1
|
Rah B, Shafarin J, Qaisar R, Karim A, Hamad M, Muhammad JS. Mouse hindlimb unloading, as a model of simulated microgravity, leads to dysregulated iron homeostasis in liver and skeletal muscle cells. LIFE SCIENCES IN SPACE RESEARCH 2025; 45:7-15. [PMID: 40280644 DOI: 10.1016/j.lssr.2025.01.003] [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: 06/11/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 04/29/2025]
Abstract
Microgravity exposure can impact various physiological systems, yet its specific effects on liver cells remain inadequately studied. To address this gap, we used a hindlimb unloading (HU) mouse model to simulate microgravity conditions and investigate alterations in iron metabolism within liver and skeletal muscle cells. 16-week-old male C57BL/6j mice were divided into three groups: (i) ground-based control (GC), (ii) hindlimb unloading treated with vehicle (HU-v), and (iii) hindlimb unloading treated with deferoxamine (DFO). After three weeks, mice were euthanized, and samples of gastrocnemius muscle, liver, and serum were collected for analysis. The HU-v group exhibited significant muscle and liver cell atrophy compared to the GC group, along with disrupted iron metabolism, as indicated by altered expression of key iron regulatory proteins, including FTH1, FPN, TFR1, IRP-1, HMOX-1, and Hepcidin. In contrast, the DFO group demonstrated restored iron homeostasis, with protein expression patterns resembling those of the GC group. Serum analysis revealed elevated levels of serum iron, ferritin, and transferrin in the DFO group compared to both HU-v and GC groups, albeit with minimal changes in total iron-binding capacity. These findings suggest that simulated microgravity induces iron overload and cellular atrophy in liver and skeletal muscle cells, highlighting the potential therapeutic benefits of iron chelation in such conditions.
Collapse
Affiliation(s)
- Bilal Rah
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates
| | - Jasmin Shafarin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates
| | - Rizwan Qaisar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates
| | - Asima Karim
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Mawieh Hamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates; Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Jibran Sualeh Muhammad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates; Department of Biomedical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom.
| |
Collapse
|
2
|
Liu Y, Wang M, Han W, Guan X, Wang Z, Guo S, Fu P. Multiparametric analysis based on 18F-AV133 PET/MR imaging for clinical application in Parkinson's disease. Eur J Radiol 2025; 187:112074. [PMID: 40194470 DOI: 10.1016/j.ejrad.2025.112074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/18/2025] [Accepted: 03/26/2025] [Indexed: 04/09/2025]
Abstract
OBJECTIVE The progressive loss of dopaminergic neurons and abnormal iron deposition in the central nervous system (CNS) are key pathogenic mechanisms of Parkinson's disease (PD). This study aimed to explore the relationship between iron deposition in specific CNS regions and striatal dysfunction using 18F-AV133 PET/MR imaging. METHODS Based on the Hoehn-Yahr stage, 24 patients with early-stage PD (EPD, stage ≤ 2.5), 17 patients with late-stage PD (LPD, stage ≥ 3), and 30 healthy controls (HCs) were recruited for scale evaluation. The specific uptake ratio (SUR) of striatal subregions was calculated using the occipital cortex as the reference region. Quantitative Susceptibility Mapping (QSM) values of major subcortical nuclei were derived through QSM imaging. Spearman correlation analysis was conducted to assess the relationships between SUR in striatal subregions, QSM values in nuclear groups, and PD clinical symptoms, as well as the correlation between SUR and QSM values. RESULTS Compared to HC, EPD and LPD patients showed significantly reduced VMAT2 distribution in the bilateral caudate nuclei and anteroposterior putamen, particularly in the contralateral posterior putamen. In PD patients, the SUR of striatal subregions and QSM values of the substantia nigra (SN), globus pallidus (GP), and external segment of the GP (GPe) were significantly correlated with disease duration, H&Y stage, UPDRS III score, and NMSS score. Moreover, SUR of striatal subregions was negatively correlated with QSM values in the SN, GP, internal segment of the GP (GPi), and GPe. CONCLUSION Multi-parameter analysis revealed a region-specific correlation between striatal dysfunction and iron deposition in PD, offering new avenues to elucidate the underlying mechanisms of the disease.
Collapse
Affiliation(s)
- Yansong Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China
| | - Mengjiao Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China
| | - Wei Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China
| | - Xinghe Guan
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China
| | - Zeyu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China
| | - Shibo Guo
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China
| | - Peng Fu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China.
| |
Collapse
|
3
|
Sil R, Chakraborti AS. Major heme proteins hemoglobin and myoglobin with respect to their roles in oxidative stress - a brief review. Front Chem 2025; 13:1543455. [PMID: 40070406 PMCID: PMC11893434 DOI: 10.3389/fchem.2025.1543455] [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: 12/11/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Oxidative stress is considered as the root-cause of different pathological conditions. Transition metals, because of their redox-active states, are capable of free radical generation contributing oxidative stress. Hemoglobin and myoglobin are two major heme proteins, involved in oxygen transport and oxygen storage, respectively. Heme prosthetic group of heme proteins is a good reservoir of iron, the most abundant transition metal in human body. Although iron is tightly bound in the heme pocket of these proteins, it is liberated under specific circumstances yielding free ferrous iron. This active iron can react with H2O2, a secondary metabolite, forming hydroxyl radical via Fenton reaction. Hydroxyl radical is the most harmful free radical among all the reactive oxygen species. It causes oxidative stress by damaging lipid membranes, proteins and nucleic acids, activating inflammatory pathways and altering membrane channels, resulting disease conditions. In this review, we have discussed how heme-irons of hemoglobin and myoglobin can promote oxidative stress under different pathophysiological conditions including metabolic syndrome, diabetes, cardiovascular, neurodegenerative and renal diseases. Understanding the association of heme proteins to oxidative stress may be important for knowing the complications as well as therapeutic management of different pathological conditions.
Collapse
Affiliation(s)
| | - Abhay Sankar Chakraborti
- Department of Biophysics, Molecular Biology and Bioinformatics, University College of Science, University of Calcutta, Kolkata, India
| |
Collapse
|
4
|
Gomes BAQ, dos Santos SM, Gato LDS, Espíndola KMM, da Silva RKM, Davis K, Navegantes-Lima KC, Burbano RMR, Romao PRT, Coleman MD, Monteiro MC. Alpha-Lipoic Acid Reduces Neuroinflammation and Oxidative Stress Induced by Dapsone in an Animal Model. Nutrients 2025; 17:791. [PMID: 40077661 PMCID: PMC11901491 DOI: 10.3390/nu17050791] [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: 12/01/2024] [Revised: 02/07/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Chronic treatment with dapsone (DDS) has been linked to adverse reactions involving all organ systems, such as dapsone hypersensitivity syndrome, methemoglobinemia and hemolytic anemia, besides neuroinflammation and neurodegeneration due to iron accumulation and oxidative stress. These effects probably occur due to the presence of its toxic metabolite DDS-NOH, which can generate reactive oxygen species (ROS) and iron overload. In this sense, antioxidant compounds with chelating properties, such as alpha-lipoic acid (ALA), may be an interesting adjuvant therapy strategy in treating or preventing these effects. Thus, the aim of this study was to evaluate the effects of ALA on oxidative and neuroinflammatory changes caused by DDS treatment in the prefrontal cortex and hippocampus of mice. Materials and Methods:Mus musculus male mice that were pre-treated with DDS (40 mg/kg) and post-treated with ALA (25 mg/kg) underwent analyses for oxidative stress, antioxidant capacity, cytokine expression and microglial/astrocytic activity. Results: DDS did not activate macrophages/microglia or astrocytes in the prefrontal cortex but induced their activation in the hippocampus. ALA stimulated a protective microglial profile and reduced astrocyte reactivity, especially in the hippocampus. DDS increased the pro-inflammatory cytokine IL-1β and reduced brain-derived neurotrophic factor (BDNF), effects reversed by ALA. DDS also reduced antioxidant capacity (TEAC, GSH, SOD, CAT) and increased oxidative damage (lipid peroxidation, iron accumulation), while ALA restored antioxidant levels and reduced oxidative stress. Conclusions: ALA was able to reduce the effects of DDS, such as reducing microglial and astrocytic activation, as well as to decrease the levels of pro-inflammatory cytokines and increase BDNF, in addition to increasing antioxidant capacity and reducing oxidative damage caused by iron accumulation. Therefore, ALA is considered a useful and promising therapeutic alternative for the treatment of diseases related to oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Bruno Alexandre Quadros Gomes
- Postgraduate Program in Neuroscience and Cell Biology, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil; (B.A.Q.G.); (S.M.d.S.); (K.M.M.E.); (R.K.M.d.S.)
| | - Savio Monteiro dos Santos
- Postgraduate Program in Neuroscience and Cell Biology, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil; (B.A.Q.G.); (S.M.d.S.); (K.M.M.E.); (R.K.M.d.S.)
- Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil;
| | - Lucas da Silva Gato
- Laboratory Immunology, Microbiology and In Vitro Assays (LABEIM), Faculty of Pharmacy, Federal University of Pará/UFPA, Belém 66075-110, PA, Brazil;
| | - Kaio Murilo Monteiro Espíndola
- Postgraduate Program in Neuroscience and Cell Biology, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil; (B.A.Q.G.); (S.M.d.S.); (K.M.M.E.); (R.K.M.d.S.)
- Postgraduate Program in Pharmacology and Biochemistry, Faculty of Pharmacy, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil;
| | - Rana Karen Mesquita da Silva
- Postgraduate Program in Neuroscience and Cell Biology, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil; (B.A.Q.G.); (S.M.d.S.); (K.M.M.E.); (R.K.M.d.S.)
| | - Kelly Davis
- Postgraduate Program in Pharmacology and Biochemistry, Faculty of Pharmacy, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil;
| | - Kely Campos Navegantes-Lima
- Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil;
- Laboratory Immunology, Microbiology and In Vitro Assays (LABEIM), Faculty of Pharmacy, Federal University of Pará/UFPA, Belém 66075-110, PA, Brazil;
| | | | - Pedro Roosevelt Torres Romao
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil;
| | - Michael D. Coleman
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK;
| | - Marta Chagas Monteiro
- Postgraduate Program in Neuroscience and Cell Biology, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil; (B.A.Q.G.); (S.M.d.S.); (K.M.M.E.); (R.K.M.d.S.)
- Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil;
- Laboratory Immunology, Microbiology and In Vitro Assays (LABEIM), Faculty of Pharmacy, Federal University of Pará/UFPA, Belém 66075-110, PA, Brazil;
- Postgraduate Program in Pharmacology and Biochemistry, Faculty of Pharmacy, Federal University of Pará/UFPA, Rua Augusto Corrêa 01, Bairro Guamá, Belém 66075-110, PA, Brazil;
| |
Collapse
|
5
|
Cornelis MC, Fazlollahi A, Bennett DA, Schneider JA, Ayton S. Genetic Markers of Postmortem Brain Iron. J Neurochem 2025; 169:e16309. [PMID: 39918201 PMCID: PMC11804167 DOI: 10.1111/jnc.16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025]
Abstract
Brain iron (Fe) dyshomeostasis is implicated in neurodegenerative diseases. Genome-wide association studies (GWAS) have identified plausible loci correlated with peripheral levels of Fe. Systemic organs and the brain share several Fe regulatory proteins but there likely exist different homeostatic pathways. We performed the first GWAS of inductively coupled plasma mass spectrometry measures of postmortem brain Fe from 635 Rush Memory and Aging Project (MAP) participants. Sixteen single nucleotide polymorphisms (SNPs) associated with Fe in at least one of four brain regions were measured (p < 5 × 10-8). Promising SNPs (p < 5 × 10-6) were followed up for replication in published GWAS of blood, spleen, and brain imaging Fe traits and mapped to candidate genes for targeted cortical transcriptomic and epigenetic analysis of postmortem Fe in MAP. Results for SNPs previously associated with other Fe traits were also examined. Ninety-eight SNPs associated with postmortem brain Fe were at least nominally (p < 0.05) associated with one or more related Fe traits. Most novel loci identified had no direct links to Fe regulatory pathways but rather endoplasmic reticulum-Golgi trafficking (SORL1, SORCS2, MARCH1, CLTC), heparan sulfate (HS3ST4, HS3ST1), and coenzyme A (SLC5A6, PANK3); supported by nearest gene function and omic analyses. We replicated (p < 0.05) several previously published Fe loci mapping to candidate genes in cellular and systemic Fe regulation. Finally, novel loci (BMAL, COQ5, SLC25A11) and replication of prior loci (PINK1, PPIF, LONP1) lend support to the role of circadian rhythms and mitochondria function in Fe regulation more generally. In summary, we provide support for novel loci linked to pathways that may have greater relevance to brain Fe accumulation; some of which are implicated in neurodegeneration. However, replication of a subset of prior loci for blood Fe suggests that genetic determinants or biological pathways underlying Fe accumulation in the brain are not completely distinct from those of Fe circulating in the periphery.
Collapse
Affiliation(s)
- Marilyn C. Cornelis
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Amir Fazlollahi
- Department of Radiology, Royal Melbourne HospitalUniversity of MelbourneMelbourneVictoriaAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
| | | | | | - Scott Ayton
- The Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
6
|
Xu ZH, Xie MM, Xie CL, Yang XW, Wang JS. Deep-Sea-Derived Isobisvertinol Targets TLR4 to Exhibit Neuroprotective Activity via Anti-Inflammatory and Ferroptosis-Inhibitory Effects. Mar Drugs 2025; 23:49. [PMID: 39852551 PMCID: PMC11766622 DOI: 10.3390/md23010049] [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: 12/20/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Neuroinflammation and neuronal cell death are leading causes of death in the elderly and underlie various neurodegenerative diseases. These diseases involve complex pathophysiological mechanisms, including inflammatory responses, oxidative stress, and ferroptosis. Compounds derived from deep-sea fungi exhibit low toxicity and potent neuroprotective effects, offering a promising source for drug development. In this study, we isolated 44 natural products from deep-sea-derived fungi and identified isobisvertinol (17) as a compound with anti-inflammatory and ferroptosis-inhibiting effects. Using LPS-induced microglial inflammation and RSL3-induced neuronal ferroptosis models, we found that 17 targets TLR4 to provide neuroprotection. Molecular docking studies revealed that 17 inhibits TLR4 activation by occupying the hydrophobic pocket at the TLR4-MD2 binding site. Additionally, 17 suppresses TLR4, reducing p38 MAPK phosphorylation, and inhibits ferroptosis by decreasing lipid peroxidation and modulating mitochondrial membrane potential. Metabolomic analysis showed that 17 rescues alterations in multiple metabolic pathways induced by RSL3 and increases levels of antioxidant metabolites, including glutamine, glutamate, and glutathione. In summary, our results indicate that isobisvertinol (17) targets TLR4 in neural cells to reduce inflammation and inhibit p38 MAPK phosphorylation, while regulating metabolic pathways, mainly GSH synthesis, to provide antioxidant effects and prevent ferroptosis in neurons.
Collapse
Affiliation(s)
- Zi-Han Xu
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing 210094, China;
- School of Basic Medicine and Life Science, Hainan Academy of Medical Sciences, Hainan Medical University, 3 Xueyuan Road, Haikou 571199, China;
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China;
| | - Ming-Min Xie
- Key Laboratory of Marine Genetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, 184 Daxue Road, Xiamen 361005, China;
| | - Chun-Lan Xie
- School of Basic Medicine and Life Science, Hainan Academy of Medical Sciences, Hainan Medical University, 3 Xueyuan Road, Haikou 571199, China;
| | - Xian-Wen Yang
- School of Basic Medicine and Life Science, Hainan Academy of Medical Sciences, Hainan Medical University, 3 Xueyuan Road, Haikou 571199, China;
| | - Jun-Song Wang
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing 210094, China;
| |
Collapse
|
7
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [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: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
8
|
Hosseininasab SSM, Ebrahimi R, Yaghoobpoor S, Kazemi K, Khakpour Y, Hajibeygi R, Mohamadkhani A, Fathi M, Vakili K, Tavasol A, Tutunchian Z, Fazel T, Fathi M, Hajiesmaeili M. Alzheimer's disease and infectious agents: a comprehensive review of pathogenic mechanisms and microRNA roles. Front Neurosci 2025; 18:1513095. [PMID: 39840010 PMCID: PMC11747386 DOI: 10.3389/fnins.2024.1513095] [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: 10/17/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Alzheimer's Disease (AD) is the most prevalent type of dementia and is characterized by the presence of senile plaques and neurofibrillary tangles. There are various theories concerning the causes of AD, but the connection between viral and bacterial infections and their potential role in the pathogenesis of AD has become a fascinating area of research for the field. Various viruses such as Herpes simplex virus 1 (HSV-1), Epstein-Barr virus (EBV), Cytomegalovirus (CMV), influenza viruses, and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), as well as bacteria such as Chlamydia pneumoniae (CP), Helicobacter pylori (HP), Porphyromonas gingivalis (P. gingivalis), Spirochetes and eukaryotic unicellular parasites (e.g., Toxoplasma gondii), have been linked to AD due to their ability to activate the immune system, induce inflammation and increase oxidative stress, thereby leading to cognitive decline and AD. In addition, microRNAs (miRNAs) might play a crucial role in the pathogenesis mechanisms of these pathogens since they are utilized to target various protein-coding genes, allowing for immune evasion, maintaining latency, and suppressing cellular signaling molecules. Also, they can regulate gene expression in human cells. This article provides an overview of the association between AD and various infectious agents, with a focus on the mechanisms by which these pathogens may be related to the pathogenesis of AD. These findings suggest important areas for further research to be explored in future studies.
Collapse
Affiliation(s)
- Seyyed Sam Mehdi Hosseininasab
- Critical Care Quality Improvement Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Yaghoobpoor
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiarash Kazemi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Khakpour
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramtin Hajibeygi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashraf Mohamadkhani
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arian Tavasol
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Tutunchian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tara Fazel
- Student Research Committee, School of International Campus, Guilan University of Medical Sciences, Tehran, Iran
| | - Mohammad Fathi
- Department of Anesthesiology, Critical Care Quality Improvement Research Center, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Hajiesmaeili
- Critical Care Quality Improvement Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Althobaiti NA. Heavy metals exposure and Alzheimer's disease: Underlying mechanisms and advancing therapeutic approaches. Behav Brain Res 2025; 476:115212. [PMID: 39187176 DOI: 10.1016/j.bbr.2024.115212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Heavy metals such as lead, cadmium, mercury, and arsenic are prevalent in the environment due to both natural and anthropogenic sources, leading to significant public health concerns. These heavy metals are known to cause damage to the nervous system, potentially leading to a range of neurological conditions including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and attention-deficit hyperactivity disorder (ADHD). The present study examines the complex relationship between heavy metal exposure and AD, focusing on the underlying mechanisms of toxicity and potential therapeutic approaches. This review article highlights how these metals can impair brain function through mechanisms such as oxidative stress, inflammation, and neurotransmitter disruption, ultimately contributing to neurodegenerative diseases like AD. It also addresses the challenges in diagnosing heavy metal-induced cognitive impairments and emphasizes the need for further research to explore effective treatment strategies and preventive measures against heavy metal exposure.
Collapse
Affiliation(s)
- Norah A Althobaiti
- Biology Department, College of Science and Humanities, Shaqra University, Saudi Arabia.
| |
Collapse
|
10
|
Deng L, Luo Q, Liu Y, Wang Y, Xiong Z, Wang H, Zhao L, Jia L, Shi R, Huang C, Chen Z. Progressive iron overload in middle-aged mice impairs olfactory function, triggers lipid oxidation and induces apoptosis. Front Pharmacol 2024; 15:1506944. [PMID: 39749201 PMCID: PMC11693683 DOI: 10.3389/fphar.2024.1506944] [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: 10/06/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction This study aims to investigate the progressive impact of chronic iron overload on the olfactory bulb, a region significantly affected in early neurodegenerative diseases like Parkinson's and Alzheimer's. The focus is on understanding how iron accumulation leads to oxidative stress, mitochondrial dysfunction, and neuronal damage over time in middle-aged mice. Method The mice were continuously administered FC for a duration of 16 weeks, and the olfactory behavior of the mice was observed at intervals of 4 weeks. Inductively coupled plasma mass spectrometry (ICP-MS) was employed to detect alterations in iron content within the olfactory bulb of the mice, while levels of lipid peroxidation and antioxidant indexes were assessed using biochemical kits. Additionally, western blotting and qPCR techniques were utilized to analyze transcriptional and expression changes in proteins and genes related to iron metabolism. Furthermore, microstructural modifications as well as mitochondrial observations were conducted through paraffin sectioning and transmission electron microscopy (TEM). Result A significant and progressive increase in iron accumulation in the olfactory bulb, starting from week 8 and peaking at week 16. This accumulation coincided with a decline in olfactory function observed at week 12. Key markers of oxidative stress, such as 4-HNE and MDA, were elevated in specific layers, and antioxidant defenses were reduced. Mitochondrial damage became evident from week 8, with caspase-3 activation indicating increased apoptosis, particularly in the granular layer. This study is to demonstrate the link between chronic iron overload and progressive olfactory dysfunction in the context of neurodegenerative diseases. It provides evidence that iron-induced oxidative stress and mitochondrial damage in the olfactory bulb contribute to early sensory deficits, suggesting that the olfactory bulb's selective vulnerability can serve as an early biomarker for neurodegenerative conditions. Conclusion Chronic iron overload leads to progressive oxidative damage, mitochondrial dysfunction, and apoptosis in the olfactory bulb, causing sensory deficits. Targeting iron accumulation and oxidative damage may offer new strategies for early intervention in neurodegenerative diseases, highlighting the importance of addressing iron dysregulation.
Collapse
Affiliation(s)
- Lin Deng
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, China
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Yucong Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Yao Wang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Zongliang Xiong
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Hongping Wang
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, China
| | - Lu Zhao
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, China
| | - Lanlan Jia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Riyi Shi
- Department of Basic Medical Sciences, Center for Paralysis Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| |
Collapse
|
11
|
Rafati N, Zarepour A, Bigham A, Khosravi A, Naderi-Manesh H, Iravani S, Zarrabi A. Nanosystems for targeted drug Delivery: Innovations and challenges in overcoming the Blood-Brain barrier for neurodegenerative disease and cancer therapy. Int J Pharm 2024; 666:124800. [PMID: 39374818 DOI: 10.1016/j.ijpharm.2024.124800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
The evolution of sophisticated nanosystems has revolutionized biomedicine, notably in treating neurodegenerative diseases and cancer. These systems show potential in delivering medication precisely to affected tissues, improving treatment effectiveness while minimizing side effects. Nevertheless, a major hurdle in targeted drug delivery is breaching the blood-brain barrier (BBB), a selective shield separating the bloodstream from the brain and spinal cord. The tight junctions between endothelial cells in brain capillaries create a formidable physical barrier, alongside efflux transporters that expel harmful molecules. This presents a notable challenge for brain drug delivery. Nanosystems present distinct advantages in overcoming BBB challenges, offering enhanced drug efficacy, reduced side effects, improved stability, and controlled release. Despite their promise, challenges persist, such as the BBB's regional variability hindering uniform drug distribution. Efflux transporters can also limit therapeutic agent efficacy, while nanosystem toxicity necessitates rigorous safety evaluations. Understanding the long-term impact of nanomaterials on the brain remains crucial. Additionally, addressing nanosystem scalability, cost-effectiveness, and safety profiles is vital for widespread clinical implementation. This review delves into the advancements and obstacles of advanced nanosystems in targeted drug delivery for neurodegenerative diseases and cancer therapy, with a focus on overcoming the BBB.
Collapse
Affiliation(s)
- Nesa Rafati
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Ashkan Bigham
- Institute of Polymers, Composites, and Biomaterials, National Research Council (IPCB-CNR), Naples 80125, Italy; Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkiye
| | - Hossein Naderi-Manesh
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran; Departments of Biophysics, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran.
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
12
|
Ferretti S, Zanella I. The Underestimated Role of Iron in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:12987. [PMID: 39684697 DOI: 10.3390/ijms252312987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The term frontotemporal dementia (FTD) comprises a group of neurodegenerative disorders characterized by the progressive degeneration of the frontal and temporal lobes of the brain with language impairment and changes in cognitive, behavioral and executive functions, and in some cases motor manifestations. A high proportion of FTD cases are due to genetic mutations and inherited in an autosomal-dominant manner with variable penetrance depending on the implicated gene. Iron is a crucial microelement that is involved in several cellular essential functions in the whole body and plays additional specialized roles in the central nervous system (CNS) mainly through its redox-cycling properties. Such a feature may be harmful under aerobic conditions, since it may lead to the generation of highly reactive hydroxyl radicals. Dysfunctions of iron homeostasis in the CNS are indeed involved in several neurodegenerative disorders, although it is still challenging to determine whether the dyshomeostasis of this essential but harmful metal is a direct cause of neurodegeneration, a contributor factor or simply a consequence of other neurodegenerative mechanisms. Unlike many other neurodegenerative disorders, evidence of the dysfunction in brain iron homeostasis in FTD is still scarce; nonetheless, the recent literature intriguingly suggests its possible involvement. The present review aims to summarize what is currently known about the contribution of iron dyshomeostasis in FTD based on clinical, imaging, histological, biochemical and molecular studies, further suggesting new perspectives and offering new insights for future investigations on this underexplored field of research.
Collapse
Affiliation(s)
- Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Medical Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
13
|
Zhao X, Zhang M, He J, Li X, Zhuang X. Emerging insights into ferroptosis in cholangiocarcinoma (Review). Oncol Lett 2024; 28:606. [PMID: 39483963 PMCID: PMC11526429 DOI: 10.3892/ol.2024.14739] [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: 05/20/2024] [Accepted: 08/21/2024] [Indexed: 11/03/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor that arises within the biliary system, which exhibits a progressively increasing incidence and a poor patient prognosis. A thorough understanding of the molecular pathogenesis that drives the progression of CCA is essential for the development of effective molecular target therapeutic approaches. Ferroptosis is driven by excessive iron accumulation and catalysis, lipid peroxidation and the failure of antioxidant defense systems. Key molecular targets of iron metabolism, lipid metabolism and antioxidant defense systems involve molecules such as transferrin receptor, ACSL4 and GPX4, respectively. Inhibitors of ferroptosis include ferrostatin-1, liproxstatin-1, vitamin E and coenzyme Q10. By contrast, compounds such as erastin, RSL3 and FIN56 have been identified as inducers of ferroptosis. Ferroptosis serves a notable role in the onset and progression of CCA. CCA cells exhibit high sensitivity to ferroptosis and aberrant iron metabolism in these cells increases oxidative stress and iron accumulation. The induction of ferroptosis markedly reduces the ability of CCA cells to proliferate and migrate. Certain ferroptosis agonists, such as RSL3 and erastin, cause lipid peroxide build up and GPX4 inhibition to induce ferroptosis in CCA cells. Current serological markers, such as CA-199, have low specificity and cause difficulties in the diagnosis of CCA. However, novel techniques, such as non-invasive liquid biopsy and assays for oxidative stress markers and double-cortin-like kinase 1, could improve diagnostic accuracy. CCA is primarily treated with surgery and chemotherapy. A close association between the progression of CCA with ferroptosis mechanisms and related regulatory pathways has been demonstrated. Therefore, it could be suggested that multi-targeted therapeutic approaches, such as ferroptosis inducers, iron chelating agents and novel modulators such as YL-939, may improve treatment efficacy. Iron death-related genes, such as GPX4, that are highly expressed in CCA and are associated with a poor prognosis for patients may represent potential prognostic markers for CCA. The present review focused on molecular targets such as p53 and ACSL4, the process of targeted medications in combination with PDT in CCA and the pathways of lipid peroxidation, the Xc-system and GSH-GPX4 in ferroptosis. The present review thus offered novel perspectives to improve the current understanding of CCA.
Collapse
Affiliation(s)
- Xiaoyue Zhao
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250002, P.R. China
| | - Miao Zhang
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250002, P.R. China
| | - Jing He
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250002, P.R. China
| | - Xin Li
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250002, P.R. China
| | - Xuewei Zhuang
- Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, Jinan, Shandong 250002, P.R. China
| |
Collapse
|
14
|
Kim S, Jung UJ, Kim SR. Role of Oxidative Stress in Blood-Brain Barrier Disruption and Neurodegenerative Diseases. Antioxidants (Basel) 2024; 13:1462. [PMID: 39765790 PMCID: PMC11673141 DOI: 10.3390/antiox13121462] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Upregulation of reactive oxygen species (ROS) levels is a principal feature observed in the brains of neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD). In these diseases, oxidative stress can disrupt the blood-brain barrier (BBB). This disruption allows neurotoxic plasma components, blood cells, and pathogens to enter the brain, leading to increased ROS production, mitochondrial dysfunction, and inflammation. Collectively, these factors result in protein modification, lipid peroxidation, DNA damage, and, ultimately, neural cell damage. In this review article, we present the mechanisms by which oxidative damage leads to BBB breakdown in brain diseases. Additionally, we summarize potential therapeutic approaches aimed at reducing oxidative damage that contributes to BBB disruption in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
15
|
Wu J, Wu J, Chen T, Cai J, Ren R. Protein aggregation and its affecting mechanisms in neurodegenerative diseases. Neurochem Int 2024; 180:105880. [PMID: 39396709 DOI: 10.1016/j.neuint.2024.105880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Protein aggregation serves as a critical pathological marker in a spectrum of neurodegenerative diseases (NDs), including the formation of amyloid β (Aβ) and Tau neurofibrillary tangles in Alzheimer's disease, as well as α-Synuclein (α-Syn) aggregates in Parkinson's disease, Parkinson's disease-related dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). A significant proportion of patients with amyotrophic lateral sclerosis (ALS) exhibit TDP-43 aggregates. Moreover, a confluence of brain protein pathologies, such as Aβ, Tau, α-Syn, and TDP-43, has been identified in individual NDs cases, highlighting the intricate interplay among these proteins that is garnering heightened scrutiny. Importantly, protein aggregation is modulated by an array of factors, with burgeoning evidence suggesting that it frequently results from perturbations in protein homeostasis, influenced by the cellular membrane milieu, metal ion concentrations, post-translational modifications, and genetic mutations. This review delves into the pathological underpinnings of protein aggregation across various NDs and elucidates the intercommunication among disparate proteins within the same disease context. Additionally, we examine the pathogenic mechanisms by which diverse factors impinge upon protein aggregation, offering fresh perspectives for the future therapeutic intervention of NDs.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jianan Wu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jing Cai
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
16
|
Marshall Moscon S, Neely E, Proctor E, Connor J. A common variant in the iron regulatory gene (Hfe) alters the metabolic and transcriptional landscape in brain regions vulnerable to neurodegeneration. J Neurochem 2024; 168:3132-3153. [PMID: 39072788 DOI: 10.1111/jnc.16171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/13/2024] [Accepted: 06/19/2024] [Indexed: 07/30/2024]
Abstract
The role of iron dyshomeostasis in neurodegenerative disease has implicated the involvement of genes that regulate brain iron. The homeostatic iron regulatory gene (HFE) has been at the forefront of these studies given the role of the H63D variant (H67D in mice) in increasing brain iron load. Despite iron's role in oxidative stress production, H67D mice have shown robust protection against neurotoxins and improved recovery from intracerebral hemorrhage. Previous data support the notion that H67D mice adapt to the increased brain iron concentrations and hence develop a neuroprotective environment. This adaptation is particularly evident in the lumbar spinal cord (LSC) and ventral midbrain (VM), both relevant to neurodegeneration. We studied C57BL6/129 mice with homozygous H67D compared to WT HFE. Immunohistochemistry was used to analyze dopaminergic (in the VM) and motor (in the LSC) neuron population maturation in the first 3 months. Immunoblotting was used to measure protein carbonyl content and the expression of oxidative phosphorylation complexes. Seahorse assay was used to analyze metabolism of mitochondria isolated from the LSC and VM. Finally, a Nanostring transcriptomic analysis of genes relevant to neurodegeneration within these regions was performed. Compared to WT mice, we found no difference in the viability of motor neurons in the LSC, but the dopaminergic neurons in H67D mice experienced significant decline before 3 months of age. Both regions in H67D mice had alterations in oxidative phosphorylation complex expression indicative of stress adaptation. Mitochondria from both regions of H67D mice demonstrated metabolic differences compared to WT. Transcriptional differences in these regions of H67D mice were related to cell structure and adhesion as well as cell signaling. Overall, we found that the LSC and VM undergo significant and distinct metabolic and transcriptional changes in adaptation to iron-related stress induced by the H67D HFE gene variant.
Collapse
Affiliation(s)
- Savannah Marshall Moscon
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Elizabeth Neely
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Elizabeth Proctor
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - James Connor
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
17
|
Rowaiye A, Ibeanu GC, Bur D, Nnadi S, Mgbeke OE, Morikwe U. Gut microbiota alteration - Cancer relationships and synbiotic roles in cancer therapies. THE MICROBE 2024; 4:100096. [DOI: 10.1016/j.microb.2024.100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Jin H, Liu J, Wang D. Antioxidant Potential of Exosomes in Animal Nutrition. Antioxidants (Basel) 2024; 13:964. [PMID: 39199210 PMCID: PMC11351667 DOI: 10.3390/antiox13080964] [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: 06/28/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
This review delves into the advantages of exosomes as novel antioxidants in animal nutrition and their potential for regulating oxidative stress. Although traditional nutritional approaches promote oxidative stress defense systems in mammalian animals, several issues remain to be solved, such as low bioavailability, targeted tissue efficiency, and high-dose by-effect. As an important candidate offering regulation opportunities concerned with cellular communication, disease prevention, and physiology regulation in multiple biological systems, the potential of exosomes in mediating redox status in biological systems has not been well described. A previously reported relationship between redox system regulation and circulating exosomes suggested exosomes as a fundamental candidate for both a regulator and biomarker for a redox system. Herein, we review the effects of oxidative stress on exosomes in animals and the potential application of exosomes as antioxidants in animal nutrition. Then, we highlight the advantages of exosomes as redox regulators due to their higher bioavailability and physiological heterogeneity-targeted properties, providing a theoretical foundation and feed industry application. Therefore, exosomes have shown great potential as novel antioxidants in the field of animal nutrition. They can overcome the limitations of traditional antioxidants in terms of dosage and side effects, which will provide unprecedented opportunities in nutritional management and disease prevention, and may become a major breakthrough in the field of animal nutrition.
Collapse
Affiliation(s)
| | | | - Diming Wang
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (H.J.); (J.L.)
| |
Collapse
|
19
|
Garbossa L, Joaquim L, Danielski LG, Goldim MPDS, Machado RS, Metzker K, Bernades G, Lanzzarin E, Bagio E, Farias AD, Rosa ND, Medeiros FDD, Carli RJD, Oliveira BH, Ferreira NC, Palandi J, Bobinski F, Martins DF, Fortunato JJ, Barichello T, Petronilho F. The effect of modafinil on passive avoidance memory, brain level of BDNF and oxidative stress markers in sepsis survivor rats. Int J Neurosci 2024; 134:849-857. [PMID: 36448768 DOI: 10.1080/00207454.2022.2154076] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 08/25/2021] [Accepted: 11/10/2021] [Indexed: 12/03/2022]
Abstract
Material and method: Male Wistar rats (250-350g) were submitted to CLP, or sham as control, and divided into the sham + water, sham + MD (300 mg/kg), CLP + water, and CLP + MD (300 mg/kg) groups. Ten days after the administration of MD and CLP, the rats were submitted to a memory test by passive avoidance apparatus being sacrificed. The nitrite and nitrate (N/N) concentration, myeloperoxidase (MPO) and catalase (CAT) activity, lipid and protein oxidative damage, and brain-derived neurotrophic factor (BDNF) levels were measured in the prefrontal cortex and hippocampus. Results: The passive avoidance test verified an increase in the latency time compared training and test section in the groups sham + water and CLP + MD. Decreased N/N concentration and MPO activity were verified in the prefrontal cortex of rats submitted to CLP and MD treatment, as well as reduced protein and lipid oxidative damage in the hippocampus, which was accompanied by increased CAT activity and BDNF levels.Conclusion: Our data indicate the role of MD in attenuating oxidative stress parameters, the alteration of BDNF, and an improvement in memory impairment in rats ten days after induction of sepsis.
Collapse
Affiliation(s)
- Leandro Garbossa
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Larissa Joaquim
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Lucineia Gainski Danielski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | | | - Richard Simon Machado
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Kiuanne Metzker
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Gabriela Bernades
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Everton Lanzzarin
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Erick Bagio
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Adriele de Farias
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Naiana da Rosa
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabiana Durante de Medeiros
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Raquel Jaconi de Carli
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Bruna Hoffman Oliveira
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Nivaldo Correia Ferreira
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Juliete Palandi
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Daniel Fernandes Martins
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Jucelia Jeremias Fortunato
- Programa de Pos graduação em Ciências da Saúde, University of Southern Santa Catarina (UNISUL), Tubarão, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| |
Collapse
|
20
|
Girón-Hernández J, Rodríguez YB, Corbezzolo N, Blanco DO, Gutiérrez CC, Cheung W, Gentile P. Exploiting residual cocoa biomass to extract advanced materials as building blocks for manufacturing nanoparticles aimed at alleviating formation-induced oxidative stress on human dermal fibroblasts. NANOSCALE ADVANCES 2024; 6:3809-3824. [PMID: 39050955 PMCID: PMC11265571 DOI: 10.1039/d4na00248b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/27/2024] [Indexed: 07/27/2024]
Abstract
The global adoption of by-product valorisation processes aligns with the circular economy framework, ensuring sustainability in the agricultural sector. In cocoa production, residual biomass can offer the opportunity to extract advanced materials, contributing to nanotherapeutic solutions for biomedical applications. This study explores extraction processes for valorising cocoa pod husks (CPHs) and optimising valuable cocoa-derived biocompounds for enhanced health benefits. Various extraction processes are compared, revealing the significant influence of CPH powder amount and extraction time. Furthermore, metabolic analysis identifies 124 compounds in the metabolite mix, including tartaric acid, gluconic acid and bioactive agents with antioxidant properties, resulting in a high total phenolic content of 3.88 ± 0.06 mg g-1. Moreover, the extracted pectin, obtained through alkaline and enzymatic routes, shows comparable yields but exhibits superior antioxidant capacity compared to commercial pectin. The study progresses to using these extracted biocompounds to develop Layer-by-Layer multifunctionalised nanoparticles (LbL-MNPs). Physico-chemical characterisation via ζ-potential, FTIR-ATR, and XPS confirms the successful multilayer coating on mesoporous silica nanoparticles (MNPs). TEM analysis demonstrates a uniform and spherical nanoparticle morphology, with a size increase after coating. In vitro biological characterisation with neo-dermal human fibroblast cells reveals enhanced metabolic activity and biocompatibility of LbL-MNPs compared to bare MNPs. Also, the engineered nanoparticles demonstrate a protective effect against H2O2-induced intracellular oxidative stress on human dermal fibroblast cell lines, showcasing their potential as antioxidant carriers for biomedical applications.
Collapse
Affiliation(s)
- Joel Girón-Hernández
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University NE1 8ST Newcastle Upon Tyne UK
| | - Yeison Barrios Rodríguez
- i-Food, Instituto Universitario de Ingeniería de Alimentos-FoodUPV, Universitat Politècnica de València 46021 Valencia Spain
- Centro Surcolombiano de Investigación en Café (CESURCAFÉ), Universidad Surcolombiana 410010 Neiva Colombia
| | - Noemi Corbezzolo
- School of Engineering, Newcastle University NE1 7RU Newcastle Upon Tyne UK
| | - Dayana Orozco Blanco
- Centro Surcolombiano de Investigación en Café (CESURCAFÉ), Universidad Surcolombiana 410010 Neiva Colombia
| | - Carlos Carranza Gutiérrez
- Escuela de Ciencias Agrícolas, Pecuarias y del Medio Ambiente, Universidad Nacional Abierta a Distancia 111511 Bogotá Colombia
| | - William Cheung
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University NE1 8ST Newcastle Upon Tyne UK
| | | |
Collapse
|
21
|
Wang S, Guo Q, Zhou L, Xia X. Ferroptosis: A double-edged sword. Cell Death Discov 2024; 10:265. [PMID: 38816377 PMCID: PMC11139933 DOI: 10.1038/s41420-024-02037-9] [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: 11/08/2023] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Ferroptosis represents a form of programmed cell death that is propelled by iron-dependent lipid peroxidation, thereby being distinguished by the prominent features of iron accumulation and lipid peroxidation. Ferroptosis has been implicated in numerous physiological and pathological phenomena, with mounting indications that it holds significant implications for cancer and other medical conditions. On one side, it demonstrates anti-cancer properties by triggering ferroptosis within malignant cells, and on the other hand, it damages normal cells causing other diseases. Therefore, in this paper, we propose to review the paradoxical regulation of ferroptosis in tumors and other diseases. First, we introduce the development history, concept and mechanism of ferroptosis. The second part focuses on the methods of inducing ferroptosis in tumors. The third section emphasizes the utilization of ferroptosis in different medical conditions and strategies to inhibit ferroptosis. The fourth part elucidates the key contradictions in the control of ferroptosis. Finally, potential research avenues in associated domains are suggested.
Collapse
Affiliation(s)
- Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
22
|
Zhao L, Wei Y, Liu Q, Cai J, Mo X, Tang X, Wang X, Qin L, Liang Y, Cao J, Huang C, Lu Y, Zhang T, Luo L, Rong J, Wu S, Jin W, Guan Q, Teng K, Li Y, Qin J, Zhang Z. Association between multiple-heavy-metal exposures and systemic immune inflammation in a middle-aged and elderly Chinese general population. BMC Public Health 2024; 24:1192. [PMID: 38679723 PMCID: PMC11057124 DOI: 10.1186/s12889-024-18638-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Exposure to heavy metals alone or in combination can promote systemic inflammation. The aim of this study was to investigate potential associations between multiple plasma heavy metals and markers of systemic immune inflammation. METHODS Using a cross-sectional study, routine blood tests were performed on 3355 participants in Guangxi, China. Eight heavy metal elements in plasma were determined by inductively coupled plasma mass spectrometry. Immunoinflammatory markers were calculated based on peripheral blood WBC and its subtype counts. A generalised linear regression model was used to analyse the association of each metal with the immunoinflammatory markers, and the association of the metal mixtures with the immunoinflammatory markers was further assessed using weighted quantile sum (WQS) regression. RESULTS In the single-metal model, plasma metal Fe (log10) was significantly negatively correlated with the levels of immune-inflammatory markers SII, NLR and PLR, and plasma metal Cu (log10) was significantly positively correlated with the levels of immune-inflammatory markers SII and PLR. In addition, plasma metal Mn (log10 conversion) was positively correlated with the levels of immune inflammatory markers NLR and PLR. The above associations remained after multiple corrections. In the mixed-metal model, after WQS regression analysis, plasma metal Cu was found to have the greatest weight in the positive effects of metal mixtures on SII and PLR, while plasma metals Mn and Fe had the greatest weight in the positive effects of metal mixtures on NLR and LMR, respectively. In addition, blood Fe had the greatest weight in the negative effects of the metal mixtures for SII, PLR and NLR. CONCLUSION Plasma metals Cu and Mn were positively correlated with immunoinflammatory markers SII, NLR and PLR. While plasma metal Fe was negatively correlated with immunoinflammatory markers SII, NLR, and PLR.
Collapse
Affiliation(s)
- Linhai Zhao
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yanfei Wei
- Department of Epidemiology, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, Guangdong, China
| | - Qiumei Liu
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiansheng Cai
- School of Public Health, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Entire Lifecycle Health and Care, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Xiaoting Mo
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xu Tang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xuexiu Wang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Lidong Qin
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yujian Liang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiejing Cao
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Chuwu Huang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yufu Lu
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Tiantian Zhang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Lei Luo
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiahui Rong
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Songju Wu
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Wenjia Jin
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Qinyi Guan
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Kaisheng Teng
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - You Li
- School of Public Health, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Jian Qin
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Zhiyong Zhang
- Department of Occupational and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
- School of Public Health, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China.
- Guangxi Key Laboratory of Entire Lifecycle Health and Care, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
23
|
Alqahtani SAM, Alsaleem MA, Ghazy RM. Association between serum ferritin level and lipid profile among diabetic patients: A retrospective cohort study. Medicine (Baltimore) 2024; 103:e37631. [PMID: 38552070 PMCID: PMC10977537 DOI: 10.1097/md.0000000000037631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/26/2024] [Indexed: 04/02/2024] Open
Abstract
High serum ferritin (SF) levels have been linked to obesity, metabolic syndrome, atherosclerosis, diabetes, dyslipidemia, and cancer. This study aimed to investigate the association between SF and dyslipidemia in adults diagnosed with diabetes mellitus. This cross-sectional study retrospectively analyzed the electronic medical records of eligible patients from 3 primary locations in Saudi Arabia namely - Abha, Khamis Mushyt, and Jeddah - from 2010 to 2020. The study included adult patients aged 18 years or older who were diagnosed with diabetes mellitus and identified with an HbA1c level of ≥6.5. This study involved 3674 participants, with males accounting for 26.6% of the total. The mean age of the studied population was 48.0 ± 18.4 years. The median [interquartile range] of SF among males was higher than females, however, this difference was not statistically significant (60.0 [23.4-125.8] vs 55.4 [24.0-113.4], P = 0.204). On the other hand, age and region were significantly associated with SF (P = .032 and 0.035). SF had a significant positive correlation with cholesterol (r = 0.081, P < .001), low-density lipoprotein cholesterol (r = .087, P < .001), and triglycerides (r = 0.068, P < .001) and negative correlation with high-density lipoprotein cholesterol (r = -0.13, P < .001). Multivariate analysis revealed that age, sex, residence, and HbA1c were significantly affecting the lipid profile. Clinicians should consider including SF testing as part of the comprehensive evaluation of patients with diabetes and dyslipidemia.
Collapse
Affiliation(s)
- Saif Aboud M Alqahtani
- Internal Medicine Department, College of Medicine, King Khalid University, Abha, Saudia Arabia
| | - Mohammed Abadi Alsaleem
- Department of Family and Community Medicine, College of Medicine, King Khalid University, Abha, Saudia Arabia
| | - Ramy Mohamed Ghazy
- Department of Family and Community Medicine, College of Medicine, King Khalid University, Abha, Saudia Arabia
- Tropical Health Department, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| |
Collapse
|
24
|
Ceramella J, De Maio AC, Basile G, Facente A, Scali E, Andreu I, Sinicropi MS, Iacopetta D, Catalano A. Phytochemicals Involved in Mitigating Silent Toxicity Induced by Heavy Metals. Foods 2024; 13:978. [PMID: 38611284 PMCID: PMC11012104 DOI: 10.3390/foods13070978] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/30/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
Heavy metals (HMs) are natural elements present in the Earth's crust, characterised by a high atomic mass and a density more than five times higher than water. Despite their origin from natural sources, extensive usage and processing of raw materials and their presence as silent poisons in our daily products and diets have drastically altered their biochemical balance, making them a threat to the environment and human health. Particularly, the food chain polluted with toxic metals represents a crucial route of human exposure. Therefore, the impact of HMs on human health has become a matter of concern because of the severe chronic effects induced by their excessive levels in the human body. Chelation therapy is an approved valid treatment for HM poisoning; however, despite the efficacy demonstrated by chelating agents, various dramatic side effects may occur. Numerous data demonstrate that dietary components and phytoantioxidants play a significant role in preventing or reducing the damage induced by HMs. This review summarises the role of various phytochemicals, plant and herbal extracts or probiotics in promoting human health by mitigating the toxic effects of different HMs.
Collapse
Affiliation(s)
- Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (J.C.); (A.C.D.M.); (G.B.); (A.F.); (D.I.)
| | - Azzurra Chiara De Maio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (J.C.); (A.C.D.M.); (G.B.); (A.F.); (D.I.)
| | - Giovanna Basile
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (J.C.); (A.C.D.M.); (G.B.); (A.F.); (D.I.)
| | - Anastasia Facente
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (J.C.); (A.C.D.M.); (G.B.); (A.F.); (D.I.)
| | - Elisabetta Scali
- Unit of Dermatology, Spoke Hospital, Locri, 89044 Reggio Calabria, Italy;
| | - Inmaculada Andreu
- Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
- Unidad Mixta de Investigación UPV-IIS La Fe, Hospital Universitari i Politècnic La Fe, Avenida de Fernando, Abril Martorell 106, 46026 Valencia, Spain
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (J.C.); (A.C.D.M.); (G.B.); (A.F.); (D.I.)
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (J.C.); (A.C.D.M.); (G.B.); (A.F.); (D.I.)
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy;
| |
Collapse
|
25
|
Zhang Y, Wang H, Jia R, Chen D, Li Z. Serum ferritin is associated with the presence of ischemic stroke among individuals with type 2 diabetes. Heliyon 2024; 10:e27898. [PMID: 38486737 PMCID: PMC10938112 DOI: 10.1016/j.heliyon.2024.e27898] [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: 09/06/2023] [Revised: 03/03/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024] Open
Abstract
Background Epidemiological evidence regarding the possible link between serum ferritin (SF) level and ischemic stroke risk among individuals with type 2 diabetes mellitus (T2DM) is sparse. Aim To evaluate the association between SF level in plasma and ischemic stroke risk among individuals with T2DM. Methods SF levels were measured in 210 T2DM patients with (n = 165) or without ischemic stroke (n = 45). Multivariate logistic regression analyses were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). Results The SF level of T2DM patients with ischemic stroke was significantly higher than that of patients without ischemic stroke (P = 0.003). The multivariate logistic regression analyses revealed that each 1-SD increase in SF (OR: 1.92; 95%CI: 1.22, 3.03) was significantly associated with increased ischemic stroke risk among T2DM patients. In addition, interaction effect of SF and BMI on ischemic stroke risk were also observed (Pfor interaction = 0.037). Conclusions Higher levels of SF were independently associated with increased risk of ischemic stroke among individuals with T2DM.
Collapse
Affiliation(s)
- Youyou Zhang
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xi'wu Road, Xi'an, 710004, Shaanxi, China
| | - Hui Wang
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xi'wu Road, Xi'an, 710004, Shaanxi, China
| | - Ruirui Jia
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xi'wu Road, Xi'an, 710004, Shaanxi, China
| | - Dong Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xi'wu Road, Xi'an, 710004, Shaanxi, China
| | - Zhaoyang Li
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University, No. 76 West Yanta Road, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
26
|
Kamel AA, Nassar AY, Meligy FY, Omar YA, Nassar GAY, Ezzat GM. Acetylated oligopeptide and N-acetylcysteine protect against iron overload-induced dentate gyrus hippocampal degeneration through upregulation of Nestin and Nrf2/HO-1 and downregulation of MMP-9/TIMP-1 and GFAP. Cell Biochem Funct 2024; 42:e3958. [PMID: 38396357 DOI: 10.1002/cbf.3958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/29/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
Iron accumulation in the brain causes oxidative stress, blood-brain barrier (BBB) breakdown, and neurodegeneration. We examined the preventive effects of acetylated oligopeptides (AOP) from whey protein on iron-induced hippocampal damage compared to N-acetyl cysteine (NAC). This 5-week study used 40 male albino rats. At the start, all rats received 150 mg/kg/day of oral NAC for a week. The 40 animals were then randomly divided into four groups: Group I (control) received a normal diet; Group II (iron overload) received 60 mg/kg/day intraperitoneal iron dextran 5 days a week for 4 weeks; Group III (NAC group) received 150 mg/kg/day NAC and iron dextran; and Group IV (AOP group) received 150 mg/kg/day AOP and iron dextran. Enzyme-linked immunosorbent assay, spectrophotometry, and qRT-PCR were used to measure MMP-9, tissue inhibitor metalloproteinase-1 (TIMP-1), MDA, reduced glutathione (GSH) levels, and nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) gene expression. Histopathological and immunohistochemical detection of nestin, claudin, caspase, and GFAP was also done. MMP-9, TIMP-1, MDA, caspase, and GFAP rose in the iron overload group, while GSH, Nrf2, HO-1, nestin, and claudin decreased. The NAC and AOP administrations improved iron overload-induced biochemical and histological alterations. We found that AOP and NAC can protect the brain hippocampus from iron overload, improve BBB disruption, and provide neuroprotection with mostly no significant difference from healthy controls.
Collapse
Affiliation(s)
- Amira A Kamel
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Y Nassar
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Fatma Y Meligy
- Department of Restorative Dentistry and Basic Medical Sciences, Faculty of Dentistry, University of Petra, Amman, Jordan
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Yomna A Omar
- Department of Biochemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Gamal A Y Nassar
- Metabolic and Genetic Disorders Unit, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ghada M Ezzat
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
27
|
Simon Machado R, Mathias K, Joaquim L, Willig de Quadros R, Petronilho F, Tezza Rezin G. From diabetic hyperglycemia to cerebrovascular Damage: A narrative review. Brain Res 2023; 1821:148611. [PMID: 37793604 DOI: 10.1016/j.brainres.2023.148611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/04/2023] [Accepted: 09/28/2023] [Indexed: 10/06/2023]
Abstract
Diabetes mellitus is a globally significant disease that can lead to systemic complications, particularly vascular damage, including cardiovascular and cerebrovascular diseases of relevance. The physiological changes resulting from the imbalance in blood glucose levels play a crucial role in initiating vascular endothelial damage. Elevated glucose levels can also penetrate the central nervous system, triggering diabetic encephalopathy characterized by oxidative damage to brain components and activation of alternative and neurotoxic pathways. This brain damage increases the risk of ischemic stroke, a leading cause of mortality worldwide and a major cause of disability among surviving patients. The aim of this review is to highlight important pathways related to hyperglycemic damage that extend to the brain and result in vascular dysfunction, ultimately leading to the occurrence of a stroke. Understanding how diabetes mellitus contributes to the development of ischemic stroke and its impact on patient outcomes is crucial for implementing therapeutic strategies that reduce the incidence of diabetes mellitus and its complications, ultimately decreasing morbidity and mortality associated with the disease.
Collapse
Affiliation(s)
- Richard Simon Machado
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil; Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil.
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Larissa Joaquim
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Rafaella Willig de Quadros
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| |
Collapse
|
28
|
Shen R, Ardianto C, Celia C, Sidharta VM, Sasmita PK, Satriotomo I, Turana Y. Brain-derived neurotrophic factor interplay with oxidative stress: neuropathology approach in potential biomarker of Alzheimer's disease. Dement Neuropsychol 2023; 17:e20230012. [PMID: 38053647 PMCID: PMC10695442 DOI: 10.1590/1980-5764-dn-2023-0012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/05/2023] [Accepted: 07/30/2023] [Indexed: 12/07/2023] Open
Abstract
The aging population poses a serious challenge concerning an increased prevalence of Alzheimer's disease (AD) and its impact on global burden, morbidity, and mortality. Oxidative stress, as a molecular hallmark that causes susceptibility in AD, interplays to other AD-related neuropathology cascades and decreases the expression of central and circulation brain-derived neurotrophic factor (BDNF), an essential neurotrophin that serves as nerve development and survival, and synaptic plasticity in AD. By its significant correlation with the molecular and clinical progression of AD, BDNF can potentially be used as an objectively accurate biomarker for AD diagnosis and progressivity follow-up in future clinical practice. This comprehensive review highlights the oxidative stress interplay with BDNF in AD neuropathology and its potential use as an AD biomarker.
Collapse
Affiliation(s)
- Robert Shen
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Christian Ardianto
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Celia Celia
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Veronika Maria Sidharta
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Poppy Kristina Sasmita
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| | - Irawan Satriotomo
- University of Florida, Gainesville, Department of Neurology, Florida, USA
- Satriotomo Foundation, Indonesia Neuroscience Institute, Jakarta, Indonesia
| | - Yuda Turana
- Atma Jaya Catholic University of Indonesia, School of Medicine and Health Sciences, Jakarta, Indonesia
| |
Collapse
|
29
|
Jin J, Han W, Yang T, Xu Z, Zhang J, Cao R, Wang Y, Wang J, Hu X, Gu T, He F, Huang J, Li G. Artificial light at night, MRI-based measures of brain iron deposition and incidence of multiple mental disorders. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 902:166004. [PMID: 37544462 DOI: 10.1016/j.scitotenv.2023.166004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/21/2023] [Accepted: 08/01/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Epidemiologic evidence on whether iron accumulation in brain modified the association between artificial light at night (ALAN) and incident mental disorders is lacking. The authors aims to investigate modification of brain iron deposition on the associations of ALAN with multiple mental disorders in the middle-aged and older adults. METHODS This prospective study used data from the UK Biobank. ALAN was drawn from satellite datasets. Susceptibility-weighted magnetic resonance imaging was used to ascertain iron content of each brain region. T2* signal loss was used as indices of iron deposition. The main outcomes are impacts of ALAN exposure on onset of wide spectrum of physician-diagnosed mental disorders, which was estimated by time-varying Cox proportional hazard model. The authors further conducted stratified analyses by levels of iron brain deposition to examine the potential modifying effects. RESULTS Among 298,283 participants followed for a median of 10.91 years, higher ALAN exposure was associated with increased risk of mental disorders. An IQR (11.37 nW/cm2/sr) increase in annual levels of ALAN was associated with an HR of 1.050 (95 % CI: 1.034,1.066) for any mental disorder, 1.076 (95 % CI: 1.053,1.099) for substance use disorder, and 1.036 (95 % CI: 1.004,1.069) for depression disorder in fully adjusted models. The exposure-response curves showed steeper trends at lower ALAN levels and a plateau at higher exposures. The associations were stronger in participants with high iron deposition in left hippocampus, left accumbens and left pallidum. CONCLUSIONS ALAN was associated with multiple mental disorders in the middle-aged and older adults, and the findings indicated stricter standards of ALAN is needed and targeted preventive measures are warranted, especially with high brain iron deposition.
Collapse
Affiliation(s)
- Jianbo Jin
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Wenxing Han
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Teng Yang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Zhihu Xu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Jin Zhang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Ru Cao
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Yuxin Wang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Jiawei Wang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Xin Hu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Tiantian Gu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China.
| | - Fan He
- Beijing Anding Hospital, Capital Medical University, Beijing, China.
| | - Jing Huang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China; Peking University Institute for Global Health and Development, Beijing, China.
| | - Guoxing Li
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, Beijing, China; Environmental Research Group, School of Public Health, Imperial college London, London, UK.
| |
Collapse
|
30
|
Genchi G, Lauria G, Catalano A, Carocci A, Sinicropi MS. Prevalence of Cobalt in the Environment and Its Role in Biological Processes. BIOLOGY 2023; 12:1335. [PMID: 37887045 PMCID: PMC10604320 DOI: 10.3390/biology12101335] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/08/2023] [Accepted: 10/14/2023] [Indexed: 10/28/2023]
Abstract
Cobalt (Co) is an essential trace element for humans and other animals, but high doses can be harmful to human health. It is present in some foods such as green vegetables, various spices, meat, milk products, seafood, and eggs, and in drinking water. Co is necessary for the metabolism of human beings and animals due to its key role in the formation of vitamin B12, also known as cobalamin, the biological reservoir of Co. In high concentrations, Co may cause some health issues such as vomiting, nausea, diarrhea, bleeding, low blood pressure, heart diseases, thyroid damage, hair loss, bone defects, and the inhibition of some enzyme activities. Conversely, Co deficiency can lead to anorexia, chronic swelling, and detrimental anemia. Co nanoparticles have different and various biomedical applications thanks to their antioxidant, antimicrobial, anticancer, and antidiabetic properties. In addition, Co and cobalt oxide nanoparticles can be used in lithium-ion batteries, as a catalyst, a carrier for targeted drug delivery, a gas sensor, an electronic thin film, and in energy storage. Accumulation of Co in agriculture and humans, due to natural and anthropogenic factors, represents a global problem affecting water quality and human and animal health. Besides the common chelating agents used for Co intoxication, phytoremediation is an interesting environmental technology for cleaning up soil contaminated with Co. The occurrence of Co in the environment is discussed and its involvement in biological processes is underlined. Toxicological aspects related to Co are also examined in this review.
Collapse
Affiliation(s)
- Giuseppe Genchi
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Arcavacata di Rende, 87036 Cosenza, Italy; (G.G.); (G.L.); (M.S.S.)
| | - Graziantonio Lauria
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Arcavacata di Rende, 87036 Cosenza, Italy; (G.G.); (G.L.); (M.S.S.)
| | - Alessia Catalano
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “A. Moro”, 70125 Bari, Italy;
| | - Alessia Carocci
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “A. Moro”, 70125 Bari, Italy;
| | - Maria Stefania Sinicropi
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Arcavacata di Rende, 87036 Cosenza, Italy; (G.G.); (G.L.); (M.S.S.)
| |
Collapse
|
31
|
Wang Q, Sun J, Chen T, Song S, Hou Y, Feng L, Fan C, Li M. Ferroptosis, Pyroptosis, and Cuproptosis in Alzheimer's Disease. ACS Chem Neurosci 2023; 14:3564-3587. [PMID: 37703318 DOI: 10.1021/acschemneuro.3c00343] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
Alzheimer's disease (AD), the most common type of dementia, is a neurodegenerative disorder characterized by progressive cognitive dysfunction. Epidemiological investigation has demonstrated that, after cardiovascular and cerebrovascular diseases, tumors, and other causes, AD has become a major health issue affecting elderly individuals, with its mortality rate acutely increasing each year. Regulatory cell death is the active and orderly death of genetically determined cells, which is ubiquitous in the development of living organisms and is crucial to the regulation of life homeostasis. With extensive research on regulatory cell death in AD, increasing evidence has revealed that ferroptosis, pyroptosis, and cuproptosis are closely related to the occurrence, development, and prognosis of AD. This paper will review the molecular mechanisms of ferroptosis, pyroptosis, and cuproptosis and their regulatory roles in AD to explore potential therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Qi Wang
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Jingyi Sun
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Tian Chen
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Siyu Song
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Yajun Hou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Lina Feng
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Cundong Fan
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Mingquan Li
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| |
Collapse
|
32
|
Tian HY, Huang BY, Nie HF, Chen XY, Zhou Y, Yang T, Cheng SW, Mei ZG, Ge JW. The Interplay between Mitochondrial Dysfunction and Ferroptosis during Ischemia-Associated Central Nervous System Diseases. Brain Sci 2023; 13:1367. [PMID: 37891735 PMCID: PMC10605666 DOI: 10.3390/brainsci13101367] [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: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Cerebral ischemia, a leading cause of disability and mortality worldwide, triggers a cascade of molecular and cellular pathologies linked to several central nervous system (CNS) disorders. These disorders primarily encompass ischemic stroke, Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and other CNS conditions. Despite substantial progress in understanding and treating the underlying pathological processes in various neurological diseases, there is still a notable absence of effective therapeutic approaches aimed specifically at mitigating the damage caused by these illnesses. Remarkably, ischemia causes severe damage to cells in ischemia-associated CNS diseases. Cerebral ischemia initiates oxygen and glucose deprivation, which subsequently promotes mitochondrial dysfunction, including mitochondrial permeability transition pore (MPTP) opening, mitophagy dysfunction, and excessive mitochondrial fission, triggering various forms of cell death such as autophagy, apoptosis, as well as ferroptosis. Ferroptosis, a novel type of regulated cell death (RCD), is characterized by iron-dependent accumulation of lethal reactive oxygen species (ROS) and lipid peroxidation. Mitochondrial dysfunction and ferroptosis both play critical roles in the pathogenic progression of ischemia-associated CNS diseases. In recent years, growing evidence has indicated that mitochondrial dysfunction interplays with ferroptosis to aggravate cerebral ischemia injury. However, the potential connections between mitochondrial dysfunction and ferroptosis in cerebral ischemia have not yet been clarified. Thus, we analyzed the underlying mechanism between mitochondrial dysfunction and ferroptosis in ischemia-associated CNS diseases. We also discovered that GSH depletion and GPX4 inactivation cause lipoxygenase activation and calcium influx following cerebral ischemia injury, resulting in MPTP opening and mitochondrial dysfunction. Additionally, dysfunction in mitochondrial electron transport and an imbalanced fusion-to-fission ratio can lead to the accumulation of ROS and iron overload, which further contribute to the occurrence of ferroptosis. This creates a vicious cycle that continuously worsens cerebral ischemia injury. In this study, our focus is on exploring the interplay between mitochondrial dysfunction and ferroptosis, which may offer new insights into potential therapeutic approaches for the treatment of ischemia-associated CNS diseases.
Collapse
Affiliation(s)
- He-Yan Tian
- School of Medical Technology and Nursing, Shenzhen Polytechnic University, Xili Lake, Nanshan District, Shenzhen 518000, China;
| | - Bo-Yang Huang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Hui-Fang Nie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xiang-Yu Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shao-Wu Cheng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhi-Gang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jin-Wen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
- Hunan Academy of Traditional Chinese Medicine, Changsha 410208, China
| |
Collapse
|
33
|
Mukem S, Sayoh I, Maungchanburi S, Thongbuakaew T. Ebselen, Iron Uptake Inhibitor, Alleviates Iron Overload-Induced Senescence-Like Neuronal Cells SH-SY5Y via Suppressing the mTORC1 Signaling Pathway. Adv Pharmacol Pharm Sci 2023; 2023:6641347. [PMID: 37731679 PMCID: PMC10509000 DOI: 10.1155/2023/6641347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/17/2023] [Accepted: 09/02/2023] [Indexed: 09/22/2023] Open
Abstract
Increasing evidence highlights that excessive iron accumulation in the brain plays a vital role in neuronal senescence and is implicated in the pathogenesis of age-related neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Therefore, the chemical compounds that eliminate an iron overload may provide better protection against oxidative stress conditions that cause the accumulation of senescent cells during brain aging. Ebselen has been identified as a strongly useful compound in the research on redox biology mechanisms. We hypothesized that ebselen could alleviate an iron overload-induced oxidative stress and consequently reverses the senescence-like phenotypes in the neuronal cells. In the present study, SH-SY5Y cells were treated with ferric ammonium citrate (FAC) before ebselen, and the evaluation of the cellular iron homeostasis, the indicators of oxidative stress, and the onset of senescence phenotypes and mechanisms were carried out accordingly. Our findings showed that ebselen ameliorated the FAC-mediated iron overload by decreasing the expression of divalent metal transporter 1 (DMT1) and ferritin light chain (FT-L) proteins. In contrast, it increased the expression of ferroportin 1 (FPN1) protein and its correlation led to a decrease in the expression of the cytosolic labile iron pool (LIP). Furthermore, ebselen significantly reduced reactive oxygen species (ROS) and rescued the mitochondrial membrane potential (ΔΨm). Notably, ebselen restored the biomarkers of cellular senescence by reducing the number of senescence-associated β-galactosidase (SA-β-gal) positive cells and senescence-associated secretory phenotypes (SASP). This also suppressed the expression of p53 protein targeting DNA damage response (DDR)/p21 cyclin-dependent kinase (CDK) inhibitor through a mTORC1 signaling pathway. Potentially, ebselen could be a therapeutic agent for treating brain aging and AD by mitigating iron accumulation and restoring senescence in SH-SY5Y cells.
Collapse
Affiliation(s)
- Sirirak Mukem
- School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Ibrahim Sayoh
- Department of Anatomy, Faculty of Science and Technology, Princess of Naradhiwas University, Narathiwat 96000, Thailand
| | - Saowanee Maungchanburi
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | | |
Collapse
|
34
|
Martins AC, Virgolini MB, Ávila DS, Scharf P, Li J, Tinkov AA, Skalny AV, Bowman AB, Rocha JBT, Aschner M. Mitochondria in the Spotlight: C. elegans as a Model Organism to Evaluate Xenobiotic-Induced Dysfunction. Cells 2023; 12:2124. [PMID: 37681856 PMCID: PMC10486742 DOI: 10.3390/cells12172124] [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: 07/18/2023] [Revised: 08/19/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023] Open
Abstract
Mitochondria play a crucial role in cellular respiration, ATP production, and the regulation of various cellular processes. Mitochondrial dysfunctions have been directly linked to pathophysiological conditions, making them a significant target of interest in toxicological research. In recent years, there has been a growing need to understand the intricate effects of xenobiotics on human health, necessitating the use of effective scientific research tools. Caenorhabditis elegans (C. elegans), a nonpathogenic nematode, has emerged as a powerful tool for investigating toxic mechanisms and mitochondrial dysfunction. With remarkable genetic homology to mammals, C. elegans has been used in studies to elucidate the impact of contaminants and drugs on mitochondrial function. This review focuses on the effects of several toxic metals and metalloids, drugs of abuse and pesticides on mitochondria, highlighting the utility of C. elegans as a model organism to investigate mitochondrial dysfunction induced by xenobiotics. Mitochondrial structure, function, and dynamics are discussed, emphasizing their essential role in cellular viability and the regulation of processes such as autophagy, apoptosis, and calcium homeostasis. Additionally, specific toxins and toxicants, such as arsenic, cadmium, and manganese are examined in the context of their impact on mitochondrial function and the utility of C. elegans in elucidating the underlying mechanisms. Furthermore, we demonstrate the utilization of C. elegans as an experimental model providing a promising platform for investigating the intricate relationships between xenobiotics and mitochondrial dysfunction. This knowledge could contribute to the development of strategies to mitigate the adverse effects of contaminants and drugs of abuse, ultimately enhancing our understanding of these complex processes and promoting human health.
Collapse
Affiliation(s)
- Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Miriam B. Virgolini
- Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Instituto de Farmacología Experimental de Córdoba-Consejo Nacional de Investigaciones Técnicas (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Daiana Silva Ávila
- Laboratory of Biochemistry and Toxicology in Caenorhabditis Elegans, Universidade Federal do Pampa, Campus Uruguaiana, BR-472 Km 592, Uruguaiana 97500-970, RS, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Jung Li
- College of Osteopathic Medicine, Des Moines University, Des Moines, IA 50312, USA
| | - Alexey A. Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Anatoly V. Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Peoples Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - João B. T. Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| |
Collapse
|
35
|
Wang C, Xie L, Xing Y, Liu M, Yang J, Gao N, Cai Y. Iron-overload-induced ferroptosis in mouse cerebral toxoplasmosis promotes brain injury and could be inhibited by Deferiprone. PLoS Negl Trop Dis 2023; 17:e0011607. [PMID: 37651502 PMCID: PMC10508604 DOI: 10.1371/journal.pntd.0011607] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/19/2023] [Accepted: 08/20/2023] [Indexed: 09/02/2023] Open
Abstract
Iron is a trace metal element that is essential for the survival of cells and parasites. The role of iron in cerebral toxoplasmosis (CT) is still unclear. Deferiprone (DFP) is the orally active iron chelator that binds iron in a molar ratio of 3:1 (ligand:iron) and promotes urinary iron excretion to remove excess iron from the body. The aims of this experiment were to observe the alterations in iron in brains with Toxoplasma gondii (T. gondii) acute infections and to investigate the mechanism of ferroptosis in CT using DFP. We established a cerebral toxoplasmosis model in vivo using TgCtwh3, the dominant strains of which are prevalent in China, and treated the mice with DFP at a dose of 75 mg/kg/d. Meanwhile, we treated the HT-22 cells with 100 μM DFP for half an hour and then infected cells with TgCtwh3 in vitro. A qRT-PCR assay of TgSAG1 levels showed a response to the T. gondii burden. We used inductively coupled plasma mass spectrometry, an iron ion assay kit, Western blot analysis, glutathione and glutathione disulfide assay kits, a malonaldehyde assay kit, and immunofluorescence to detect the ferroptosis-related indexes in the mouse hippocampus and HT-22 cells. The inflammatory factors interferon-γ, tumor necrosis factor-α, transforming growth factor-β, and arginase 1 in the hippocampus and cells were detected using the Western blot assay. Hematoxylin and eosin staining, electron microscopy, and the Morris water maze experiment were used to evaluate the brain injuries of the mice. The results showed that TgCtwh3 infection is followed by the activation of ferroptosis-related signaling pathways and hippocampal pathological damage in mice. The use of DFP led to ferroptosis resistance and attenuated pathological changes, inflammatory reactions and T. gondii burden of the mice, prolonging their survival time. The HT-22 cells with TgCtwh3 activated the ferroptosis pathway and was inhibit by DFP in vitro. In TgCtwh3-infected cells, inflammatory response and mitochondrial damage were severe, but these effects could be reduced by DFP. Our study elucidates the mechanism by which T. gondii interferes with the host's iron metabolism and activates ferroptosis, complementing the pathogenic mechanism of CT and further demonstrating the potential value of DFP for the treatment of CT.
Collapse
Affiliation(s)
- Chong Wang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Microbiology and Parasitology, and Anhui Provincial Laboratory of Zoonoses of High Institutions, Anhui Medical University, Hefei, China
| | - Linding Xie
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Microbiology and Parasitology, and Anhui Provincial Laboratory of Zoonoses of High Institutions, Anhui Medical University, Hefei, China
| | - Yien Xing
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Microbiology and Parasitology, and Anhui Provincial Laboratory of Zoonoses of High Institutions, Anhui Medical University, Hefei, China
| | - Min Liu
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Microbiology and Parasitology, and Anhui Provincial Laboratory of Zoonoses of High Institutions, Anhui Medical University, Hefei, China
| | - Jun Yang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Microbiology and Parasitology, and Anhui Provincial Laboratory of Zoonoses of High Institutions, Anhui Medical University, Hefei, China
| | - Nannan Gao
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Microbiology and Parasitology, and Anhui Provincial Laboratory of Zoonoses of High Institutions, Anhui Medical University, Hefei, China
| | - Yihong Cai
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, China
- Anhui Provincial Laboratory of Microbiology and Parasitology, and Anhui Provincial Laboratory of Zoonoses of High Institutions, Anhui Medical University, Hefei, China
| |
Collapse
|
36
|
Lee S, Hwang N, Seok BG, Lee S, Lee SJ, Chung SW. Autophagy mediates an amplification loop during ferroptosis. Cell Death Dis 2023; 14:464. [PMID: 37491375 PMCID: PMC10368698 DOI: 10.1038/s41419-023-05978-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/09/2023] [Accepted: 07/11/2023] [Indexed: 07/27/2023]
Abstract
Ferroptosis, a programmed cell death, has been identified and associated with cancer and various other diseases. Ferroptosis is defined as a reactive oxygen species (ROS)-dependent cell death related to iron accumulation and lipid peroxidation, which is different from apoptosis, necrosis, autophagy, and other forms of cell death. However, accumulating evidence has revealed a link between autophagy and ferroptosis at the molecular level and has suggested that autophagy is involved in regulating the accumulation of iron-dependent lipid peroxidation and ROS during ferroptosis. Understanding the roles and pathophysiological processes of autophagy during ferroptosis may provide effective strategies for the treatment of ferroptosis-related diseases. In this review, we summarize the current knowledge regarding the regulatory mechanisms underlying ferroptosis, including iron and lipid metabolism, and its association with the autophagy pathway. In addition, we discuss the contribution of autophagy to ferroptosis and elucidate the role of autophagy as a ferroptosis enhancer during ROS-dependent ferroptosis.
Collapse
Affiliation(s)
- Seunghee Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, VA Palo Alto Health Care System and Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Narae Hwang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Byeong Geun Seok
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea
| | - Sangguk Lee
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea
| | - Seon-Jin Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon, 34141, South Korea
| | - Su Wol Chung
- Department of Biological Sciences, College of Natural Sciences, University of Ulsan, 93 Daehak-ro, Nam-gu, Ulsan, 44610, South Korea.
- Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan, 44610, South Korea.
| |
Collapse
|
37
|
Guo L, Hu C, Yao M, Han G. Mechanism of sorafenib resistance associated with ferroptosis in HCC. Front Pharmacol 2023; 14:1207496. [PMID: 37351514 PMCID: PMC10282186 DOI: 10.3389/fphar.2023.1207496] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/26/2023] [Indexed: 06/24/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most familiar primary hepatic malignancy with a poor prognosis. The incidence of HCC and the associated deaths have risen in recent decades. Sorafenib is the first drug to be approved by the Food and Drug Administration (FDA) for routine use in the first-line therapy of patients with advanced HCC. However, only about 30% of patients with HCC will be benefited from sorafenib therapy, and drug resistance typically develops within 6 months. In recent years, the mechanisms of resistance to sorafenib have gained the attention of a growing number of researchers. A promising field of current studies is ferroptosis, which is a novel form of cell death differing from apoptosis, necroptosis, and autophagy. This process is dependent on the accumulation of intracellular iron and reactive oxygen species (ROS). Furthermore, the increase in intracellular iron levels and ROS can be significantly observed in cells resistant to sorafenib. This article reviews the mechanisms of resistance to sorafenib that are related to ferroptosis, evaluates the relationship between ferroptosis and sorafenib resistance, and explores new therapeutic approaches capable of reversing sorafenib resistance in HCC through the modulation of ferroptosis.
Collapse
|
38
|
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: 4.5] [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
|
39
|
Langenbacher RE, Horoszko CP, Kim M, Heller DA. Hematoxylin Nuclear Stain Reports Oxidative Stress via Near-Infrared Emission. ACS Chem Biol 2023; 18:1237-1245. [PMID: 37070948 PMCID: PMC10289833 DOI: 10.1021/acschembio.3c00156] [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: 04/19/2023]
Abstract
Hematoxylin & eosin (H&E) is the gold standard histological stain used for medical diagnosis and has been used for over a century. Herein, we examined the near-infrared II (NIR-II) fluorescence of this stain. We observed significant NIR-II emission from the hematoxylin component of the H&E stain. We found that the emission intensity, using the common aluminum(III) hematoxylin mordant, could be modulated by the availability of endogenous iron(III), and this emission intensity increased at higher oxidative stress. Our mechanistic investigations found that hematoxylin emission reported the nuclear translocation of the iron via the protein ferritin. In human tumor tissue samples, oxidative stress biomarkers correlated with hematoxylin NIR-II emission intensity. Emission response of the stain was also observed in human Alzheimer's disease brain tissue regions affected by disease progression, suggesting that ferritin nuclear translocation is preserved in these regions as an oxidative stress response. These findings indicate that NIR-II emission from the H&E stain provides a new source of redox information in tissues with implications for biomedical research and clinical practice.
Collapse
Affiliation(s)
| | | | - Mijin Kim
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Daniel A. Heller
- Weill Cornell Medicine, Cornell University, New York, NY 10065, United States
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| |
Collapse
|
40
|
Lotan A, Luza S, Opazo CM, Ayton S, Lane DJR, Mancuso S, Pereira A, Sundram S, Weickert CS, Bousman C, Pantelis C, Everall IP, Bush AI. Perturbed iron biology in the prefrontal cortex of people with schizophrenia. Mol Psychiatry 2023; 28:2058-2070. [PMID: 36750734 PMCID: PMC10575779 DOI: 10.1038/s41380-023-01979-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
Despite loss of grey matter volume and emergence of distinct cognitive deficits in young adults diagnosed with schizophrenia, current treatments for schizophrenia do not target disruptions in late maturational reshaping of the prefrontal cortex. Iron, the most abundant transition metal in the brain, is essential to brain development and function, but in excess, it can impair major neurotransmission systems and lead to lipid peroxidation, neuroinflammation and accelerated aging. However, analysis of cortical iron biology in schizophrenia has not been reported in modern literature. Using a combination of inductively coupled plasma-mass spectrometry and western blots, we quantified iron and its major-storage protein, ferritin, in post-mortem prefrontal cortex specimens obtained from three independent, well-characterised brain tissue resources. Compared to matched controls (n = 85), among schizophrenia cases (n = 86) we found elevated tissue iron, unlikely to be confounded by demographic and lifestyle variables, by duration, dose and type of antipsychotic medications used or by copper and zinc levels. We further observed a loss of physiologic age-dependent iron accumulation among people with schizophrenia, in that the iron level among cases was already high in young adulthood. Ferritin, which stores iron in a redox-inactive form, was paradoxically decreased in individuals with the disorder. Such iron-ferritin uncoupling could alter free, chemically reactive, tissue iron in key reasoning and planning areas of the young-adult schizophrenia cortex. Using a prediction model based on iron and ferritin, our data provide a pathophysiologic link between perturbed cortical iron biology and schizophrenia and indicate that achievement of optimal cortical iron homeostasis could offer a new therapeutic target.
Collapse
Affiliation(s)
- Amit Lotan
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Psychiatry and the Biological Psychiatry Laboratory, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sandra Luza
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia.
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Serafino Mancuso
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Avril Pereira
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
- Mental Health Program, Monash Health, Melbourne, VIC, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Chad Bousman
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
| | - Ian P Everall
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia.
| |
Collapse
|
41
|
Li G, Yu W, Yang H, Wang X, Ma T, Luo X. Relationship between Serum Ferritin Level and Dyslipidemia in US Adults Based on Data from the National Health and Nutrition Examination Surveys 2017 to 2020. Nutrients 2023; 15:1878. [PMID: 37111096 PMCID: PMC10143246 DOI: 10.3390/nu15081878] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Previous research has suggested that high serum ferritin (SF) levels may be associated with dyslipidemia. This study investigated the association between SF levels and dyslipidemia in American adults, which held relevance for both clinical and public health areas concerned with screening and prevention. Data from the pre-pandemic National Health and Nutrition Examination Surveys (NHANES), conducted between 2017 and 2020, were utilized for this analysis. Multivariate linear regression models were used to explore the correlation between lipid and SF concentrations, and the connection between SF and the four types of dyslipidemia was further assessed by using multivariate logistic regression analysis. Odds ratios (ORs; 95% CI) for dyslipidemia were calculated for quartiles of SF concentrations, with the lowest ferritin quartile as the reference. The final subjects consisted of 2676 participants (1290 males and 1386 females). ORs for dyslipidemia were the highest in the fourth quartile (Q4) of SF both in males (OR: 1.60, 95% CI: 1.12-2.28) and females (OR: 1.52, 95% CI: 1.07-2.17). The crude ORs (95% CI) for the risk of High TC and High LDL-C increased progressively in both genders. However, after adjusting for covariates, the trend of significance was only present in females. Finally, the association between total daily iron intake and the four types of dyslipidemia was examined, revealing that the risk of High TG in the third quartile of the total daily iron intake was 2.16 times greater in females (adjusted OR: 3.16, 95% CI: 1.38-7.23). SF concentrations were remarkably associated with dyslipidemia. In females, daily dietary iron intake was associated with High-TG dyslipidemia.
Collapse
Affiliation(s)
| | | | | | | | - Tianyou Ma
- Department of Nutrition and Food Safety, School of Public Health, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiaoqin Luo
- Department of Nutrition and Food Safety, School of Public Health, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
42
|
Kannan M, Sil S, Oladapo A, Thangaraj A, Periyasamy P, Buch S. HIV-1 Tat-mediated microglial ferroptosis involves the miR-204–ACSL4 signaling axis. Redox Biol 2023; 62:102689. [PMID: 37023693 PMCID: PMC10106521 DOI: 10.1016/j.redox.2023.102689] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/20/2023] [Accepted: 03/26/2023] [Indexed: 04/04/2023] Open
Abstract
This study was focused on exploring the role of the HIV-1 Tat protein in mediating microglial ferroptosis. Exposure of mouse primary microglial cells (mPMs) to HIV-1 Tat protein resulted in induction of ferroptosis, which was characterized by increased expression of Acyl-CoA synthetase long-chain family member 4 (ACSL4), in turn, leading to increased generation of oxidized phosphatidylethanolamine, elevated levels of lipid peroxidation, upregulated labile iron pool (LIP) and ferritin heavy chain-1 (FTH1), decreased glutathione peroxidase-4 and mitochondrial outer membrane rupture. Also, inhibition of ferroptosis by ferrostatin-1 (Fer-1) or deferoxamine (DFO) treatment suppressed ferroptosis-related changes in mPMs. Similarly, the knockdown of ACSL4 by gene silencing also inhibited ferroptosis induced by HIV-1 Tat. Furthermore, increased lipid peroxidation resulted in increased release of proinflammatory cytokines, such as TNFα, IL6, and IL1β and microglial activation. Pretreatment of mPMs with Fer-1 or DFO further blocked HIV-1 Tat-mediated microglial activation in vitro and reduced the expression and release of proinflammatory cytokines. We identified miR-204 as an upstream modulator of ACSL4, which was downregulated in mPMs exposed to HIV-1 Tat. Transient transfection of mPMs with miR-204 mimics reduced the expression of ACSL4 while inhibiting HIV-1 Tat-mediated ferroptosis and the release of proinflammatory cytokines. These in vitro findings were further validated in HIV-1 transgenic rats as well as HIV + ve human brain samples. Overall, this study underscores a novel mechanism(s) underlying HIV-1 Tat-mediated ferroptosis and microglial activation involving miR-204-ACSL4 signaling.
Collapse
|
43
|
Martins TS, Costa RS, Vilaça R, Lemos C, Teixeira V, Pereira C, Costa V. Iron Limitation Restores Autophagy and Increases Lifespan in the Yeast Model of Niemann-Pick Type C1. Int J Mol Sci 2023; 24:6221. [PMID: 37047194 PMCID: PMC10094029 DOI: 10.3390/ijms24076221] [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: 02/07/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Niemann-Pick type C1 (NPC1) is an endolysosomal transmembrane protein involved in the export of cholesterol and sphingolipids to other cellular compartments such as the endoplasmic reticulum and plasma membrane. NPC1 loss of function is the major cause of NPC disease, a rare lysosomal storage disorder characterized by an abnormal accumulation of lipids in the late endosomal/lysosomal network, mitochondrial dysfunction, and impaired autophagy. NPC phenotypes are conserved in yeast lacking Ncr1, an orthologue of human NPC1, leading to premature aging. Herein, we performed a phosphoproteomic analysis to investigate the effect of Ncr1 loss on cellular functions mediated by the yeast lysosome-like vacuoles. Our results revealed changes in vacuolar membrane proteins that are associated mostly with vesicle biology (fusion, transport, organization), autophagy, and ion homeostasis, including iron, manganese, and calcium. Consistently, the cytoplasm to vacuole targeting (Cvt) pathway was increased in ncr1∆ cells and autophagy was compromised despite TORC1 inhibition. Moreover, ncr1∆ cells exhibited iron overload mediated by the low-iron sensing transcription factor Aft1. Iron deprivation restored the autophagic flux of ncr1∆ cells and increased its chronological lifespan and oxidative stress resistance. These results implicate iron overload on autophagy impairment, oxidative stress sensitivity, and cell death in the yeast model of NPC1.
Collapse
Affiliation(s)
- Telma S. Martins
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Rafaela S. Costa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Rita Vilaça
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Carolina Lemos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vitor Teixeira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Clara Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Vítor Costa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
44
|
Sivagurunathan N, Gnanasekaran P, Calivarathan L. Mitochondrial Toxicant-Induced Neuronal Apoptosis in Parkinson's Disease: What We Know so Far. Degener Neurol Neuromuscul Dis 2023; 13:1-13. [PMID: 36726995 PMCID: PMC9885882 DOI: 10.2147/dnnd.s361526] [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: 07/10/2022] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common progressive neurodegenerative diseases caused by the loss of dopamine-producing neuronal cells in the region of substantia nigra pars compacta of the brain. During biological aging, neuronal cells slowly undergo degeneration, but the rate of cell death increases tremendously under some pathological conditions, leading to irreversible neurodegenerative diseases. By the time symptoms of PD usually appear, more than 50 to 60% of neuronal cells have already been destroyed. PD symptoms often start with tremors, followed by slow movement, stiffness, and postural imbalance. The etiology of PD is still unknown; however, besides genetics, several factors contribute to neurodegenerative disease, including exposure to pesticides, environmental chemicals, solvents, and heavy metals. Postmortem brain tissues of patients with PD show mitochondrial abnormalities, including dysfunction of the electron transport chain. Most chemicals present in our environment have been shown to target the mitochondria; remarkably, patients with PD show a mild deficiency in NADH dehydrogenase activity, signifying a possible link between PD and mitochondrial dysfunction. Inhibition of electron transport complexes generates free radicals that further attack the macromolecules leading to neuropathological conditions. Apart from that, oxidative stress also causes neuroinflammation-mediated neurodegeneration due to the activation of microglial cells. However, the mechanism that causes mitochondrial dysfunction, especially the electron transport chain, in the pathogenesis of PD remains unclear. This review discusses the recent updates and explains the possible mechanisms of mitochondrial toxicant-induced neuroinflammation and neurodegeneration in PD.
Collapse
Affiliation(s)
- Narmadhaa Sivagurunathan
- Molecular Pharmacology and Toxicology Laboratory, Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Priyadharshini Gnanasekaran
- Molecular Pharmacology and Toxicology Laboratory, Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology and Toxicology Laboratory, Department of Biotechnology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India,Correspondence: Latchoumycandane Calivarathan, Molecular Pharmacology and Toxicology Laboratory, Department of Biotechnology (Sponsored by DST-FIST), School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, 610005, India, Tel +91-6381989116, Email
| |
Collapse
|
45
|
Conceição M, Beserra FP, Aldana Mejia JA, Caldas GR, Tanimoto MH, Luzenti AM, Gaspari PDM, Evans ND, Bastos JK, Pellizzon CH. Guttiferones: An insight into occurrence, biosynthesis, and their broad spectrum of pharmacological activities. Chem Biol Interact 2023; 370:110313. [PMID: 36566914 DOI: 10.1016/j.cbi.2022.110313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
Guttiferones belong to the polyisoprenylated benzophenone, a class of compounds, a very restricted group of natural plant products, especially in the Clusiaceae family. They are commonly found in bark, stem, leaves, and fruits of plants of the genus Garcinia and Symphonia. Guttiferones have the following classifications according to their chemical structure: A, B, C, D, E, F, G, H, I, J, K, L, M, N, O, P, Q, R, S, and T. All of them have received growing attention due to its multiple biological activities. This review provides a first comprehensive approach to plant sources, phytochemical profile, specific pharmacological effects, and mechanisms of guttiferones already described. Studies indicate a broad spectrum of pharmacological activities, such as: anti-inflammatory, immunomodulatory, antioxidant, antitumor, antiparasitic, antiviral, and antimicrobial. Despite the low toxicity of these compounds in healthy cells, there is a lack of studies in the literature related to toxicity in general. Given their beneficial effects, guttiferones are expected to be great potential drug candidates for treating cancer and infectious and transmissible diseases. However, further studies are needed to elucidate their toxicity, specific molecular mechanisms and targets, and to perform more in-depth pharmacokinetic studies. This review highlights chemical properties, biological characteristics, and mechanisms of action so far, offering a broad view of the subject and perspectives for the future of guttiferones in therapeutics.
Collapse
Affiliation(s)
- Mariana Conceição
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Fernando Pereira Beserra
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil; Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.
| | - Jennyfer Andrea Aldana Mejia
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Gabriel Rocha Caldas
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Matheus Hikaru Tanimoto
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Andréia Marincek Luzenti
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Priscyla Daniely Marcato Gaspari
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Nicholas David Evans
- Human Development and Health, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Jairo Kenupp Bastos
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Cláudia Helena Pellizzon
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
46
|
Nayeem A, Ali MF, Shariffuddin JH. The recent development of inverse vulcanized polysulfide as an alternative adsorbent for heavy metal removal in wastewater. ENVIRONMENTAL RESEARCH 2023; 216:114306. [PMID: 36191616 DOI: 10.1016/j.envres.2022.114306] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/11/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023]
Abstract
Inverse vulcanized polysulfides have been used as low-cost and effective adsorbents to remediate heavy metals in wastewater. Inverse vulcanization introduces sustainable polysulfide synthesis by solving the rapid desulfurization problem of unstable polysulfides, and provides superior performance compared to conventional commercial adsorbents. The review discussed the brief applications of the inverse vulcanized polysulfides to remove heavy metal wastewater and emphasized the modified synthesis processes for enhanced uptake ratios. The characteristics of polysulfide adsorbents, which play a vital role during the removal process are highlighted with a proper discussion of the interaction between metal ions and polysulfides. The review paper concludes with remarks on the future outlook of these low-cost adsorbents with high selectivity to heavy metals. These polysulfide adsorbents can be prepared using a wide variety of crosslinker monomers including organic hydrocarbons, cooking oils, and agro-based waste materials. They have shown good surface area and excellent metal-binding capabilities compared to the commercially available adsorbents. Proper postmodification processes have enabled the benefits of repetitive uses of the polysulfide adsorbents. The improved surface area obtained by appropriate choice of crosslinkers, modified synthesis techniques, and regeneration through post-modification has made inverse vulcanized polysulfides capable of removing.
Collapse
Affiliation(s)
- Abdullah Nayeem
- Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang, 26300 Gambang, Pahang, Malaysia
| | - Mohd Faizal Ali
- Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang, 26300 Gambang, Pahang, Malaysia
| | - Jun Haslinda Shariffuddin
- Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang, 26300 Gambang, Pahang, Malaysia; Centre for Sustainability of Ecosystem & Earth Resources, Lebuhraya Tun Razak, 26300 Gambang, Kuantan, Pahang Darul Makmur, Malaysia.
| |
Collapse
|
47
|
Yang M, Wang C, Zhao G, Kong D, Liu L, Yuan S, Chen W, Feng C, Li Z. Comparative Analysis of the Pre- and Post-Medication Effects of Antipsychotic Agents on the Blood-Based Oxidative Stress Biomarkers in Patients with Schizophrenia: A Meta-Analysis. Curr Neuropharmacol 2023; 21:340-352. [PMID: 35794775 PMCID: PMC10190148 DOI: 10.2174/1570159x20666220706101021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 04/19/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Studies have shown that oxidative stress (OS) is related to the pathophysiology of schizophrenia (SCZ), but whether antipsychotics can induce OS has not been investigated well. Moreover, antipsychotics have differential effects on the OS level modulation, i.e., different types of antipsychotics have different effects on the cellular antioxidants or pro-oxidants. METHODS We followed the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines and investigated the OS indicators including both enzymatic and nonenzymatic markers, such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), malondialdehyde (MDA), glutathione (GSH), vitamin C, etc., of SCZ patients at baseline and follow-up of mono-medication. RESULTS Twenty studies met the inclusion criteria, with a total of 1162 patients enrolled at baseline, and 1105 patients completed the follow-up. OS markers were changed after a period of antipsychotic treatment in SCZ patients. The GPx activity and MDA level decreased in the whole blood (P<0.05), also the serum MDA level decreased (P<0.05). For the first-episode SCZ patients, the activity of GPx and the level of MDA decreased, while the level of vitamin C increased (all P<0.05). The levels of MDA in patients receiving atypical antipsychotics decreased (P<0.05), while the level of GSH in patients with typical antipsychotics decreased (P=0.05). CONCLUSION Antipsychotic medication may cause changes in the levels of OS markers in different blood samples of SCZ patients. However, the available studies might not be sufficient to reveal the underlying facts accurately due to the poor quality of experimental designs in the published literature.
Collapse
Affiliation(s)
- Mi Yang
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Chunzhi Wang
- Department of Psychiatry, Qingdao Mental Health Center, Qingdao, China
| | - Guocheng Zhao
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Di Kong
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Liju Liu
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Shuai Yuan
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Wei Chen
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Can Feng
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Zezhi Li
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Psychiatry, Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| |
Collapse
|
48
|
Valencia-Hernández I, González-Piña R, García-Díaz G, Ramos-Languren L, Parra-Cid C, Lomelí J, Montes S, Ríos C, Bueno-Nava A. Alpha 2-adrenergic receptor activation reinstates motor deficits in rats recovering from cortical injury. Neural Regen Res 2023; 18:875-880. [DOI: 10.4103/1673-5374.353501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
49
|
Han S, Gim Y, Jang EH, Hur EM. Functions and dysfunctions of oligodendrocytes in neurodegenerative diseases. Front Cell Neurosci 2022; 16:1083159. [PMID: 36605616 PMCID: PMC9807813 DOI: 10.3389/fncel.2022.1083159] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Neurodegenerative diseases (NDDs) are characterized by the progressive loss of selectively vulnerable populations of neurons, which is responsible for the clinical symptoms. Although degeneration of neurons is a prominent feature that undoubtedly contributes to and defines NDD pathology, it is now clear that neuronal cell death is by no means mediated solely by cell-autonomous mechanisms. Oligodendrocytes (OLs), the myelinating cells of the central nervous system (CNS), enable rapid transmission of electrical signals and provide metabolic and trophic support to neurons. Recent evidence suggests that OLs and their progenitor population play a role in the onset and progression of NDDs. In this review, we discuss emerging evidence suggesting a role of OL lineage cells in the pathogenesis of age-related NDDs. We start with multiple system atrophy, an NDD with a well-known oligodendroglial pathology, and then discuss Alzheimer's disease (AD) and Parkinson's disease (PD), NDDs which have been thought of as neuronal origins. Understanding the functions and dysfunctions of OLs might lead to the advent of disease-modifying strategies against NDDs.
Collapse
Affiliation(s)
- Seungwan Han
- Laboratory of Neuroscience, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
- BK21 Four Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yunho Gim
- Laboratory of Neuroscience, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
- BK21 Four Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Eun-Hae Jang
- Laboratory of Neuroscience, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea
| | - Eun-Mi Hur
- Laboratory of Neuroscience, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
- BK21 Four Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, South Korea
- Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
50
|
Sergi CM. Epigallocatechin gallate for Parkinson's disease. Clin Exp Pharmacol Physiol 2022; 49:1029-1041. [PMID: 35748799 DOI: 10.1111/1440-1681.13691] [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: 02/02/2021] [Revised: 01/03/2022] [Accepted: 06/19/2022] [Indexed: 11/28/2022]
Abstract
In the last couple of decades, we have experienced increased use of nutraceuticals worldwide with a demand for organic foods, which has been elevated to an extent probably unmatched with other periods of our civilization. One of the nutraceuticals that gained attention is epigallocatechin gallate (EGCG), a polyphenol in green tea. It has been suggested that diseases of the central nervous system can benefit from consuming some antioxidants, despite current results showing little evidence for their use in preventing and treating these diseases. ECGC may be beneficial in delaying the neurodegeneration of the substantia nigra regardless of the origin of Parkinson's disease (PD). This review covers the effect of EGCG on vitro and animal models of PD, the potential mechanisms of neuroprotection involved and summaries recent clinical trials in human PD. This review also aims to provide an investigative analysis of the current knowledge in this field and to identify putative crucial issues. Environmental factors such as dietary habits, drug use and social interaction are all factors that influence the evolution of neurodegenerative diseases. Therefore, the use of nutraceuticals requires further investigation.
Collapse
Affiliation(s)
- Consolato M Sergi
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
- Anatomic Pathology, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
- Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|