost and time constraints during the preclinical phase. Zebrafish, owing to their simplicity, well-defined behavioural patterns, and relevance to neurodegenerative research, have emerged as a promising complementary model. Zebrafish possess glutaminergic and cholinergic pathways implicated in learning and memory, actively contributing to our understanding of neural transmission processes. This review sheds light on the molecular mechanisms by which various neurotoxic agents, including okadaic acid (OKA), cigarette smoke extract, metals, and transgenic zebrafish models with genetic similarities to AD patients, induce cognitive impairments and neuronal degeneration in mammalian systems. These insights may facilitate the identification of effective neurotoxic agents for replicating AD pathogenesis in the zebrafish brain. In this comprehensive review, the pivotal role of zebrafish models in advancing our comprehension of AD is emphasized. These models hold immense potential for shaping future research directions and clinical interventions, ultimately contributing to the development of novel AD therapies.
Collapse
Affiliation(s)
- Ruksar Sande
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Angel Godad
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| |
Collapse
|
27
|
Yadav VK, Choudhary N, Gacem A, Verma RK, Abul Hasan M, Tarique Imam M, Almalki ZS, Yadav KK, Park HK, Ghosh T, Kumar P, Patel A, Kalasariya H, Jeon BH, Ali AlMubarak H. Deeper insight into ferroptosis: association with Alzheimer's, Parkinson's disease, and brain tumors and their possible treatment by nanomaterials induced ferroptosis. Redox Rep 2023; 28:2269331. [PMID: 38010378 PMCID: PMC11001282 DOI: 10.1080/13510002.2023.2269331] [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: 11/29/2023] Open
Abstract
Ferroptosis is an emerging and novel type of iron-dependent programmed cell death which is mainly caused by the excessive deposition of free intracellular iron in the brain cells. This deposited free iron exerts a ferroptosis pathway, resulting in lipid peroxidation (LiPr). There are mainly three ferroptosis pathways viz. iron metabolism-mediated cysteine/glutamate, and LiPr-mediated. Iron is required by the brain as a redox metal for several physiological activities. Due to the iron homeostasis balance disruption, the brain gets adversely affected which further causes neurodegenerative diseases (NDDs) like Parkinson's and Alzheimer's disease, strokes, and brain tumors like glioblastoma (GBS), and glioma. Nanotechnology has played an important role in the prevention and treatment of these NDDs. A synergistic effect of nanomaterials and ferroptosis could prove to be an effective and efficient approach in the field of nanomedicine. In the current review, the authors have highlighted all the latest research in the field of ferroptosis, specifically emphasizing on the role of major molecular key players and various mechanisms involved in the ferroptosis pathway. Moreover, here the authors have also addressed the correlation of ferroptosis with the pathophysiology of NDDs and theragnostic effect of ferroptosis and nanomaterials for the prevention and treatment of NDDs.
Collapse
Affiliation(s)
- Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Nisha Choudhary
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Amel Gacem
- Department of Physics, Faculty of Sciences, University 20 Août 1955, Skikda, Algeria
| | - Rakesh Kumar Verma
- Department of Biosciences, School of Liberal Arts & Sciences, Mody University of Science and Technology, Sikar, India
| | - Mohd Abul Hasan
- Civil Engineering Department, College of Engineering, King Khalid University, Abha, Kingdom of Saudi Arabia (KSA)
| | - Mohammad Tarique Imam
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Ziyad Saeed Almalki
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Krishna Kumar Yadav
- Faculty of Science and Technology, Madhyanchal Professional University, Bhopal, India
- Environmental and Atmospheric Sciences Research Group, Scientific Research Center, Al-Ayen University, Nasiriyah, Iraq
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Tathagata Ghosh
- Department of Arts, School of Liberal Arts & Sciences, Mody University of Science and Technology, Sikar, India
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, India
| | - Haresh Kalasariya
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hassan Ali AlMubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University (KKU), Abha, Kingdom of Saudi Arabia
| |
Collapse
|
28
|
SHI J, XIE Y, LI Y, REN D, ZHANG Y, SHAO H, LIU Y, WANG X, LI Y. Effects of food-grade iron(III) oxide nanoparticles on cecal digesta- and mucosa-associated microbiota and short-chain fatty acids in rats. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 43:43-54. [PMID: 38188661 PMCID: PMC10767317 DOI: 10.12938/bmfh.2023-012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/21/2023] [Indexed: 01/09/2024]
Abstract
Although iron(III) oxide nanoparticles (IONPs) are widely used in diverse applications ranging from food to biomedicine, the effects of IONPs on different locations of gut microbiota and short-chain fatty acids (SCFAs) are unclear. So, a subacute repeated oral toxicity study on Sprague Dawley (SD) rats was performed, administering low (50 mg/kg·bw), medium (100 mg/kg·bw), and high (200 mg/kg·bw) doses of IONPs. In this study, we found that a high dose of IONPs increased animal weight, and 16S rRNA sequencing revealed that IONPs caused intestinal flora disorders in both the cecal digesta- and mucosa-associated microbiota. However, only high-dose IONP exposure changed the abundance and composition of the mucosa-associated microbiota. IONPs increased the relative abundances of Firmicutes, Ruminococcaceae_UCG-014, Ruminiclostridium_9, Romboutsia, and Bilophila and decreased the relative abundance of Bifidobacterium, and many of these microorganisms are associated with weight gain, obesity, inflammation, diabetes, and mucosal damage. Functional analysis showed that changes in the gut microbiota induced by a high dose of IONPs were mainly related to metabolism, infection, immune, and endocrine disease functions. IONPs significantly elevated the levels of valeric, isobutyric, and isovaleric acid, promoting the absorption of iron. This is the first description of intestinal microbiota dysbiosis in SD rats caused by IONPs, and the effects and mechanisms of action of IONPs on intestinal and host health need to be further studied and confirmed.
Collapse
Affiliation(s)
- Jiangchun SHI
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yumeng XIE
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yulin LI
- Department of Hospital-acquired Infection Management, Guizhou
Provincial People’s Hospital, Guiyang 550002, China
| | - Dongxia REN
- Department of Blood Transfusion, Tangdu Hospital, Fourth
Military Medical University, Xi’an 710032, China
| | - Yiqi ZHANG
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Huangfang SHAO
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yang LIU
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Xue WANG
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yun LI
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
- Provincial Key Laboratory of Food Safety Monitoring and Risk
Assessment of Sichuan, Chengdu 610041, China
| |
Collapse
|
29
|
Chen S, Huang W, Tan H, Yin G, Chen S, Zhao K, Huang Y, Zhang Y, Li H, Wu C. A large Stokes shift NIR fluorescent probe for visual monitoring of mitochondrial peroxynitrite during inflammation and ferroptosis and in an Alzheimer's disease model. Analyst 2023; 148:4331-4338. [PMID: 37547973 DOI: 10.1039/d3an00956d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The excessive formation of peroxynitrite (ONOO-) in mitochondria has been implicated in various pathophysiological processes and diseases. However, owing to short emission wavelengths and small Stokes shifts, previously reported fluorescent probes pose significant challenges for mitochondrial ONOO- imaging in biological systems. In this study, a near-infrared (NIR) fluorescent probe, denoted as DCO-POT, is designed for the visual monitoring of mitochondrial ONOO-, displaying a remarkable Stokes shift of 170 nm. The NIR fluorophore of DCO-CHO is released by DCO-POT upon the addition of ONOO-, resulting in off-on NIR fluorescence at 670 nm. This phenomenon facilitates the high-resolution confocal laser scanning imaging of ONOO- generated in biological systems. The practical applications of DCO-POT as an efficient fluorescence imaging tool are verified in this study. DCO-POT enables the fluorometric visualization of ONOO- in organelles, cells, and organisms. In particular, ONOO- generation is analyzed during cellular and organism-level (zebrafish) inflammation during ferroptosis and in an Alzheimer's disease mouse model. The excellent visual monitoring performance of DCO-POT in vivo makes it a promising tool for exploring the pathophysiological effects of ONOO-.
Collapse
Affiliation(s)
- Shiying Chen
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Wei Huang
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Hongli Tan
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Guoxing Yin
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Shengyou Chen
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Kuicheng Zhao
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Yinghui Huang
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Youyu Zhang
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Haitao Li
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Cuiyan Wu
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
- School of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo 315211, PR China
| |
Collapse
|
30
|
Wang H, Liu X, Song L, Yang W, Li M, Chen Q, Lv H, Zhao P, Yang Z, Liu W, Wang ZC. Dysfunctional Coupling of Cerebral Blood Flow and Susceptibility Value in the Bilateral Hippocampus is Associated with Cognitive Decline in Nondialysis Patients with CKD. J Am Soc Nephrol 2023; 34:1574-1588. [PMID: 37476849 PMCID: PMC10482064 DOI: 10.1681/asn.0000000000000185] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/13/2023] [Indexed: 07/22/2023] Open
Abstract
SIGNIFICANCE STATEMENT Patients with end stage CKD often develop cognitive decline, but whether this is related to the underlying disease or to hemodialysis remains unclear. We performed three-dimensional pseudocontinuous arterial spin labeling and quantitative susceptibility mapping prospectively in 40 patients with stage 1-4 CKD, 47 nondialysis patients with stage 5 CKD, and 44 healthy controls. Our magnetic resonance imaging data demonstrate that changes in cerebral blood flow-susceptibility coupling might underlie this cognitive decline, perhaps in the hippocampus and thalamus. These results suggest that magnetic resonance imaging parameters are potential biomarkers of cognitive decline in patients with CKD. Moreover, our findings may lead to discovery of novel therapeutic targets to prevent cognitive decline in patients with CKD. BACKGROUND Cerebral blood flow (CBF) and susceptibility values reflect vascular and iron metabolism, providing mechanistic insights into conditions of health and disease. Nondialysis patients with CKD show a cognitive decline, but the pathophysiological mechanisms underlying this remain unclear. METHODS Three-dimensional pseudocontinuous arterial spin labeling and quantitative susceptibility mapping were prospectively performed in 40 patients with stage 1-4 CKD (CKD 1-4), 47 nondialysis patients with stage 5 CKD (CKD 5ND), and 44 healthy controls (HCs). Voxel-based global and regional analyses of CBF, susceptibility values, and vascular-susceptibility coupling were performed. Furthermore, the association between clinical performance and cerebral perfusion and iron deposition was analyzed. RESULTS For CBF, patients with CKD 5ND had higher normalized CBF in the hippocampus and thalamus than HCs. Patients with CKD 5ND had higher normalized CBF in the hippocampus and thalamus than those with CKD 1-4. The susceptibility values in the hippocampus and thalamus were lower in patients with CKD 5ND than in HCs. Patients with CKD 5ND had higher susceptibility value in the caudate nucleus than those with CKD 1-4. More importantly, patients with CKD 5ND had lower CBF-susceptibility coupling than HCs. In addition, CBF and susceptibility values were significantly associated with clinical performance. CONCLUSIONS Our findings demonstrate a new neuropathological mechanism in patients with CKD, which leads to regional changes in CBF-susceptibility coupling. These changes are related to cognitive decline, providing potential imaging markers for assessing clinical disability and cognitive decline in these patients.
Collapse
Affiliation(s)
- Hao Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lijun Song
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenbo Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingan Li
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qian Chen
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Han Lv
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Pengfei Zhao
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhen-chang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Schreiner OD, Schreiner TG. Iron chelators as a therapeutic option for Alzheimer's disease-A mini-review. FRONTIERS IN AGING 2023; 4:1234958. [PMID: 37602277 PMCID: PMC10433644 DOI: 10.3389/fragi.2023.1234958] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Neurodegenerative disorders, particularly Alzheimer's disease (AD), remain a great challenge regarding the finding of effective treatment, one main reason being the incomplete understanding of their etiology. With many intensely debated hypotheses, a newer approach based on the impact of iron imbalance in sustaining neurodegeneration in the central nervous system becomes increasingly popular. Altered iron homeostasis leads to increased iron accumulation in specific brain areas, explaining the clinical picture of AD patients. Moreover, growing evidence sustains the significant impact of iron metabolism in relationship to other pathological processes encountered in the AD-affected brain, such as the amyloidogenic pathway, chronic inflammation, or oxidative stress. In this context, this mini-review aims to summarize the novel data from the continuously expanding literature on this topic in a didactic manner. Thus, in the first part, the authors briefly highlight the most relevant aspects related to iron absorption, transport, regulation, and elimination at the cerebral level, focusing on the role of the blood-brain barrier and the newer concept of ferroptosis. Subsequently, currently available iron chelation therapies are discussed, including an overview of the most relevant clinical trials on this topic. In the final part, based on the latest results from in vitro and in vivo studies, new research directions are suggested to enhance the development of effective antidementia therapies.
Collapse
Affiliation(s)
- Oliver Daniel Schreiner
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Medical Oncology Department, Regional Institute of Oncology, Iasi, Romania
| | - Thomas Gabriel Schreiner
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, Iasi, Romania
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| |
Collapse
|
32
|
Wang Y, Wu S, Li Q, Sun H, Wang H. Pharmacological Inhibition of Ferroptosis as a Therapeutic Target for Neurodegenerative Diseases and Strokes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300325. [PMID: 37341302 PMCID: PMC10460905 DOI: 10.1002/advs.202300325] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/23/2023] [Indexed: 06/22/2023]
Abstract
Emerging evidence suggests that ferroptosis, a unique regulated cell death modality that is morphologically and mechanistically different from other forms of cell death, plays a vital role in the pathophysiological process of neurodegenerative diseases, and strokes. Accumulating evidence supports ferroptosis as a critical factor of neurodegenerative diseases and strokes, and pharmacological inhibition of ferroptosis as a therapeutic target for these diseases. In this review article, the core mechanisms of ferroptosis are overviewed and the roles of ferroptosis in neurodegenerative diseases and strokes are described. Finally, the emerging findings in treating neurodegenerative diseases and strokes through pharmacological inhibition of ferroptosis are described. This review demonstrates that pharmacological inhibition of ferroptosis by bioactive small-molecule compounds (ferroptosis inhibitors) could be effective for treatments of these diseases, and highlights a potential promising therapeutic avenue that could be used to prevent neurodegenerative diseases and strokes. This review article will shed light on developing novel therapeutic regimens by pharmacological inhibition of ferroptosis to slow down the progression of these diseases in the future.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care MedicineAerospace Center HospitalPeking University Aerospace School of Clinical MedicineBeijing100049P. R. China
| | - Shuang Wu
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhan430000P. R. China
| | - Qiang Li
- Department of NeurologyThe Affiliated Hospital of Chifeng UniversityChifeng024005P. R. China
| | - Huiyan Sun
- Chifeng University Health Science CenterChifeng024000P. R. China
| | - Hongquan Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060P. R. China
| |
Collapse
|
33
|
Chen B, Ouyang X, Cheng C, Chen D, Su J, Hu Y, Li X. Bioactive peptides derived from Radix Angelicae sinensis inhibit ferroptosis in HT22 cells through direct Keap1-Nrf2 PPI inhibition. RSC Adv 2023; 13:22148-22157. [PMID: 37492506 PMCID: PMC10363710 DOI: 10.1039/d3ra04057g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
The development of natural peptides as direct Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid2-related factor 2 (Nrf2) protein-protein interaction (PPI) inhibitors for antioxidant and anti-ferroptotic purposes has attracted increasing interest from chemists. Radix Angelicae sinensis (RAS) is a widely used traditional Chinese medicine with antioxidant capability. However, few studies have screened Keap1-Nrf2 PPI inhibitory RAS peptides (RASPs). This study optimized the extraction and hydrolysis protocols of RAS protein using response surface methodology coupled with Box-Behnken design. The molecular weight distribution of the prepared hydrolysates was analysed to obtain active fractions. Subsequently, ultra-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight tandem mass spectrometry was employed to identify RASPs. Various in vitro and in silico assays were conducted to evaluate the antioxidant and anti-ferroptotic effects of RASPs. The results revealed that at least 50 RASPs could be obtained through the optimized protocols. RASPs containing active residues effectively scavenged 2,2-diphenyl-1-picrylhydrazyl radical and 2,2'-azinobis(3-ethylbenzothiazoline)-6-sulfonic acid radical cation. They also showed cytoprotective effect against erastin-induced ferroptosis in HT22 cells, which was characterized by the activation of Nrf2 and weakened under the incubation of an Nrf2 inhibitor. Moreover, RASPs could bind to Keap1 and then dissociate Nrf2 in molecular dynamics simulations. In conclusion, RASPs exhibit antioxidant activity through hydrogen atom transfer and electron transfer mechanisms. Importantly, they also inhibit ferroptosis by directly inhibiting Keap1-Nrf2 PPI.
Collapse
Affiliation(s)
- Ban Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology Wuhan 430068 China
| | | | - Chunfeng Cheng
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine Shenzhen 518000 China
| | - Dongfeng Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine Guangzhou 510000 China
| | - Jiangtao Su
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology Wuhan 430068 China
| | - Yuchen Hu
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology Wuhan 430068 China
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine Guangzhou 510000 China
| |
Collapse
|
34
|
Cerasuolo M, Di Meo I, Auriemma MC, Trojsi F, Maiorino MI, Cirillo M, Esposito F, Polito R, Colangelo AM, Paolisso G, Papa M, Rizzo MR. Iron and Ferroptosis More than a Suspect: Beyond the Most Common Mechanisms of Neurodegeneration for New Therapeutic Approaches to Cognitive Decline and Dementia. Int J Mol Sci 2023; 24:9637. [PMID: 37298586 PMCID: PMC10253771 DOI: 10.3390/ijms24119637] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Neurodegeneration is a multifactorial process that involves multiple mechanisms. Examples of neurodegenerative diseases are Parkinson's disease, multiple sclerosis, Alzheimer's disease, prion diseases such as Creutzfeldt-Jakob's disease, and amyotrophic lateral sclerosis. These are progressive and irreversible pathologies, characterized by neuron vulnerability, loss of structure or function of neurons, and even neuron demise in the brain, leading to clinical, functional, and cognitive dysfunction and movement disorders. However, iron overload can cause neurodegeneration. Dysregulation of iron metabolism associated with cellular damage and oxidative stress is reported as a common event in several neurodegenerative diseases. Uncontrolled oxidation of membrane fatty acids triggers a programmed cell death involving iron, ROS, and ferroptosis, promoting cell death. In Alzheimer's disease, the iron content in the brain is significantly increased in vulnerable regions, resulting in a lack of antioxidant defenses and mitochondrial alterations. Iron interacts with glucose metabolism reciprocally. Overall, iron metabolism and accumulation and ferroptosis play a significant role, particularly in the context of diabetes-induced cognitive decline. Iron chelators improve cognitive performance, meaning that brain iron metabolism control reduces neuronal ferroptosis, promising a novel therapeutic approach to cognitive impairment.
Collapse
Affiliation(s)
- Michele Cerasuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Maria Chiara Auriemma
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Maria Ida Maiorino
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Mario Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Fabrizio Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Anna Maria Colangelo
- Laboratory of Neuroscience “R. Levi-Montalcini”, Department of Biotechnology and Biosciences, NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy;
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania ‘‘Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (F.T.); (M.I.M.); (M.C.); (F.E.); (G.P.)
| |
Collapse
|
35
|
Choi Y, Lee WS, Lee J, Park SH, Kim S, Kim KH, Park S, Kim EH, Kim JK. Capacitive Electrode-Based Electric Field Treatments on Redox-Toxic Iron Deposits in Transgenic AD Mouse Models: The Electroceutical Targeting of Alzheimer's Disease Feasibility Study. Int J Mol Sci 2023; 24:ijms24119552. [PMID: 37298502 DOI: 10.3390/ijms24119552] [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: 04/29/2023] [Revised: 05/24/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Iron accumulation in the brain accelerates Alzheimer's disease progression. To cure iron toxicity, we assessed the therapeutic effects of noncontact transcranial electric field stimulation to the brain on toxic iron deposits in either the Aβ fibril structure or the Aβ plaque in a mouse model of Alzheimer's disease (AD) as a pilot study. A capacitive electrode-based alternating electric field (AEF) was applied to a suspension of magnetite (Fe3O4) to measure field-sensitized reactive oxygen species (ROS) generation. The increase in ROS generation compared to the untreated control was both exposure-time and AEF-frequency dependent. The frequency-specific exposure of AEF to 0.7-1.4 V/cm on a magnetite-bound Aβ-fibril or a transgenic Alzheimer's disease (AD) mouse model revealed the degradation of the Aβ fibril or the removal of the Aβ-plaque burden and ferrous magnetite compared to the untreated control. The results of the behavioral tests show an improvement in impaired cognitive function following AEF treatment on the AD mouse model. Tissue clearing and 3D-imaging analysis revealed no induced damage to the neuronal structures of normal brain tissue following AEF treatment. In conclusion, our results suggest that the effective degradation of magnetite-bound amyloid fibrils or plaques in the AD brain by the electro-Fenton effect from electric field-sensitized magnetite offers a potential electroceutical treatment option for AD.
Collapse
Affiliation(s)
- Younshick Choi
- Departments of Biomedical Engineering & Radiology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Won-Seok Lee
- Departments of Biochemistry, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Jaemeun Lee
- Korea R&D Center for Advanced Pharmaceuticals & Evaluation, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Sun-Hyun Park
- Korea R&D Center for Advanced Pharmaceuticals & Evaluation, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Sunwoung Kim
- Departments of Biomedical Engineering & Radiology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Ki-Hong Kim
- Department of Optometry and Vision Science, Daegu Catholic University, Kyungsan 38430, Republic of Korea
| | - Sua Park
- Department of Neurology, Inje University Ilsan Paik Hospital, Koyang 10380, Republic of Korea
| | - Eun Ho Kim
- Departments of Biochemistry, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Jong-Ki Kim
- Departments of Biomedical Engineering & Radiology, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| |
Collapse
|
36
|
Wang H, Song L, Li M, Yang Z, Wang ZC. Association between susceptibility value and cerebral blood flow in the bilateral putamen in patients undergoing hemodialysis. J Cereb Blood Flow Metab 2023; 43:433-445. [PMID: 36284493 PMCID: PMC9941863 DOI: 10.1177/0271678x221134384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hemodialysis (HD) is the most regularly applied replacement therapy for end-stage renal disease, but it may result in brain injuries. The correlation between cerebral blood flow (CBF) alteration and iron deposition has not been investigated in patients undergoing HD. Ferritin level may be a dominant factor in CBF and iron deposition change. We hypothesize that ferritin level might be the key mediator between iron deposition and CBF alteration. The correlation in the putamen was estimated between the susceptibility values and CBF in patients undergoing HD. Compared with healthy controls, patients showed more altered global susceptibility values and CBF. The susceptibility value was negatively correlated with CBF in the putamen in patients. Moreover, the susceptibility value was negatively correlated with ferritin level and positively correlated with serum iron level in the putamen of patients. CBF was positively correlated with ferritin level and negatively correlated with serum iron level in the putamen of patients. These findings indicate that iron dyshomeostasis and vascular damage might exist in the putamen in patients. The results revealed that iron dyshomeostasis and vascular damage in the putamen may be potential neural mechanisms for neurodegenerative processes in patients undergoing HD.
Collapse
Affiliation(s)
- Hao Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lijun Song
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingan Li
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhen-Chang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Guo S, Lei Q, Guo H, Yang Q, Xue Y, Chen R. Edaravone Attenuates Aβ 1-42-Induced Inflammatory Damage and Ferroptosis in HT22 Cells. Neurochem Res 2023; 48:570-578. [PMID: 36333599 DOI: 10.1007/s11064-022-03782-y] [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: 06/02/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022]
Abstract
Ferroptosis and neuroinflammation play a crucial role in the pathogenesis of Alzheimer's disease (AD), and Edaravone (EDA) has been demonstrated to have anti-inflammatory, antioxidant and neuroprotective effects in neurodegenerative diseases. However, the relationship between EDA and ferroptosis in AD is unidentified. This research aimed to elucidate the mechanism of EDA in AD with Aβ 1-42-induced HT22 cells as in vitro cell model. The results showed that EDA could significantly reduce Aβ1-42-induced apoptosis of HT22 cells and formation of pro-inflammatory factors TNF-α, IL-1β and IL-6, prevent the activation of TLR4/NF-κB /NLRP3 signaling pathway, and inhibit ferroptosis and lipid peroxidation. Taken together, EDA contributes to inhibiting neuroinflammatory injury and ferroptosis in Aβ 1-42-induced HT22 cells, and thus may be a potential candidate for the treatment of AD.
Collapse
Affiliation(s)
- Shenglong Guo
- Department II of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Qi Lei
- Department II of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Hena Guo
- Department II of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Qian Yang
- Department II of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Yanli Xue
- Department II of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Ruili Chen
- Department II of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
38
|
Qu Y, Wu Y, Cheng W, Wang D, Zeng L, Wang Y, Li T, Zhang L, Yang J, Sun L, Ai J. Amelioration of cognitive impairment using epigallocatechin-3-gallate in ovariectomized mice fed a high-fat diet involves remodeling with Prevotella and Bifidobacteriales. Front Pharmacol 2023; 13:1079313. [PMID: 36686657 PMCID: PMC9846573 DOI: 10.3389/fphar.2022.1079313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/30/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Estrogen deficiency and a high-fat diet (HFD) are both risk factors for Alzheimer's disease (AD). HFD can accelerate cognitive impairment in estrogen-deficient patients, but there is currently no effective treatment. Epigallocatechin-3-galate (EGCG) is widely studied for its anti-inflammatory, anti-cancer, and anti-neurodegeneration effects. Nevertheless, whether EGCG can ameliorate cognitive impairment in HFD-fed estrogen-deficient mice has not been studied. Methods and Results: Ovariectomized (OVX) mice fed an HFD (HFOVX) for 8 weeks experienced impaired object recognition and spatial memory, but this damage was significantly attenuated by the administration of EGCG at a dose of 45 mg/kg. Through 16S rRNA gene sequencing, we found that HFOVX changed the diversity and structure of the gut microbiota in mice, which could be restored with EGCG. Further analysis showed that HFOVX exposure not only resulted in a decrease of Alloprevotella in Bacteroidetes, Lactobacillaceae in Firmicutes, and Prevotella in Bacteroidetes but also in an increase of Bifidobacteriales in Actinobacteria. EGCG effectively reversed the decrease of Prevotella and inhibited the increase of Bifidobacteriales but had no effect on the decrease of Alloprevotella or Lactobacillaceae or on the increase of Enterorhabdus in HFOVX mice. Additionally, using Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, we found that EGCG significantly reversed the five functional gut microbiota genes elevated by HFOVX, including iron complex transport system substrate-binding protein, iron complex transport system permease protein, 3-oxoacyl- [acyl-carrier protein] reductase, transketolase, and 8-oxo-dGTP diphosphatase. Conclusions: We concluded that EGCG improved cognitive impairment in mice with estrogen deficiency exacerbated by an HFD involved a rebuilding of the disrupted gut microbiota composition.
Collapse
|
39
|
Yang J, Shi X, Wang Y, Ma M, Liu H, Wang J, Xu Z. Multi-Target Neuroprotection of Thiazolidinediones on Alzheimer's Disease via Neuroinflammation and Ferroptosis. J Alzheimers Dis 2023; 96:927-945. [PMID: 37927258 PMCID: PMC10741341 DOI: 10.3233/jad-230593] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 11/07/2023]
Abstract
Alzheimer's disease (AD) is the main cause of dementia in older age. The prevalence of AD is growing worldwide, causing a tremendous burden to societies and families. Due to the complexity of its pathogenesis, the current treatment of AD is not satisfactory, and drugs acting on a single target may not prevent AD progression. This review summarizes the multi-target pharmacological effects of thiazolidinediones (TZDs) on AD. TZDs act as peroxisome proliferator-activated receptor gamma (PPARγ) agonists and long-chain acyl-CoA synthetase family member 4 (ACSL4) inhibitors. TZDs ameliorated neuroinflammation and ferroptosis in preclinical models of AD. Here, we discussed recent findings from clinical trials of pioglitazone in the treatment of AD, ischemic stroke, and atherosclerosis. We also dissected the major limitations in the clinical application of pioglitazone and explained the potential benefit of pioglitazone in AD. We recommend the use of pioglitazone to prevent cognitive decline and lower AD risk in a specific group of patients.
Collapse
Affiliation(s)
- Jiahui Yang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Shi
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yingying Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ming Ma
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hongyu Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaoqi Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongxin Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
40
|
iTRAQ-Based Proteomic Analysis of APP Transgenic Mouse Urine Exosomes. Int J Mol Sci 2022; 24:ijms24010672. [PMID: 36614115 PMCID: PMC9820663 DOI: 10.3390/ijms24010672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is a common dementia disease in the elderly. To get a better understanding of the pathophysiology, we performed a proteomic analysis of the urine exosomes (U-exo) in AD model mice (J20). The polymer precipitation method was used to isolate U-exo from the urine of 3-month-old J20 and wild-type (WT) mice. Neuron-derived exosome (N-exo) was isolated from U-exo by immunoprecipitation. iTRAQ-based MALDI TOF MS/MS was used for proteomic analysis. The results showed that compared to WT, the levels of 61 and 92 proteins were increased in the J20 U-exo and N-exo, respectively. Gene ontology enrichment analysis demonstrated that the sphingolipid catabolic process, ceramide catabolic process, membrane lipid catabolic process, Aβ clearance, and Aβ metabolic process were highly enriched in U-exo and N-exo. Among these, Asah1 was shown to be the key protein in lipid metabolism, and clusterin, ApoE, neprilysin, and ACE were related to Aβ metabolism and clearance. Furthermore, protein-protein interaction analysis identified four protein complexes where clusterin and ApoE participated as partner proteins. Thus, J20 U-exo and N-exo contain proteins related to lipid- and Aβ-metabolism in the early stages of AD, providing a new insight into the underlying pathological mechanism of early AD.
Collapse
|
41
|
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
|
42
|
Liu H, Xie Y, Wang X, Abboud MI, Ma C, Ge W, Schofield CJ. Exploring links between 2-oxoglutarate-dependent oxygenases and Alzheimer's disease. Alzheimers Dement 2022; 18:2637-2668. [PMID: 35852137 PMCID: PMC10083964 DOI: 10.1002/alz.12733] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/12/2022] [Accepted: 06/10/2022] [Indexed: 01/31/2023]
Abstract
Hypoxia, that is, an inadequate oxygen supply, is linked to neurodegeneration and patients with cardiovascular disease are prone to Alzheimer's disease (AD). 2-Oxoglutarate and ferrous iron-dependent oxygenases (2OGDD) play a key role in the regulation of oxygen homeostasis by acting as hypoxia sensors. 2OGDD also have roles in collagen biosynthesis, lipid metabolism, nucleic acid repair, and the regulation of transcription and translation. Many biological processes in which the >60 human 2OGDD are involved are altered in AD patient brains, raising the question as to whether 2OGDD are involved in the transition from normal aging to AD. Here we give an overview of human 2OGDD and critically discuss their potential roles in AD, highlighting possible relationships with synapse dysfunction/loss. 2OGDD may regulate neuronal/glial differentiation through enzyme activity-dependent mechanisms and modulation of their activity has potential to protect against synapse loss. Work linking 2OGDD and AD is at an early stage, especially from a therapeutic perspective; we suggest integrated pathology and in vitro discovery research to explore their roles in AD is merited. We hope to help enable long-term research on the roles of 2OGDD and, more generally, oxygen/hypoxia in AD. We also suggest shorter term empirically guided clinical studies concerning the exploration of 2OGDD/oxygen modulators to help maintain synaptic viability are of interest for AD treatment.
Collapse
Affiliation(s)
- Haotian Liu
- State Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Yong Xie
- State Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijingChina
- National Clinical Research Center for OrthopedicsSports Medicine & RehabilitationDepartment of OrthopedicsGeneral Hospital of Chinese PLABeijingChina
| | - Xia Wang
- State Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Martine I. Abboud
- The Chemistry Research LaboratoryDepartment of Chemistry and the Ineos Oxford Institute for Antimicrobial ResearchUniversity of OxfordOxfordUK
| | - Chao Ma
- Department of Human Anatomy, Histology and EmbryologyNeuroscience CenterNational Human Brain Bank for Development and FunctionInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology & Department of ImmunologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Christopher J. Schofield
- The Chemistry Research LaboratoryDepartment of Chemistry and the Ineos Oxford Institute for Antimicrobial ResearchUniversity of OxfordOxfordUK
| |
Collapse
|
43
|
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
|
44
|
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: 2] [Impact Index Per Article: 1.0] [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
|
45
|
Ferroptosis is involved in regulating perioperative neurocognitive disorders: emerging perspectives. J Neuroinflammation 2022; 19:219. [PMID: 36068571 PMCID: PMC9450301 DOI: 10.1186/s12974-022-02570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
Since the twenty-first century, the development of technological advances in anesthesia and surgery has brought benefits to human health. However, the adverse neurological effects of perioperative-related factors (e.g., surgical trauma, anesthesia, etc.) as stressors cannot be ignored as well. The nervous system appears to be more "fragile" and vulnerable to damage in developing and aging individuals. Ferroptosis is a novel form of programmed cell death proposed in 2012. In recent years, the regulation of ferroptosis to treat cancer, immune system disorders, and neurodegenerative diseases have seen an unprecedented surge of interest. The association of ferroptosis with perioperative neurocognitive disorders has also received much attention. Cognitive impairment can not only affect the individual's quality of life, but also impose a burden on the family and society. Therefore, the search for effective preventive and therapeutic methods to alleviate cognitive impairment caused by perioperative-related factors is a challenge that needs to be urgently addressed. In our review, we first briefly describe the connection between iron accumulation in neurons and impairment of brain function during development and aging. It is followed by a review of the pathways of ferroptosis, mainly including iron metabolism, amino acid metabolism, and lipid metabolism pathway. Furthermore, we analyze the connection between ferroptosis and perioperative-related factors. The surgery itself, general anesthetic drugs, and many other relevant factors in the perioperative period may affect neuronal iron homeostasis. Finally, we summarize the experimental evidence for ameliorating developmental and degenerative neurotoxicity by modulating ferroptosis. The suppression of ferroptosis seems to provide the possibility to prevent and improve perioperative neurocognitive impairment.
Collapse
|
46
|
Yang Y, Wang X, Xiao A, Han J, Wang Z, Wen M. Ketogenic diet prevents chronic sleep deprivation-induced Alzheimer’s disease by inhibiting iron dyshomeostasis and promoting repair via Sirt1/Nrf2 pathway. Front Aging Neurosci 2022; 14:998292. [PMID: 36118706 PMCID: PMC9475074 DOI: 10.3389/fnagi.2022.998292] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
Sleep deprivation (SD) is one of the main risk factors for Alzheimer’s disease (AD), but the underlying mechanism is still unclear. Ketogenic diet (KD) has been shown widely neuroprotective effects but less known about its effect on SD-induced AD. In the present study, a continuous 21 days SD mouse model with or without KD was established. The changes of cognitive function, pathological hallmarks of AD, ferroptosis, and intracellular signal pathways in mice were detected by Morris water maze, ThS staining, diaminobenzidine (DAB)-enhanced Perls’ stain, antioxidant assay, immuno-histochemistry, and western blot. The results showed that KD can prevent the cognitive deficiency, amyloid deposition and hyperphosphorylated tau induced by chronic SD. Analysis of ferroptosis revealed that KD can inhibit iron dyshomeostasis by down-regulating the expression of TfR1 and DMT1 and up-regulating the expression of FTH1, FPN1. Meanwhile, KD alleviated oxidative stress with elevated xCT/GPX4 axis, FSP1 and reduced MDA. In addition, KD could promote neuronal repair by enhancing BDNF and DCX. Further studies demonstrated that KD activated Sirt1/Nrf2 signaling pathway in the hippocampus in SD-exposed mice. Our finding firstly suggested that KD could prevent chronic SD-induced AD by inhibiting ferroptosis and improving the neuronal repair ability via Sirt1/Nrf2 signaling pathway.
Collapse
|
47
|
Serotonin 5-HT 6 Receptor Ligands and Butyrylcholinesterase Inhibitors Displaying Antioxidant Activity-Design, Synthesis and Biological Evaluation of Multifunctional Agents against Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23169443. [PMID: 36012707 PMCID: PMC9409043 DOI: 10.3390/ijms23169443] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Neurodegeneration leading to Alzheimer’s disease results from a complex interplay of a variety of processes including misfolding and aggregation of amyloid beta and tau proteins, neuroinflammation or oxidative stress. Therefore, to address more than one of these, drug discovery programmes focus on the development of multifunctional ligands, preferably with disease-modifying and symptoms-reducing potential. Following this idea, herein we present the design and synthesis of multifunctional ligands and biological evaluation of their 5-HT6 receptor affinity (radioligand binding assay), cholinesterase inhibitory activity (spectroscopic Ellman’s assay), antioxidant activity (ABTS assay) and metal-chelating properties, as well as a preliminary ADMET properties evaluation. Based on the results we selected compound 14 as a well-balanced and potent 5-HT6 receptor ligand (Ki = 22 nM) and human BuChE inhibitor (IC50 = 16 nM) with antioxidant potential expressed as a reduction of ABTS radicals by 35% (150 μM). The study also revealed additional metal-chelating properties of compounds 15 and 18. The presented compounds modulating Alzheimer’s disease-related processes might be further developed as multifunctional ligands against the disease.
Collapse
|
48
|
Doroszkiewicz J, Mroczko B. New Possibilities in the Therapeutic Approach to Alzheimer's Disease. Int J Mol Sci 2022; 23:8902. [PMID: 36012193 PMCID: PMC9409036 DOI: 10.3390/ijms23168902] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 01/17/2023] Open
Abstract
Despite the fact that Alzheimer's disease (AD) is the most common cause of dementia, after many years of research regarding this disease, there is no casual treatment. Regardless of the serious public health threat it poses, only five medical treatments for Alzheimer's disease have been authorized, and they only control symptoms rather than changing the course of the disease. Numerous clinical trials of single-agent therapy did not slow the development of disease or improve symptoms when compared to placebo. Evidence indicates that the pathological alterations linked to AD start many years earlier than a manifestation of the disease. In this pre-clinical period before the neurodegenerative process is established, pharmaceutical therapy might prove invaluable. Although recent findings from the testing of drugs such as aducanumab are encouraging, they should nevertheless be interpreted cautiously. Such medications may be able to delay the onset of dementia, significantly lowering the prevalence of the disease, but are still a long way from having a clinically effective disease-modifying therapy.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland
| |
Collapse
|
49
|
Wang H, Dai JY, He YZ, Xia ZW, Chen XF, Hong ZY, Chai YF. Therapeutic effect and mechanism of Anemarrhenae Rhizoma on Alzheimer’s disease based on multi-platform metabolomics analyses. Front Pharmacol 2022; 13:940555. [PMID: 35991874 PMCID: PMC9385998 DOI: 10.3389/fphar.2022.940555] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Anemarrhenae Rhizoma (AR) has multiple pharmacological activities to prevent and treat Alzheimer’s disease (AD). However, the effect and its molecular mechanism are not elucidated clear. This study aims to evaluate AR’s therapeutic effect and mechanism on AD model rats induced by D-galactose and AlCl3 with serum metabolomics. Behavior study, histopathological observations, and biochemical analyses were applied in the AD model assessment. Gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-QTOF/MS) were combined with multivariate statistical analysis to identify potential biomarkers of AD and evaluate the therapeutic effect of AR on AD from the perspective of metabolomics. A total of 49 biomarkers associated with the AD model were identified by metabolomics, and pathway analysis was performed to obtain the metabolic pathways closely related to the model. With the pre-treatment of AR, 32 metabolites in the serum of AD model rats were significantly affected by AR compared with the AD model group. The regulated metabolites affected by AR were involved in the pathway of arginine biosynthesis, arginine and proline metabolism, ether lipid metabolism, glutathione metabolism, primary bile acid biosynthesis, and steroid biosynthesis. These multi-platform metabolomics analyses were in accord with the results of behavior study, histopathological observations, and biochemical analyses. This study explored the therapeutic mechanism of AR based on multi-platform metabolomics analyses and provided a scientific basis for the application of AR in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Hui Wang
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jian-Ying Dai
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yu-Zhen He
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Zhe-Wei Xia
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xiao-Fei Chen
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Zhan-Ying Hong
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai, China
- *Correspondence: Zhan-Ying Hong,
| | - Yi-Feng Chai
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Naval Medical University, Shanghai, China
| |
Collapse
|