1
|
Zhu J, Liu L, Wu J, Bai L. Rodent models for dry eye syndrome (DES). Cont Lens Anterior Eye 2025:102383. [PMID: 39956692 DOI: 10.1016/j.clae.2025.102383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Dry eye syndrome (DES) is a range of ophthalmic conditions characterized by compromised tear film homeostasis, resulting from various pathological factors and primarily manifesting as ocular discomfort and impaired ocular surface integrity. With the rise in screen time due to modern lifestyles, the prevalence of DES is increasing annually, posing a significant global public health challenge. Pathophysiologically, DES involves damage to the lacrimal functional unit (LFU), including the lacrimal glands, meibomian glands, and corneoconjunctival epithelium, highlighting its multifactorial etiology. Current treatments mainly focus on artificial tears for moisture replacement and anti-inflammatory therapies, but both are limited. Consequently, animal models are crucial for understanding the complex pathological mechanisms of DES and identifying potential therapeutic agents. Rodent eyes, with their structural and physiological similarities to human eyes and cost-effectiveness, have become widely used in DES research. This manuscript reviews the current understanding of DES pathogenesis and rodent models, discussing their strengths, weaknesses, and relevant genetic models. The aim is to furnish critical insights and provide a scholarly resource to propel future investigative endeavors into the pathogenesis of and therapy for DES.
Collapse
Affiliation(s)
- Jingyun Zhu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liu Liu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Wu
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lang Bai
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Baldensperger T, Preissler M, Becker CFW. Non-enzymatic posttranslational protein modifications in protein aggregation and neurodegenerative diseases. RSC Chem Biol 2025; 6:129-149. [PMID: 39722676 PMCID: PMC11667106 DOI: 10.1039/d4cb00221k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Highly reactive metabolic intermediates and other small molecules frequently react with amino acid side chains, leading to non-enzymatic posttranslational modifications (nPTMs) of proteins. The abundance of these modifications increases under high metabolic activity or stress conditions and can dramatically impact protein structure and function. Although protein quality control mechanisms typically mitigate the effects of these impaired proteins, in long-lived and degradation-resistant proteins, nPTMs accumulate. In some cases, such as cataract development and diabetes, clear links between nPTMs, aging, and disease progression have been established. In neurodegenerative diseases such as Alzheimer's and Parkinson's disease, a key question is whether accumulation of nPTMs is a cause or consequence of protein aggregation. This review focuses on major nPTMs found on proteins with central roles in neurodegenerative diseases such as α-synuclein, β-amyloid, and tau. We summarize current knowledge on the formation of these modifications and discuss their potential impact on disease onset and progression. Additionally, we examine what is known to date about how nPTMs impair cellular detoxification, repair, and degradation systems. Finally, we critically discuss the available methodologies to systematically investigate nPTMs at the molecular level and outline suitable approaches to study their effects on protein aggregation. We aim to foster more research into the role of nPTMs in neurodegeneration by adapting methodologies that have proven successful in studying enzymatic posttranslational modifications. Specifically, we advocate for site-specific incorporation of these modifications into target proteins using advanced chemical and molecular biology techniques.
Collapse
Affiliation(s)
- Tim Baldensperger
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
| | - Miriam Preissler
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
- University of Vienna, Vienna Doctoral School in Chemistry (DoSChem) Währinger Str. 42 1090 Vienna Austria
| | - Christian F W Becker
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry Währinger Str. 38 1090 Vienna Austria
| |
Collapse
|
3
|
Baek Y, Kim H, Lee D, Kim D, Jo E, Roh SH, Ha NC. Structural insights into the role of reduced cysteine residues in SOD1 amyloid filament formation. Proc Natl Acad Sci U S A 2025; 122:e2408582122. [PMID: 39874287 PMCID: PMC11804504 DOI: 10.1073/pnas.2408582122] [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: 04/30/2024] [Accepted: 12/25/2024] [Indexed: 01/30/2025] Open
Abstract
The formation of superoxide dismutase 1 (SOD1) filaments has been implicated in amyotrophic lateral sclerosis (ALS). Although the disulfide bond formed between Cys57 and Cys146 in the active state has been well studied, the role of the reduced cysteine residues, Cys6 and Cys111, in SOD1 filament formation remains unclear. In this study, we investigated the role of reduced cysteine residues by determining and comparing cryoelectron microscopy (cryo-EM) structures of wild-type (WT) and C6A/C111A SOD1 filaments under thiol-based reducing and metal-depriving conditions, starting with protein samples possessing enzymatic activity. The C6A/C111A mutant SOD1 formed filaments more rapidly than the WT protein. The mutant structure had a unique paired-protofilament arrangement, with a smaller filament core than that of the single-protofilament structure observed in WT SOD1. Although the single-protofilament form developed more slowly, cross-seeding experiments demonstrated the predominance of single-protofilament morphology over paired protofilaments, regardless of the presence of the Cys6 and Cys111 mutations. These findings highlight the importance of the number of amino acid residues within the filament core in determining the energy requirements for assembly. Our study provides insights into ALS pathogenesis by elucidating the initiation and propagation of filament formation, which potentially leads to deleterious amyloid filaments.
Collapse
Affiliation(s)
- Yeongjin Baek
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, CALS, Seoul National University, Seoul08826, Republic of Korea
| | - Hyunmin Kim
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul08826, Republic of Korea
- Molecular Systems Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg69117, Germany
| | - Dukwon Lee
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, CALS, Seoul National University, Seoul08826, Republic of Korea
| | - Doyeon Kim
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, CALS, Seoul National University, Seoul08826, Republic of Korea
| | - Eunbyul Jo
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, CALS, Seoul National University, Seoul08826, Republic of Korea
| | - Soung-Hun Roh
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul08826, Republic of Korea
| | - Nam-Chul Ha
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, CALS, Seoul National University, Seoul08826, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul08826, Republic of Korea
| |
Collapse
|
4
|
Otenaike TA, Farodoye OM, de Silva MM, Loreto JS, Adedara AO, Dos Santos MM, de Prestes AS, Barbosa NV, da Rocha JBT, Lobo LE, Wagner R, Abolaji AO, Loreto ELS. Nicotine and Vape: Drugs of the Same Profile Flock Together. J Biochem Mol Toxicol 2024; 38:e70075. [PMID: 39601203 DOI: 10.1002/jbt.70075] [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: 02/22/2024] [Revised: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
Smoking, a major behavioral health burden, causes preventable and premature deaths globally. Nicotine, the addictive component present in tobacco products and Electronic cigarettes (E-cigarettes, vape), can bind to nicotinic acetylcholine receptors in the brain to trigger a dopamine release that reinforces smoking. Despite the widespread usage of nicotine, its mechanisms of toxicity, particularly in e-cigarettes, are poorly understood. Using Drosophila melanogaster as a model organism, this study aims to investigate the mechanism of the toxicity of nicotine and vape. Behavioral parameters, oxidative stress indicators, mRNA expression levels of Dopamine 1- receptor 1 (Dop1R1), Acetyl-coenzyme A synthetase (AcCoAs), and apoptotic proteins were assessed in the flies after a 5-day exposure to varying concentrations of nicotine (0.15, 0.25, and 0.35 mg/mL diet) and vape (0.06, 0.08, and 0.12 mg/mL diet). Furthermore, Gas Chromatography-Mass Spectrometry (GC/MS) and Gas Chromatography-Flame Ionization Detection (GC/FID) analyzes were conducted to gain more insight on the composition of the vape used in study. Findings indicate that both nicotine and vape exposure significantly reduced lifespan, impaired locomotor activity, and disrupted sleep patterns. Notably, nicotine exposure stimulated Dop1R1 transcription and altered Acetyl-CoA gene expression, impacting the viability and behavior of the flies. Elevated levels of reactive oxygen biomarkers were observed, contributing to cellular damage through oxidative stress and apoptotic mechanisms mediated by the Reaper and DIAP1 proteins. Additionally, the composition analysis of vape liquid revealed the presence of propylene glycol, nicotine, methyl esters, and an unidentified compound. This study highlights the complex interplay between nicotine, gene expression, and physiological responses in Drosophila.
Collapse
Affiliation(s)
- Titilayomi A Otenaike
- Doctoral Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), CEP, Porto Alegre, Brazil
- Drosophila Research and Training Centre, Ibadan, Nigeria
| | - Oluwabukola M Farodoye
- Doctoral Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), CEP, Porto Alegre, Brazil
- Drosophila Research and Training Centre, Ibadan, Nigeria
| | - Monica M de Silva
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Julia S Loreto
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Adeola O Adedara
- Drosophila Research and Training Centre, Ibadan, Nigeria
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Matheus M Dos Santos
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Alessandro S de Prestes
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Nilda V Barbosa
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - João B T da Rocha
- Center for Natural and Exact Sciences, Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Luiz E Lobo
- Department of Technology and Food Science, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Roger Wagner
- Department of Technology and Food Science, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| | - Amos O Abolaji
- Drosophila Research and Training Centre, Ibadan, Nigeria
- Drosophila Laboratory, Department of Biochemistry, College of Medicine, Molecular Drug Metabolism and Toxicology Unit, Ibadan, Nigeria
| | - Elgion L S Loreto
- Doctoral Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), CEP, Porto Alegre, Brazil
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria (UFSM), CEP, Santa Maria, Brazil
| |
Collapse
|
5
|
Liao S, Börmel L, Müller AK, Gottschalk L, Pritsch N, Preisner LZ, Samokhina O, Schwarz M, Kipp AP, Schlörmann W, Glei M, Schubert M, Schmölz L, Wallert M, Lorkowski S. α-Tocopherol Long-Chain Metabolite α-T-13'-COOH Exhibits Biphasic Effects on Cell Viability, Induces ROS-Dependent DNA Damage, and Modulates Redox Status in Murine RAW264.7 Macrophages. Mol Nutr Food Res 2024; 68:e2400455. [PMID: 39548913 DOI: 10.1002/mnfr.202400455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/13/2024] [Indexed: 11/18/2024]
Abstract
SCOPE The α-tocopherol long-chain metabolite α-tocopherol-13'-hydroxy-chromanol (α-T-13'-COOH) is a proposed regulatory intermediate of endogenous vitamin E metabolism. Effects of α-T-13'-COOH on cell viability and adaptive stress response are not well understood. The present study aims to investigate the concentration-dependent effects of α-T-13'-COOH on cellular redox homeostasis, genotoxicity, and cytotoxicity in murine RAW264.7 macrophages as a model system. METHODS AND RESULTS Murine RAW264.7 macrophages are exposed to various dosages of α-T-13'-COOH to determine its regulatory effects on reactive oxygen species (ROS) production, DNA damage, expression of stress-related markers, and the activity of ROS scavenging enzymes including superoxide dismutases, catalase, and glutathione-S-transferases. The impact on cell viability is assessed by analyzing cell proliferation, cell cycle arrest, and cell apoptosis. CONCLUSION α-T-13'-COOH influences ROS production and induces DNA damage in a dose-dependent manner. The metabolite modulates the activity of ROS-scavenging enzymes, with significant changes observed in the activities of antioxidant enzymes. A biphasic response affecting cell viability is noted: sub-micromolar doses of α-T-13'-COOH promote cell proliferation and enhance DNA synthesis, whereas supraphysiological doses lead to DNA damage and cytotoxicity. It hypothesizes an adaptive stress response, characterized by upregulation of ROS detoxification mechanisms, enhanced cell cycle arrest, and increased apoptosis, indicating a correlation with oxidative stress and subsequent cellular damage.
Collapse
Affiliation(s)
- Sijia Liao
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
| | - Lisa Börmel
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
| | - Anke Katharina Müller
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
| | - Luisa Gottschalk
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Nadine Pritsch
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Lara Zoé Preisner
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Oleksandra Samokhina
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Maria Schwarz
- Department of Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Anna P Kipp
- Department of Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Wiebke Schlörmann
- Department of Applied Nutritional Toxicology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Michael Glei
- Department of Applied Nutritional Toxicology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Martin Schubert
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
| | - Lisa Schmölz
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
- Member of Leibniz Research Alliance, Leibniz Health Technology and Leibniz Centre for Photonics in Infection Research, Leibniz Institute of Photonic Technology, Jena, Germany
| | - Maria Wallert
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
| | - Stefan Lorkowski
- Department of Nutritional Physiology and Biochemistry, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Germany
| |
Collapse
|
6
|
Wang H, Zhang L, Shang Y. DEPTOR attenuates asthma progression by suppressing endoplasmic reticulum stress through SOD1. Biol Direct 2024; 19:114. [PMID: 39533404 PMCID: PMC11556204 DOI: 10.1186/s13062-024-00557-z] [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] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Endoplasmic reticulum (ER) stress has been shown to play a pivotal role in the pathogenesis of asthma. DEPTOR (DEP Domain Containing MTOR Interacting Protein) is an endogenous mTOR inhibitor that participates in various physiological processes such as cell growth, apoptosis, autophagy, and ER homeostasis. However, the role of DEPTOR in the pathogenesis of asthma is still unknown. In this study, an ovalbumin (OVA)-induced mice model and IL-13 induced 16HBE cells were used to evaluate the effect of DEPTOR on asthma. A decreased DEPTOR expression was shown in the lung tissues of OVA-mice and IL-13 induced 16HBE cells. Upregulation of DEPTOR attenuated airway goblet cell hyperplasia, inhibited mucus hypersecretion, decreased the expression of mucin MUC5AC, and suppressed the level of inflammatory factors IL-4 and IL-5, which were all induced by OVA treatment. The increased protein expression of ER stress markers GRP78, CHOP, unfolded protein response (UPR) related proteins, and apoptosis markers in OVA mice were also inhibited by DEPTOR overexpression. In IL-13 induced 16HBE cells, overexpression of DEPTOR decreased IL-4, IL-5, and MUC5AC levels, preventing ER stress response and UPR process. Furthermore, from the proteomics results, we identified that SOD1 (Cu/Zn Superoxide Dismutase 1) may be the downstream factor of DEPTOR. Similar to DEPTOR, upregulation of SOD1 alleviated asthma progression. Rescue experiments showed that SOD1 inhibition abrogates the remission effect of DEPTOR on ER stress in vitro. In conclusion, these data suggested that DEPTOR attenuates asthma progression by suppressing endoplasmic reticulum stress through SOD1.
Collapse
Affiliation(s)
- Hao Wang
- Department of Pediatric Respiratory Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Lei Zhang
- Department of General Surgery, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Yunxiao Shang
- Department of Pediatric Respiratory Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
7
|
Gao L, Wang W, Ma H, Yin M, Yang X, Han R, Ohara S, Kim D, Wang G. Bioinformatics analysis reveals SOD1 is a prognostic factor in lung adenocarcinoma. Transl Cancer Res 2024; 13:5522-5534. [PMID: 39525006 PMCID: PMC11543046 DOI: 10.21037/tcr-24-1400] [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: 08/10/2024] [Accepted: 09/13/2024] [Indexed: 11/16/2024]
Abstract
Background Lung cancer is a major cause of cancer-related deaths worldwide. Unfortunately, non-small cell lung cancer (NSCLC) often lacks clear clinical symptoms and molecular markers for early diagnosis, which can hinder the initiation of timely treatments. In this study, we conducted an extensive bioinformatics analysis of copper-zinc superoxide dismutase (SOD1), a molecule linked to lung adenocarcinoma (LUAD) to enhance early detection and treatment approaches for this condition. Methods A bioinformatics analysis was conducted using a dataset from The Cancer Genome Atlas (TCGA) database. Several analytical methods, such as a differential expression analysis, a Kaplan-Meier survival analysis, a clinicopathological analysis, an enrichment analysis, protein-protein interaction (PPI) network construction using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database, and an immunoreactivity analysis of SOD1 expression in LUAD using TIMER were employed. We further validated the expression of SOD1 in LUAD through in vitro experiments using quantitative polymerase chain reaction (qPCR) and Western blot. Results Our findings indicate that LUAD tissues exhibited significantly higher expression levels of SOD1 than healthy tissues. The univariate Cox analysis showed that the elevated level was linked to unfavorable overall survival (OS) rates. Further, the Cox regression analysis of multiple variables suggested that elevated SOD1 expression levels acted as an autonomous prognosticator for unfavorable OS. We also conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and a gene set enrichment analysis (GSEA) and observed differential pathway enrichment among patients with high SOD1 expression. In addition, a correlation between SOD1 and immune cell infiltration was found. The in vitro experiments confirmed that SOD1 expression was upregulated in LUAD. Conclusions SOD1 could serve as a reliable prognostic indicator in individuals diagnosed with LUAD. Our findings may prove valuable in the development of therapeutic and prognostic markers for LUAD. The potential clinical utility of SOD1 in LUAD requires further investigation.
Collapse
Affiliation(s)
- Ling Gao
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Wei Wang
- Department of Neurosurgery, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Haishan Ma
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Minghui Yin
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Xuejiao Yang
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Ruihui Han
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| | - Shuta Ohara
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Dohun Kim
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Chungbuk National University and Chungbuk National University Hospital, Cheongju, Korea
| | - Guangyan Wang
- Clinical Laboratory, Chuxiong Yi Autonomous Prefecture People’s Hospital, Chuxiong, China
| |
Collapse
|
8
|
Yang Y, Wu J, Wang L, Ji G, Dang Y. Copper homeostasis and cuproptosis in health and disease. MedComm (Beijing) 2024; 5:e724. [PMID: 39290254 PMCID: PMC11406047 DOI: 10.1002/mco2.724] [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: 03/28/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Copper is a vital trace element in human physiology, essential for the synthesis of numerous crucial metabolic enzymes and facilitation of various biological processes. Regulation of copper levels within a narrow range is imperative for maintaining metabolic homeostasis. Numerous studies have demonstrated the significant roles of copper homeostasis and cuproptosis in health and disease pathogenesis. However, a comprehensive and up-to-date systematic review in this domain remains absent. This review aims to consolidate recent advancements in understanding the roles of cuproptosis and copper homeostasis in health and disease, focusing on the underlying mechanisms and potential therapeutic interventions. Dysregulation of copper homeostasis, manifesting as either copper excess or deficiency, is implicated in the etiology of various diseases. Cuproptosis, a recently identified form of cell death, is characterized by intracellular copper overload. This phenomenon mediates a diverse array of evolutionary processes in organisms, spanning from health to disease, and is implicated in genetic disorders, liver diseases, neurodegenerative disorders, and various cancers. This review provides a comprehensive summary of the pathogenic mechanisms underlying cuproptosis and copper homeostasis, along with associated targeted therapeutic agents. Furthermore, it explores future research directions with the potential to yield significant advancements in disease treatment, health management, and disease prevention.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| | - Jiaxuan Wu
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine University of Ottawa Ottawa Ontario Canada
- China-Canada Centre of Research for Digestive Diseases University of Ottawa Ottawa Ontario Canada
| | - Guang Ji
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| | - Yanqi Dang
- Institute of Digestive Diseases China-Canada Center of Research for Digestive Diseases Longhua Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine) Shanghai China
| |
Collapse
|
9
|
Xie GF, Cheng JY, Liu Y, Sun J, Hua DX, He QY, Yang J, Zhao H, Lu YJ, Fu L. Serum SOD1 level predicts the severity and prognosis of community-acquired pneumonia patients. Am J Med Sci 2024; 368:311-319. [PMID: 38825074 DOI: 10.1016/j.amjms.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 03/28/2024] [Accepted: 05/20/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Superoxide dismutase 1 (SOD1) is one of the most important participants of antioxidant enzyme system in biological system. Previous studies have found that SOD1 is associated with many inflammatory diseases. The goal of this study was to assess the associations of serum SOD1 with the severity and prognosis in community-acquired pneumonia (CAP) patients by a prospective cohort study. METHODS CAP patients were enrolled from the Second Affiliated Hospital of Anhui Medical University. Peripheral blood samples were gathered. The level of serum SOD1 was detected through enzyme linked immunosorbent assay (ELISA). Clinical characteristics and demographic information were analyzed. RESULTS The level of serum SOD1 was gradually upregulated with elevated CAP severity scores. Spearman correlation coefficient or Pearson rank correlation analyses indicated that serum SOD1 was strongly connected with many clinical parameters among CAP patients. Further linear and logistic regression analyses found that the level of serum SOD1 was positively associated with CRB-65, CURB-65, SMART-COP, and CURXO scores among CAP patients. Moreover, serum higher SOD1 at admission substantially increased the risks of ICU admission, mechanical ventilation, vasoactive agent usage, death, and longer hospital stays during hospitalization. Serum SOD1 level combination with CAP severity scores elevated the predictive abilities for severity and death compared with alone serum SOD1 and CAP severity scores in CAP patients during hospitalization. CONCLUSION The level of serum SOD1 is positively associated with the severity and poor prognosis in CAP patients, suggesting that SOD1 is implicated in the initiation and progression of CAP. Serum SOD1 may be regarded as a biomarker to appraise the severity and prognosis for CAP patients.
Collapse
Affiliation(s)
- Guo-Fang Xie
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Jia-Yi Cheng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Ying Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Jing Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Dong-Xu Hua
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Qi-Yuan He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Jin Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - You-Jin Lu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China; Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, PR China.
| |
Collapse
|
10
|
Miyake N, Ochi N, Takeyama M, Isozaki H, Ichihara E, Yamane H, Fukazawa T, Nagasaki Y, Kawahara T, Nakanishi H, Hiraki A, Kiura K, Takigawa N. A novel molecular target, superoxide dismutase 1, in ALK inhibitor-resistant lung cancer cells, detected through proteomic analysis. Exp Cell Res 2024; 442:114266. [PMID: 39313177 DOI: 10.1016/j.yexcr.2024.114266] [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: 03/30/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUNDS To the best of our knowledge, there are no reports of proteomic analysis for the identification of unknown proteins involved in resistance to anaplastic lymphoma kinase (ALK) inhibitors. In this study, we investigated the proteins involved in resistance to alectinib, a representative ALK inhibitor, through proteomic analysis and the possibility of overcoming resistance. METHODS An ALK-positive lung adenocarcinoma cell line (ABC-11) and the corresponding alectinib-resistant cell line (ABC-11/CHR2) were used. Two-dimensional difference gel electrophoresis (2D DIGE) was performed; the stained gel was scanned and the spots were analyzed using DeCyder TM2D 7.0. Mass spectrometry (MS) with the UltrafleXtreme matrix-assisted laser desorption ionization-tandem time-of-flight (MALDI-TOF/TOF) MS system was performed. For the MS/MS analysis, the samples were spotted on an AnchorChipTM 600 TF plate. The peptide masses obtained in the reflector positive mode were acquired at m/z of 400-6000. MS/MS data were searched against the NCBI protein databases. Growth inhibition was measured using an MTT assay. The isobologram and combination index were calculated based on the median-effect analysis. Western blotting was performed using antibodies, including superoxide dismutase (SOD) 1, MET, ERK, PARP, AKT, and BRCA1. RESULTS The 2D DIGE for ABC-11 and ABC-11/CHR2 showed different expression levels in about 2000 spots. SOD was identified from spots highly expressed in resistant strains. Western blotting also confirmed SOD1 overexpression in ABC-11/CHR2. siSOD1 enhanced the growth inhibitory effects of alectinib, increased cleaved PARP levels, and decreased pERK, pAKT, and BRCA1 levels with a combination of alectinib. In addition, the combination of LCS-1, an SOD1 inhibitor, and alectinib synergistically suppressed the growth in ABC-11/CHR2, but not in ABC-11. CONCLUSIONS SOD1 overexpression is thought to be a mechanism for alectinib resistance, suggesting the possibility of overcoming resistance using SOD1 inhibitors.
Collapse
Affiliation(s)
- Noriko Miyake
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan; Kajiki Hospital, Okayama, Japan
| | - Nobuaki Ochi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Masami Takeyama
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Hideko Isozaki
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan; Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Eiki Ichihara
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Japan
| | - Hiromichi Yamane
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Takuya Fukazawa
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan
| | - Yasunari Nagasaki
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Tatsuyuki Kawahara
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Hidekazu Nakanishi
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | | | - Katsuyuki Kiura
- Kajiki Hospital, Okayama, Japan; Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Nagio Takigawa
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan.
| |
Collapse
|
11
|
Wu Y, Ma B, Liu C, Li D, Sui G. Pathological Involvement of Protein Phase Separation and Aggregation in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:10187. [PMID: 39337671 PMCID: PMC11432175 DOI: 10.3390/ijms251810187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Neurodegenerative diseases are the leading cause of human disability and immensely reduce patients' life span and quality. The diseases are characterized by the functional loss of neuronal cells and share several common pathogenic mechanisms involving the malfunction, structural distortion, or aggregation of multiple key regulatory proteins. Cellular phase separation is the formation of biomolecular condensates that regulate numerous biological processes, including neuronal development and synaptic signaling transduction. Aberrant phase separation may cause protein aggregation that is a general phenomenon in the neuronal cells of patients suffering neurodegenerative diseases. In this review, we summarize the pathological causes of common neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease, among others. We discuss the regulation of key amyloidogenic proteins with an emphasis of their aberrant phase separation and aggregation. We also introduce the approaches as potential therapeutic strategies to ameliorate neurodegenerative diseases through intervening protein aggregation. Overall, this review consolidates the research findings of phase separation and aggregation caused by misfolded proteins in a context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yinuo Wu
- Aulin College, Northeast Forestry University, Harbin 150040, China;
| | - Biao Ma
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (B.M.); (C.L.)
| | - Chang Liu
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (B.M.); (C.L.)
| | - Dangdang Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (B.M.); (C.L.)
| | - Guangchao Sui
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (B.M.); (C.L.)
| |
Collapse
|
12
|
Bedja-Iacona L, Richard E, Marouillat S, Brulard C, Alouane T, Beltran S, Andres CR, Blasco H, Corcia P, Veyrat-Durebex C, Vourc’h P. Post-Translational Variants of Major Proteins in Amyotrophic Lateral Sclerosis Provide New Insights into the Pathophysiology of the Disease. Int J Mol Sci 2024; 25:8664. [PMID: 39201350 PMCID: PMC11354932 DOI: 10.3390/ijms25168664] [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: 06/30/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Post-translational modifications (PTMs) affecting proteins during or after their synthesis play a crucial role in their localization and function. The modification of these PTMs under pathophysiological conditions, i.e., their appearance, disappearance, or variation in quantity caused by a pathological environment or a mutation, corresponds to post-translational variants (PTVs). These PTVs can be directly or indirectly involved in the pathophysiology of diseases. Here, we present the PTMs and PTVs of four major amyotrophic lateral sclerosis (ALS) proteins, SOD1, TDP-43, FUS, and TBK1. These modifications involve acetylation, phosphorylation, methylation, ubiquitination, SUMOylation, and enzymatic cleavage. We list the PTM positions known to be mutated in ALS patients and discuss the roles of PTVs in the pathophysiological processes of ALS. In-depth knowledge of the PTMs and PTVs of ALS proteins is needed to better understand their role in the disease. We believe it is also crucial for developing new therapies that may be more effective in ALS.
Collapse
Affiliation(s)
- Léa Bedja-Iacona
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
| | - Elodie Richard
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
| | - Sylviane Marouillat
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
| | | | | | - Stéphane Beltran
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
- Service de Neurologie, CHRU de Tours, 37000 Tours, France
| | - Christian R. Andres
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| | - Hélène Blasco
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| | - Philippe Corcia
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
- Service de Neurologie, CHRU de Tours, 37000 Tours, France
| | - Charlotte Veyrat-Durebex
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
- UTTIL, CHRU de Tours, 37000 Tours, France
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| | - Patrick Vourc’h
- UMR 1253, iBraiN, Université de Tours, Inserm, 37000 Tours, France; lea.bedja-- (L.B.-I.); (E.R.)
- UTTIL, CHRU de Tours, 37000 Tours, France
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| |
Collapse
|
13
|
Duranti E, Villa C. Insights into Dysregulated Neurological Biomarkers in Cancer. Cancers (Basel) 2024; 16:2680. [PMID: 39123408 PMCID: PMC11312413 DOI: 10.3390/cancers16152680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The link between neurodegenerative diseases (NDs) and cancer has generated greater interest in biomedical research, with decades of global studies investigating neurodegenerative biomarkers in cancer to better understand possible connections. Tau, amyloid-β, α-synuclein, SOD1, TDP-43, and other proteins associated with nervous system diseases have also been identified in various types of solid and malignant tumors, suggesting a potential overlap in pathological processes. In this review, we aim to provide an overview of current evidence on the role of these proteins in cancer, specifically examining their effects on cell proliferation, apoptosis, chemoresistance, and tumor progression. Additionally, we discuss the diagnostic and therapeutic implications of this interconnection, emphasizing the importance of further research to completely comprehend the clinical implications of these proteins in tumors. Finally, we explore the challenges and opportunities in targeting these proteins for the development of new targeted anticancer therapies, providing insight into how to integrate knowledge of NDs in oncology research.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
14
|
Hefley BS, Ali AA, Bhattacharya P, Hjortdal J, Walker MK, Karamichos D. Systemic and Ocular Associations of Keratoconus. EXPERT REVIEW OF OPHTHALMOLOGY 2024; 19:379-391. [PMID: 39494085 PMCID: PMC11526800 DOI: 10.1080/17469899.2024.2368801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/12/2024] [Indexed: 11/05/2024]
Abstract
Introduction Keratoconus (KC) is the most prevalent corneal ectasia in the world and its pathogenesis is influenced by both ocular and systemic factors. This review explores the multifaceted associations between keratoconus and systemic health conditions, ocular characteristics, and various other environmental/exogenous factors, aiming to illuminate how these relationships influence the pathophysiology of the disease. Areas Covered This review will summarize the fundamental attributes of KC, review and discuss the systemic and ocular association of KC including molecular biomarkers, and provide an organized overview of the parallel alterations occurring within various biological pathways in KC. Expert Opinion Despite the substantial volume of research on keratoconus, the precise etiology of the disease remains elusive. Further studies are necessary to deepen our understanding of this intricate disorder and improve its management.
Collapse
Affiliation(s)
- Brenna S. Hefley
- North Texas EAye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Arsalan A. Ali
- Anne Burnett Marion School of Medicine, Texas Christian University, Fort Worth, TX, 76107, USA
| | - Pradipta Bhattacharya
- Department of Clinical Sciences, College of Optometry, University of Houston, TX, 77204, USA
- The Ocular Surface Institute, University of Houston College of Optometry, Houston, TX, 77204, USA
| | - Jesper Hjortdal
- Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Maria K. Walker
- Department of Clinical Sciences, College of Optometry, University of Houston, TX, 77204, USA
- The Ocular Surface Institute, University of Houston College of Optometry, Houston, TX, 77204, USA
| | - Dimitrios Karamichos
- North Texas EAye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| |
Collapse
|
15
|
Anjo SI, He Z, Hussain Z, Farooq A, McIntyre A, Laughton CA, Carvalho AN, Finelli MJ. Protein Oxidative Modifications in Neurodegenerative Diseases: From Advances in Detection and Modelling to Their Use as Disease Biomarkers. Antioxidants (Basel) 2024; 13:681. [PMID: 38929122 PMCID: PMC11200609 DOI: 10.3390/antiox13060681] [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: 05/02/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Oxidation-reduction post-translational modifications (redox-PTMs) are chemical alterations to amino acids of proteins. Redox-PTMs participate in the regulation of protein conformation, localization and function, acting as signalling effectors that impact many essential biochemical processes in the cells. Crucially, the dysregulation of redox-PTMs of proteins has been implicated in the pathophysiology of numerous human diseases, including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. This review aims to highlight the current gaps in knowledge in the field of redox-PTMs biology and to explore new methodological advances in proteomics and computational modelling that will pave the way for a better understanding of the role and therapeutic potential of redox-PTMs of proteins in neurodegenerative diseases. Here, we summarize the main types of redox-PTMs of proteins while providing examples of their occurrence in neurodegenerative diseases and an overview of the state-of-the-art methods used for their detection. We explore the potential of novel computational modelling approaches as essential tools to obtain insights into the precise role of redox-PTMs in regulating protein structure and function. We also discuss the complex crosstalk between various PTMs that occur in living cells. Finally, we argue that redox-PTMs of proteins could be used in the future as diagnosis and prognosis biomarkers for neurodegenerative diseases.
Collapse
Affiliation(s)
- Sandra I. Anjo
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-517 Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Zhicheng He
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Zohaib Hussain
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Aruba Farooq
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Alan McIntyre
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Charles A. Laughton
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Andreia Neves Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Mattéa J. Finelli
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
16
|
Huang M, Liu YU, Yao X, Qin D, Su H. Variability in SOD1-associated amyotrophic lateral sclerosis: geographic patterns, clinical heterogeneity, molecular alterations, and therapeutic implications. Transl Neurodegener 2024; 13:28. [PMID: 38811997 PMCID: PMC11138100 DOI: 10.1186/s40035-024-00416-x] [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: 01/29/2024] [Accepted: 04/17/2024] [Indexed: 05/31/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive loss of motor neurons, resulting in global health burden and limited post-diagnosis life expectancy. Although primarily sporadic, familial ALS (fALS) cases suggest a genetic basis. This review focuses on SOD1, the first gene found to be associated with fALS, which has been more recently confirmed by genome sequencing. While informative, databases such as ALSoD and STRENGTH exhibit regional biases. Through a systematic global examination of SOD1 mutations from 1993 to 2023, we found different geographic distributions and clinical presentations. Even though different SOD1 variants are expressed at different protein levels and have different half-lives and dismutase activities, these alterations lead to loss of function that is not consistently correlated with disease severity. Gain of function of toxic aggregates of SOD1 resulting from mutated SOD1 has emerged as one of the key contributors to ALS. Therapeutic interventions specifically targeting toxic gain of function of mutant SOD1, including RNA interference and antibodies, show promise, but a cure remains elusive. This review provides a comprehensive perspective on SOD1-associated ALS and describes molecular features and the complex genetic landscape of SOD1, highlighting its importance in determining diverse clinical manifestations observed in ALS patients and emphasizing the need for personalized therapeutic strategies.
Collapse
Affiliation(s)
- Miaodan Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China
| | - Yong U Liu
- Laboratory for Neuroimmunology in Health and Diseases, Guangzhou First People's Hospital School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaoli Yao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510799, China.
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China.
| |
Collapse
|
17
|
Tsekrekou M, Giannakou M, Papanikolopoulou K, Skretas G. Protein aggregation and therapeutic strategies in SOD1- and TDP-43- linked ALS. Front Mol Biosci 2024; 11:1383453. [PMID: 38855322 PMCID: PMC11157337 DOI: 10.3389/fmolb.2024.1383453] [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: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 06/11/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with severe socio-economic impact. A hallmark of ALS pathology is the presence of aberrant cytoplasmic inclusions composed of misfolded and aggregated proteins, including both wild-type and mutant forms. This review highlights the critical role of misfolded protein species in ALS pathogenesis, particularly focusing on Cu/Zn superoxide dismutase (SOD1) and TAR DNA-binding protein 43 (TDP-43), and emphasizes the urgent need for innovative therapeutic strategies targeting these misfolded proteins directly. Despite significant advancements in understanding ALS mechanisms, the disease remains incurable, with current treatments offering limited clinical benefits. Through a comprehensive analysis, the review focuses on the direct modulation of the misfolded proteins and presents recent discoveries in small molecules and peptides that inhibit SOD1 and TDP-43 aggregation, underscoring their potential as effective treatments to modify disease progression and improve clinical outcomes.
Collapse
Affiliation(s)
- Maria Tsekrekou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Maria Giannakou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Katerina Papanikolopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, Vari, Greece
- ResQ Biotech, Patras Science Park, Rio, Greece
| | - Georgios Skretas
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
- ResQ Biotech, Patras Science Park, Rio, Greece
- Institute for Bio-innovation, Biomedical Sciences Research Centre “Alexander Fleming”, Vari, Greece
| |
Collapse
|
18
|
Castelli L, Vasta R, Allen SP, Waller R, Chiò A, Traynor BJ, Kirby J. From use of omics to systems biology: Identifying therapeutic targets for amyotrophic lateral sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:209-268. [PMID: 38802176 DOI: 10.1016/bs.irn.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a heterogeneous progressive neurodegenerative disorder with available treatments such as riluzole and edaravone extending survival by an average of 3-6 months. The lack of highly effective, widely available therapies reflects the complexity of ALS. Omics technologies, including genomics, transcriptomic and proteomics have contributed to the identification of biological pathways dysregulated and targeted by therapeutic strategies in preclinical and clinical trials. Integrating clinical, environmental and neuroimaging information with omics data and applying a systems biology approach can further improve our understanding of the disease with the potential to stratify patients and provide more personalised medicine. This chapter will review the omics technologies that contribute to a systems biology approach and how these components have assisted in identifying therapeutic targets. Current strategies, including the use of genetic screening and biosampling in clinical trials, as well as the future application of additional technological advances, will also be discussed.
Collapse
Affiliation(s)
- Lydia Castelli
- Sheffield Institute for Translational Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom; Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Rosario Vasta
- ALS Expert Center,'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy; Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | - Scott P Allen
- Sheffield Institute for Translational Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom; Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Rachel Waller
- Sheffield Institute for Translational Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom; Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Adriano Chiò
- ALS Expert Center,'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy; Neurology 1, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza of Turin, Turin, Italy
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States; RNA Therapeutics Laboratory, National Center for Advancing Translational Sciences, NIH, Rockville, MD, United States; National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States; Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, United States; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology,University College London, London, United Kingdom
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom; Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
19
|
Srishti K, Negi O, Hota PK. Recent Development on Copper-Sensor and its Biological Applications: A Review. J Fluoresc 2024:10.1007/s10895-024-03587-y. [PMID: 38416283 DOI: 10.1007/s10895-024-03587-y] [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: 11/29/2023] [Accepted: 01/14/2024] [Indexed: 02/29/2024]
Abstract
Metal ion recognition is one of the most prospective research topics in the field of chemical sensors due to its wide range of clinical, biological and environmental applications. In this context, hydrazones are well known compounds that exhibit metal sensing and several biological properties due to the presence of N=CH- bond. Some of the biological properties includes anti-cancer, anti-tumor, anti-oxidant, anti-microbial activities. Hydrazones are also used as a ligand to detect metal ion as well as to generate metal complexes that exhibit medicinal properties. Thus, in recent years, many attempts were made to develop novel ligands with enhanced metal sensing and medicinal properties. In this review, some of the recent development on the hydrazones and their copper complexes are covered from the last few years from 2015-2023. These includes significance of copper ions, synthesis, biological properties, mechanism and metal sensing properties of some of the copper complexes were discussed.
Collapse
Affiliation(s)
- Km Srishti
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Oseen Negi
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Prasanta Kumar Hota
- Department of Chemistry, School of Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India.
| |
Collapse
|
20
|
Guo Y, Guan T, Yu Q, Sanghai N, Shafiq K, Li M, Jiao X, Na D, Zhang G, Kong J. ALS-linked SOD1 mutations impair mitochondrial-derived vesicle formation and accelerate aging. Redox Biol 2024; 69:102972. [PMID: 38056310 PMCID: PMC10746562 DOI: 10.1016/j.redox.2023.102972] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
Oxidative stress (OS) is regarded as the dominant theory for aging. While compelling correlative data have been generated to support the OS theory, a direct cause-and-effect relationship between the accumulation of oxidation-mediated damage and aging has not been firmly established. Superoxide dismutase 1 (SOD1) is a primary antioxidant in all cells. It is, however, susceptible to oxidation due to OS and gains toxic properties to cells. This study investigates the role of oxidized SOD1 derived from amyotrophic lateral sclerosis (ALS) linked SOD1 mutations in cell senescence and aging. Herein, we have shown that the cell line NSC34 expressing the G93A mutation of human SOD1 (hSOD1G93A) entered premature senescence as evidenced by a decreased number of the 5-ethynyl-2'-deoxyuridine (EdU)-positive cells. There was an upregulation of cellular senescence markers compared to cells expressing the wild-type human SOD1 (hSOD1WT). Transgenic mice carrying the hSOD1G93A gene showed aging phenotypes at an early age (135 days) with high levels of P53 and P16 but low levels of SIRT1 and SIRT6 compared with age-matched hSOD1WT transgenic mice. Notably, the levels of oxidized SOD1 were significantly elevated in both the senescent NSC34 cells and 135-day hSOD1G93A mice. Selective removal of oxidized SOD1 by our CT4-directed autophagy significantly decelerated aging, indicating that oxidized SOD1 is a causal factor of aging. Intriguingly, mitochondria malfunctioned in both senescent NSC34 cells and middle-aged hSODG93A transgenic mice. They exhibited increased production of mitochondrial-derived vesicles (MDVs) in response to mild OS in mutant humanSOD1 (hSOD1) transgenic mice at a younger age; however, the mitochondrial response gradually declined with aging. In conclusion, our data show that oxidized SOD1 derived from ALS-linked SOD1 mutants is a causal factor for cellular senescence and aging. Compromised mitochondrial responsiveness to OS may serve as an indicator of premature aging.
Collapse
Affiliation(s)
- Ying Guo
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada; Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Qiang Yu
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Nitesh Sanghai
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Canada
| | - Kashfia Shafiq
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Meiyu Li
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Xin Jiao
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Donghui Na
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China.
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada.
| |
Collapse
|
21
|
Zhou D, Zhang Y, Zhu M, Zhang X, Zhang X, Lv J, Tang W, Weng Q, Lin Y, Tong L, Zhong Z, Zhang Y, Zhang M, Lai M, Wang D. mROS‑calcium feedback loop promotes lethal ventricular arrhythmias and sudden cardiac death in early myocardial ischemia. Int J Mol Med 2024; 53:5. [PMID: 37997788 PMCID: PMC10712693 DOI: 10.3892/ijmm.2023.5329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Lethal ventricular arrhythmia‑sudden cardiac death (LVA‑SCD) occurs frequently during the early stage of myocardial ischemia (MI). However, the mechanism underlying higher LVA‑SCD incidence is still poorly understood. The present study aimed to explore the role of mitochondrial reactive oxygen species (mROS) and Ca2+ crosstalk in promoting LVA‑SCD in early MI. RyR2 S2814A mice and their wild‑type littermates were used. MitoTEMPO was applied to scavenge mitochondrial ROS (mROS). Mice were subjected to severe MI and the occurrence of LVA‑SCD was evaluated. Levels of mitochondrial ROS and calcium (mitoCa2+), cytosolic ROS (cytoROS), and calcium (cytoCa2+), RyR2 Ser‑2814 phosphorylation, CaMKII Met‑282 oxidation, mitochondrial membrane potential (MMP), and glutathione/oxidized glutathione (GSH/GSSG) ratio in the myocardia were detected. Dynamic changes in mROS after hypoxia were investigated using H9c2 cells. Moreover, the myocardial phosphoproteome was analyzed to explore the related mechanisms facilitating mROS‑Ca2+ crosstalk and LVA‑SCD. There was a high incidence (~33.9%) of LVA‑SCD in early MI. Mice who underwent SCD displayed notably elevated levels of myocardial ROS and mROS, and the latter was validated in H9c2 cells. These mice also demonstrated overloads of cytoplasmic and mitochondrial Ca2+, decreased MMP and reduced GSH/GSSG ratio, upregulated RyR2‑S2814 phosphorylation and CaMKII‑M282 oxidation and transient hyperphosphorylation of mitochondrial proteomes in the myocardium. mROS‑specific scavenging by a mitochondria‑targeted antioxidant agent (MitoTEMPO) corrected these SCD‑induced alterations. S2814A mice with a genetically inactivated CaMKII phosphorylation site in RyR2 exhibited decreased overloads in cytoplasmic and mitochondrial Ca2+ and demonstrated similar effects as MitoTEMPO to correct SCD‑induced changes and prevent SCD post‑MI. The data confirmed crosstalk between mROS and Ca2+ in promoting LVA‑SCD. Therefore, we provided evidence that there is a higher incidence of LVA‑SCD in early MI, which may be attributed to a positive feedback loop between mROS and Ca2+ imbalance.
Collapse
Affiliation(s)
- Danya Zhou
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- School of Forensic Medicine, Xinxiang Key Laboratory of Forensic Toxicology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Ye Zhang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Mengting Zhu
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiaojun Zhang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Xiaojuan Zhang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Junyao Lv
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Wanting Tang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Qi Weng
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yang Lin
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Lejun Tong
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Zhiwei Zhong
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yanmei Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Mengxuan Zhang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Minchao Lai
- Department of Neurology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Dian Wang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
22
|
Liao Y, Wu C, Li Y, Wen J, Zhao D. MIF is a critical regulator of mononuclear phagocytic infiltration in hepatocellular carcinoma. iScience 2023; 26:107273. [PMID: 37520719 PMCID: PMC10371853 DOI: 10.1016/j.isci.2023.107273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/03/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Immunotherapy targeting tumor-associated macrophages (TAMs) is a promising approach to treating cancer. However, the limited drug targets and ambiguous mechanisms impede the development of clinical immunotherapy strategies. To elucidate the underlying processes involved in mononuclear phagocyte (MNP) infiltration and phenotypic changes in hepatocellular carcinoma (HCC), we integrated single-cell RNA-sequencing data from 100,030 cells derived from patients with HCC and healthy individuals and compared the phenotypes and origins of the MNPs in the tumor core, tumor periphery, adjacent normal tissue, and healthy liver samples. Using machine learning and multi-omics analyses, we identified 445 infiltration-associated genes and potential drug targets affecting this process. Through in vitro experiments, we found that the expression of macrophage migration inhibitory factor (MIF) is the upstream regulator of secreted phosphoprotein 1 (SPP1) and promote migration in TAMs. Our findings also indicate that MIF promotes tumor metastasis and invasion and is a promising potential target for treating HCC.
Collapse
Affiliation(s)
- Yunxi Liao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Chenyang Wu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yang Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jinhua Wen
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Dongyu Zhao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| |
Collapse
|
23
|
Yamamoto Y, Kondo M, Hotta Y, Tashiro Y, Sanagawa A, Kataoka T, Furukawa-Hibi Y, Ri M, Komatsu H, Iida S, Kimura K. The Relationship between Changes in Serum Element Concentrations and Pathological Condition and Disease Status in Japanese Multiple Myeloma Patients: A Pilot Study and Literature Review. Asian Pac J Cancer Prev 2023; 24:2493-2503. [PMID: 37505784 PMCID: PMC10676486 DOI: 10.31557/apjcp.2023.24.7.2493] [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: 04/12/2023] [Accepted: 07/03/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a rare cancer, and information on its pathological condition and serum element levels is lacking. In this pilot study, we examined serum element concentrations in Japanese patients with MM by a comprehensive multi-element analysis. METHODS This is a case-control study of 12 Japanese patients diagnosed with MM at the Nagoya City University Hospital between 2008 and 2013. Blood samples were taken, at the initial diagnosis and at relapse. The serum concentrations of 12 elements were analyzed by inductively coupled plasma mass spectrometry and compared between MM patients and non-MM volunteers. We also analyzed the correlation between serum element concentrations and laboratory values related to disease status and tumor volume of MM. RESULTS We found that serum chromium (Cr), copper (Cu), molybdenum (Mo), and barium (Ba) concentrations were significantly increased in MM patients. Ba was significantly increased in MM patients, suggesting an association with bone lesions. There was no consistent trend between these elements and existing indices related to MM tumor volume and disease status. CONCLUSIONS Although this is a pilot study, serum Cr, Cu, Mo, and Ba concentrations were found to be significantly elevated in MM patients. Further studies with large sample sizes are needed, since the changes in serum concentrations of these elements may reflect the pathological condition of MM.
Collapse
Affiliation(s)
- Yuko Yamamoto
- Department of Hospital Pharmacy, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe do-ri, Mizuho-ku, Nagoya 467-8603, Japan.
- Department of Analytical Chemistry, Aichi Prefectural Institute of Public Health, 7-6 Nagare, Tsuji-machi, Kita-ku, Nagoya 462-8576, Japan.
| | - Masahiro Kondo
- Department of Pharmacy, Nagoya City University Hospital East Medical Center, 1-2-23 Wakamizu, Chikusa-ku, Nagoya 464-8547, Japan.
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Yuji Hotta
- Department of Hospital Pharmacy, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe do-ri, Mizuho-ku, Nagoya 467-8603, Japan.
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
- Department of Pharmacy, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Yuusuke Tashiro
- Department of Pharmacy, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Akimasa Sanagawa
- Department of Hospital Pharmacy, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe do-ri, Mizuho-ku, Nagoya 467-8603, Japan.
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
- Department of Pharmacy, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Tomoya Kataoka
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences Chiba Institute of Science, 15-8 Shiomi-cho, Choshi 288-0025, Japan.
| | - Yoko Furukawa-Hibi
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
- Department of Pharmacy, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Hirokazu Komatsu
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Kazunori Kimura
- Department of Hospital Pharmacy, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe do-ri, Mizuho-ku, Nagoya 467-8603, Japan.
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
- Department of Pharmacy, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| |
Collapse
|
24
|
Simmers MD, Jima DD, Tsuji Y, Cowley M. LncRNA Tuna is activated in cadmium-induced placental insufficiency and drives the NRF2-mediated oxidative stress response. Front Cell Dev Biol 2023; 11:1151108. [PMID: 37325564 PMCID: PMC10267411 DOI: 10.3389/fcell.2023.1151108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/24/2023] [Indexed: 06/17/2023] Open
Abstract
Cadmium (Cd) is a toxic heavy metal found throughout the environment and one of the top ten toxicants of major public health concern identified by the World Health Organization. In utero Cd exposure causes fetal growth restriction, malformation, and spontaneous abortion; however, the mechanisms by which Cd impacts these outcomes are poorly understood. Cd accumulates in the placenta, suggesting that these negative outcomes may be a consequence of disrupted placental function and placental insufficiency. To understand the impact of Cd on gene expression within the placenta, we developed a mouse model of Cd-induced fetal growth restriction through maternal consumption of CdCl2 and performed RNA-seq on control and CdCl2 exposed placentae. The top differentially expressed transcript was the Tcl1 Upstream Neuron-Associated (Tuna) long non-coding RNA, which was upregulated over 25-fold in CdCl2 exposed placentae. Tuna has been shown to be critical for neural stem cell differentiation. However, within the placenta, there is no evidence that Tuna is normally expressed or functional at any developmental stage. To determine the spatial expression of Cd-activated Tuna within the placenta, we used in situ hybridization as well as placental layer-specific RNA isolation and analysis. Both methods confirmed the absence of Tuna expression in control samples and determined that Cd-induced Tuna expression is specific to the junctional zone. Since many lncRNAs regulate gene expression, we hypothesized that Tuna forms part of the mechanism of Cd-induced transcriptomic changes. To test this, we over-expressed Tuna in cultured choriocarcinoma cells and compared gene expression profiles to those of control and CdCl2 exposed cells. We demonstrate significant overlap between genes activated by Tuna overexpression and genes activated by CdCl2 exposure, with enrichment in the NRF2-mediated oxidative stress response. Herein we analyze the NRF2 pathway and show that Tuna increases NRF2/NRF2 both at the transcript and protein levels. Tuna drives increased NRF2 target gene expression, a result that is abrogated with the use of an NRF2 inhibitor, confirming that Tuna activates oxidative stress response genes through this pathway. This work identifies the lncRNA Tuna as a potential novel player in Cd-induced placental insufficiency.
Collapse
Affiliation(s)
- Mark D. Simmers
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Dereje D. Jima
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States
| | - Yoshiaki Tsuji
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Michael Cowley
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
25
|
Tzeplaeff L, Wilfling S, Requardt MV, Herdick M. Current State and Future Directions in the Therapy of ALS. Cells 2023; 12:1523. [PMID: 37296644 PMCID: PMC10252394 DOI: 10.3390/cells12111523] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder affecting upper and lower motor neurons, with death resulting mainly from respiratory failure three to five years after symptom onset. As the exact underlying causative pathological pathway is unclear and potentially diverse, finding a suitable therapy to slow down or possibly stop disease progression remains challenging. Varying by country Riluzole, Edaravone, and Sodium phenylbutyrate/Taurursodiol are the only drugs currently approved in ALS treatment for their moderate effect on disease progression. Even though curative treatment options, able to prevent or stop disease progression, are still unknown, recent breakthroughs, especially in the field of targeting genetic disease forms, raise hope for improved care and therapy for ALS patients. In this review, we aim to summarize the current state of ALS therapy, including medication as well as supportive therapy, and discuss the ongoing developments and prospects in the field. Furthermore, we highlight the rationale behind the intense research on biomarkers and genetic testing as a feasible way to improve the classification of ALS patients towards personalized medicine.
Collapse
Affiliation(s)
- Laura Tzeplaeff
- Department of Neurology, Rechts der Isar Hospital, Technical University of Munich, 81675 München, Germany
| | - Sibylle Wilfling
- Department of Neurology, University of Regensburg, 93053 Regensburg, Germany;
- Center for Human Genetics Regensburg, 93059 Regensburg, Germany
| | - Maria Viktoria Requardt
- Formerly: Department of Neurology with Institute of Translational Neurology, Münster University Hospital (UKM), 48149 Münster, Germany;
| | - Meret Herdick
- Precision Neurology, University of Lübeck, 23562 Luebeck, Germany
| |
Collapse
|
26
|
Minjares M, Wu W, Wang JM. Oxidative Stress and MicroRNAs in Endothelial Cells under Metabolic Disorders. Cells 2023; 12:1341. [PMID: 37174741 PMCID: PMC10177439 DOI: 10.3390/cells12091341] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
Reactive oxygen species (ROS) are radical oxygen intermediates that serve as important second messengers in signal transduction. However, when the accumulation of these molecules exceeds the buffering capacity of antioxidant enzymes, oxidative stress and endothelial cell (EC) dysfunction occur. EC dysfunction shifts the vascular system into a pro-coagulative, proinflammatory state, thereby increasing the risk of developing cardiovascular (CV) diseases and metabolic disorders. Studies have turned to the investigation of microRNA treatment for CV risk factors, as these post-transcription regulators are known to co-regulate ROS. In this review, we will discuss ROS pathways and generation, normal endothelial cell physiology and ROS-induced dysfunction, and the current knowledge of common metabolic disorders and their connection to oxidative stress. Therapeutic strategies based on microRNAs in response to oxidative stress and microRNA's regulatory roles in controlling ROS will also be explored. It is important to gain an in-depth comprehension of the mechanisms generating ROS and how manipulating these enzymatic byproducts can protect endothelial cell function from oxidative stress and prevent the development of vascular disorders.
Collapse
Affiliation(s)
- Morgan Minjares
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
| | - Wendy Wu
- Vera P Shiffman Medical Library, Wayne State University, 320 E Canfield St., Detroit, MI 48201, USA;
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
- Center for Molecular Medicine and Genetics, Wayne State University, 320 E Canfield St., Detroit, MI 48201, USA
- Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI 48201, USA
| |
Collapse
|
27
|
Shi W, Ling L, Li C, Wu R, Zhang M, Shao F, Wang Y. LncRNA UCA1 promoted cisplatin resistance in lung adenocarcinoma with HO1 targets NRF2/HO1 pathway. J Cancer Res Clin Oncol 2023; 149:1295-1311. [PMID: 35792913 DOI: 10.1007/s00432-022-04152-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/13/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE Our previous experiments have demonstrated that lncRNA UCA1 (UCA1) promoted cisplatin resistance in lung adenocarcinoma (LUAD). This study aimed to explore the potential downstream target genes regulated by UCA1 and how this downstream gene promotes cisplatin resistance in LUAD. METHODS Here, we measured the expression level of Heme oxygenase1 (HO1) in LUAD cell lines by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) based on UCA1 overexpression cell lines and UCA1 knockdown cell lines. HO1 was knocked down in the UCA1 overexpression cell line, and HO1 was overexpressed in the UCA1 knockdown cell line, and the half maximal inhibitory concentration (IC50) trends were observed by adding cisplatin containing a certain concentration gradient. Cell functional assays were performed to observe the changes in the biological behavior of HO1 after overexpression and knockdown, and the tumorigenic assay in nude mice was performed to verify the effect of UCA1 in regulating the growth and cisplatin resistance of HO1 on LUAD cells in vivo. RESULTS The results showed that HO1 and UCA1 expression were both upregulated in LUAD tissues and LUAD cisplatin-resistant cell lines, and there was a significant positive correlation between the expression of HO1 and UCA1. In vitro experiments showed that HO1 overexpression could reverse the reduced sensitivity to cisplatin caused by UCA1 knockdown in A549/DDP cells, and HO1 knockdown could reduce cisplatin resistance in A549 UCA1 overexpressing cells. Tumorigenic assays in nude mice further confirmed the role of HO1 in the regulation of UCA1 by activating the NRF2/HO1 pathway against LUAD cisplatin resistance. CONCLUSION Our findings suggested that UCA1 regulates HO1 targets the UCA1/NRF2-HO1 pathway to exert cisplatin resistance in LUAD.
Collapse
Affiliation(s)
- Wenjing Shi
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translation Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Liqun Ling
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translation Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Changhong Li
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translation Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Ruihao Wu
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translation Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Meijuan Zhang
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translation Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Fanggui Shao
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translation Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China
| | - Yumin Wang
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Diagnosis and Translation Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
28
|
Human Pluripotent Stem Cell-Mesenchymal Stem Cell-Derived Exosomes Promote Ovarian Granulosa Cell Proliferation and Attenuate Cell Apoptosis Induced by Cyclophosphamide in a POI-like Mouse Model. Molecules 2023; 28:molecules28052112. [PMID: 36903358 PMCID: PMC10004137 DOI: 10.3390/molecules28052112] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
Premature ovarian insufficiency (POI) is a complex disease which causes amenorrhea, hypergonadotropism and infertility in patients no more than 40 years old. Recently, several studies have reported that exosomes have the potential to protect ovarian function using a POI-like mouse model induced by chemotherapy drugs. In this study, the therapeutic potential of exosomes derived from human pluripotent stem cell-mesenchymal stem cells (hiMSC exosomes) was evaluated through a cyclophosphamide (CTX)-induced POI-like mouse model. POI-like pathological changes in mice were determined by serum sex-hormones levels and the available number of ovarian follicles. The expression levels of cellular proliferation proteins and apoptosis-related proteins in mouse ovarian granulosa cells were measured using immunofluorescence, immunohistochemistry and Western blotting. Notably, a positive effect on the preservation of ovarian function was evidenced, since the loss of follicles in the POI-like mouse ovaries was slowed. Additionally, hiMSC exosomes not only restored the levels of serum sex hormones, but also significantly promoted the proliferation of granulosa cells and inhibited cell apoptosis. The current study suggests that the administration of hiMSC exosomes in the ovaries can preserve female-mouse fertility.
Collapse
|
29
|
Fehlberg LK, Guadagnin AR, Thomas BL, Ballou M, Loor JJ, Sugimoto Y, Shinzato I, Cardoso FC. Feeding rumen-protected lysine altered immune and metabolic biomarkers in dairy cows during the transition period. J Dairy Sci 2023; 106:2989-3007. [PMID: 36797190 DOI: 10.3168/jds.2022-22349] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/13/2022] [Indexed: 02/16/2023]
Abstract
This experiment was conducted to determine the effects of feeding rumen-protected lysine (RPL; AjiPro-L Generation 3, Ajinomoto Health and Nutrition North America Inc.) from -26 ± 4.6 d prepartum (0.54% RPL of dietary dry matter intake) to 28 d postpartum (0.39% RPL of dietary dry matter intake) on immunometabolic status and liver composition in dairy cows. Seventy-five multiparous Holstein cows, blocked by parity, previous 305-d mature-equivalent milk production, expected calving date, and body condition score during the far-off dry period were assigned to 1 of 4 dietary treatments in a randomized, complete block design with a 2 × 2 factorial arrangement of treatments. Treatments prepartum consisted of total mixed ration top dressed with RPL (PRE-L) or without RPL (PRE-C), and postpartum treatments consisted of total mixed ration top dressed PRE-L prepartum and postpartum, PRE-L prepartum and PRE-C postpartum, PRE-C prepartum and PRE-L postpartum, and PRE-C prepartum and postpartum in 300 g of molasses. Blood samples were taken on -7 ± 0.5, 0 ± 0.5, 7 ± 0.9, 14 ± 0.9, and 28 ± 0.5 d relative to calving. Whole blood samples were taken on -14 ± 0.5, -7 ± 0.5, 7 ± 0.9, and 14 ± 0.9 d relative to calving for oxidative burst and phagocytic capacity of monocytes and neutrophils. Liver samples were collected via a biopsy on -12 ± 4.95 and 13 ± 2.62 d relative to calving and analyzed for liver composition (triacylglyceride and carnitine concentrations), mRNA expression of hepatic genes, and protein abundance. Protein abundance was calculated by normalizing intensity bands for a specific protein with glyceraldehyde-3-phosphate dehydrogenase. Concentrations of haptoglobin and glutathione peroxidase activity in plasma were lower at d 0 for cows in PRE-L (102 µg/mL and 339 nmol/min per mL, respectively) compared with cows in PRE-C (165 µg/mL and 405 nmol/min per mL, respectively). Oxidative burst capacity in monocytes tended to be greater on d 7 postpartum for cows in PRE-L (65.6%) than cows in PRE-C (57.5%). Additionally, feeding RPL altered the mRNA expression in liver tissue prepartum [decreased INSR (insulin receptor), CPT1A (carnitine palmitoyltransferase 1A), and IL1B (interleukin 1 β)] and postpartum [increased IL8 (interleukin 8), EHMT2 (euchromatic histone lysine methyltransferase 2), TSPO (translocator protein), and SLC3A2 (solute carrier family 3 member 2); and decreased SLC7A1 (solute carrier family 7 member 1), SOD1 (superoxide dismutase 1), and SAA3 (serum amyloid A 3)] compared with cows not consuming RPL]. Additionally, cows in the PRE-C prepartum and PRE-L postpartum treatment tended to have greater protein abundance of mTOR postpartum compared with the PRE-C prepartum and postpartum treatment. Protein abundance of SLC7A7 (solute carrier family 7 member 7) pre- and postpartum tended to be greater and BBOX1 (gamma-butyrobetaine dioxygenase 1) tended to be less when RPL was consumed prepartum. In conclusion, cows that consumed RPL during the transition period had molecular changes related to liver composition, enhanced liver function indicated by greater total protein and albumin concentrations in plasma, and improved immune status indicated by decreased haptoglobin, glutathione peroxidase activity, and immune related mRNA expression.
Collapse
Affiliation(s)
- L K Fehlberg
- Department of Animal Sciences, University of Illinois, Urbana 61801
| | - A R Guadagnin
- Department of Animal Sciences, University of Illinois, Urbana 61801
| | - B L Thomas
- Department of Animal Sciences, University of Illinois, Urbana 61801
| | - M Ballou
- Department of Animal Science and Food Science, Texas Tech University, Lubbock 79409
| | - J J Loor
- Department of Animal Sciences, University of Illinois, Urbana 61801
| | - Y Sugimoto
- Ajinomoto Co. Inc., Tokyo 104-8315, Japan
| | - I Shinzato
- Ajinomoto Co. Inc., Tokyo 104-8315, Japan
| | - F C Cardoso
- Department of Animal Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
30
|
Li H, Yuan L, Yang H, Guo Y, Zheng W, Fan K, Deng S, Gong L, Xu H, Yang Z, Cheng J, Kang M, Deng H. Analysis of SOD1 Variants in Chinese Patients with Familial Amyotrophic Lateral Sclerosis. QJM 2023; 116:365-374. [PMID: 36661322 DOI: 10.1093/qjmed/hcad010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease, and genetic contributors exert a significant role in the complicated pathogenesis. Identification of the genetic causes in ALS families could be valuable for early diagnosis and management. The development of potential drugs for patients with genetic defects will shed new light on ALS therapy. AIM To identify causative variants in three Chinese families with familial ALS (FALS), reveal the pathogenic mechanism, and look for the targeted drug for ALS. DESIGN AND METHODS Whole-exome sequencing and bioinformatics were used to perform genetic analysis of the ALS families. Functional analysis was performed to study the variants' function and search for potential drug targets. RESULTS Three heterozygous missense variants of the SOD1 gene were identified in families with FALS. The clinical manifestations of these patients include spinal onset, predominant lower motor neurons presentation, and absence of cognitive involvement. Functional analysis showed that all three SOD1 variants led to increased reactive oxygen species (ROS) levels, reduced cell viability, and formation of cytoplasmic aggregates. Remarkably, the decreased cell viability induced by variants was rescued after treatment with the ROS inhibitor N-acetylcysteine. CONCLUSIONS This study identified three SOD1 variants in three families with FALS. The variant SOD1 toxicity was associated with oxidative damage and aggregation, and N-acetylcysteine could rescue the decreased cell viability induced by these variants. Our findings support a pathogenic role for ROS in SOD1 deficiencies, and provide a potential drug N-acetylcysteine for ALS therapy, especially in SOD1-patients with limb onset.
Collapse
Affiliation(s)
- H Li
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, China
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - L Yuan
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
- Disease Genome Research Center, Central South University, Changsha, China
| | - H Yang
- Department of Neurology, the Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Y Guo
- Department of Medical Information, School of Life Sciences, Central South University, Changsha, China
| | - W Zheng
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - K Fan
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, China
| | - S Deng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - L Gong
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - H Xu
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Z Yang
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - J Cheng
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - M Kang
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - H Deng
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, China
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, the Third Xiangya Hospital, Central South University, Changsha, China
- Disease Genome Research Center, Central South University, Changsha, China
| |
Collapse
|
31
|
Kabir F, Atkinson R, Cook AL, Phipps AJ, King AE. The role of altered protein acetylation in neurodegenerative disease. Front Aging Neurosci 2023; 14:1025473. [PMID: 36688174 PMCID: PMC9845957 DOI: 10.3389/fnagi.2022.1025473] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/03/2022] [Indexed: 01/06/2023] Open
Abstract
Acetylation is a key post-translational modification (PTM) involved in the regulation of both histone and non-histone proteins. It controls cellular processes such as DNA transcription, RNA modifications, proteostasis, aging, autophagy, regulation of cytoskeletal structures, and metabolism. Acetylation is essential to maintain neuronal plasticity and therefore essential for memory and learning. Homeostasis of acetylation is maintained through the activities of histone acetyltransferases (HAT) and histone deacetylase (HDAC) enzymes, with alterations to these tightly regulated processes reported in several neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). Both hyperacetylation and hypoacetylation can impair neuronal physiological homeostasis and increase the accumulation of pathophysiological proteins such as tau, α-synuclein, and Huntingtin protein implicated in AD, PD, and HD, respectively. Additionally, dysregulation of acetylation is linked to impaired axonal transport, a key pathological mechanism in ALS. This review article will discuss the physiological roles of protein acetylation and examine the current literature that describes altered protein acetylation in neurodegenerative disorders.
Collapse
|
32
|
Camponeschi F, Banci L. Metal trafficking in the cell: Combining atomic resolution with cellular dimension. FEBS Lett 2023; 597:122-133. [PMID: 36285633 DOI: 10.1002/1873-3468.14524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 01/14/2023]
Abstract
Metals are widely present in biological systems as simple ions or complex cofactors, and are involved in a variety of processes essential for life. Their transport inside cells and insertion into the binding sites of the proteins that need metals to function occur through complex and selective pathways involving dedicated multiprotein machineries specifically and transiently interacting with each other, often sharing the coordination of metal ions and/or cofactors. The understanding of these machineries requires integrated approaches, ranging from bioinformatics to experimental investigations, possibly in the cellular context. In this review, we report two case studies where the use of integrated in vitro and in cellulo approaches is necessary to clarify at atomic resolution essential aspects of metal trafficking in cells.
Collapse
Affiliation(s)
- Francesca Camponeschi
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy
| | - Lucia Banci
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy.,Department of Chemistry, University of Florence, Italy
| |
Collapse
|
33
|
Melicher P, Dvořák P, Šamaj J, Takáč T. Protein-protein interactions in plant antioxidant defense. FRONTIERS IN PLANT SCIENCE 2022; 13:1035573. [PMID: 36589041 PMCID: PMC9795235 DOI: 10.3389/fpls.2022.1035573] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
The regulation of reactive oxygen species (ROS) levels in plants is ensured by mechanisms preventing their over accumulation, and by diverse antioxidants, including enzymes and nonenzymatic compounds. These are affected by redox conditions, posttranslational modifications, transcriptional and posttranscriptional modifications, Ca2+, nitric oxide (NO) and mitogen-activated protein kinase signaling pathways. Recent knowledge about protein-protein interactions (PPIs) of antioxidant enzymes advanced during last decade. The best-known examples are interactions mediated by redox buffering proteins such as thioredoxins and glutaredoxins. This review summarizes interactions of major antioxidant enzymes with regulatory and signaling proteins and their diverse functions. Such interactions are important for stability, degradation and activation of interacting partners. Moreover, PPIs of antioxidant enzymes may connect diverse metabolic processes with ROS scavenging. Proteins like receptor for activated C kinase 1 may ensure coordination of antioxidant enzymes to ensure efficient ROS regulation. Nevertheless, PPIs in antioxidant defense are understudied, and intensive research is required to define their role in complex regulation of ROS scavenging.
Collapse
|
34
|
Tsymbal S, Li G, Agadzhanian N, Sun Y, Zhang J, Dukhinova M, Fedorov V, Shevtsov M. Recent Advances in Copper-Based Organic Complexes and Nanoparticles for Tumor Theranostics. Molecules 2022; 27:7066. [PMID: 36296659 PMCID: PMC9611640 DOI: 10.3390/molecules27207066] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 08/19/2023] Open
Abstract
Treatment of drug-resistant forms of cancer requires consideration of their hallmark features, such as abnormal cell death mechanisms or mutations in drug-responding molecular pathways. Malignant cells differ from their normal counterparts in numerous aspects, including copper metabolism. Intracellular copper levels are elevated in various cancer types, and this phenomenon could be employed for the development of novel oncotherapeutic approaches. Copper maintains the cell oxidation levels, regulates the protein activity and metabolism, and is involved in inflammation. Various copper-based compounds, such as nanoparticles or metal-based organic complexes, show specific activity against cancer cells according to preclinical studies. Herein, we summarize the major principles of copper metabolism in cancer cells and its potential in cancer theranostics.
Collapse
Affiliation(s)
- Sergey Tsymbal
- International Institute of Solution Chemistry of Advanced Materials and Technologies, ITMO University, 197101 Saint Petersburg, Russia
| | - Ge Li
- Cancer Center & Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, 2000 Xiang’an Road East, Xiamen 361101, China
- Xiamen Key Laboratory for Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, Xiamen 361101, China
| | - Nikol Agadzhanian
- International Institute of Solution Chemistry of Advanced Materials and Technologies, ITMO University, 197101 Saint Petersburg, Russia
| | - Yuhao Sun
- Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jiazhennan Zhang
- Day-Care Department, Xinjiang Medical University, Urumqi 830011, China
| | - Marina Dukhinova
- International Institute of Solution Chemistry of Advanced Materials and Technologies, ITMO University, 197101 Saint Petersburg, Russia
| | - Viacheslav Fedorov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
35
|
Dashnaw CM, Zhang AY, Gonzalez M, Koone JC, Shaw BF. Metal migration and subunit swapping in ALS-linked SOD1: Zn 2+ transfer between mutant and wild-type occurs faster than the rate of heterodimerization. J Biol Chem 2022; 298:102610. [PMID: 36265587 PMCID: PMC9667317 DOI: 10.1016/j.jbc.2022.102610] [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: 08/01/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
The heterodimerization of WT Cu, Zn superoxide dismutase-1 (SOD1), and mutant SOD1 might be a critical step in the pathogenesis of SOD1-linked amyotrophic lateral sclerosis (ALS). Rates and free energies of heterodimerization (ΔGHet) between WT and ALS-mutant SOD1 in mismatched metalation states-where one subunit is metalated and the other is not-have been difficult to obtain. Consequently, the hypothesis that under-metalated SOD1 might trigger misfolding of metalated SOD1 by "stealing" metal ions remains untested. This study used capillary zone electrophoresis and mass spectrometry to track heterodimerization and metal transfer between WT SOD1, ALS-variant SOD1 (E100K, E100G, D90A), and triply deamidated SOD1 (modeled with N26D/N131D/N139D substitutions). We determined that rates of subunit exchange between apo dimers and metalated dimers-expressed as time to reach 30% heterodimer-ranged from t30% = 67.75 ± 9.08 to 338.53 ± 26.95 min; free energies of heterodimerization ranged from ΔGHet = -1.21 ± 0.31 to -3.06 ± 0.12 kJ/mol. Rates and ΔGHet values of partially metalated heterodimers were more similar to those of fully metalated heterodimers than apo heterodimers, and largely independent of which subunit (mutant or WT) was metal-replete or metal-free. Mass spectrometry and capillary electrophoresis demonstrated that mutant or WT 4Zn-SOD1 could transfer up to two equivalents of Zn2+ to mutant or WT apo-SOD1 (at rates faster than the rate of heterodimerization). This result suggests that zinc-replete SOD1 can function as a chaperone to deliver Zn2+ to apo-SOD1, and that WT apo-SOD1 might increase the toxicity of mutant SOD1 by stealing its Zn2+.
Collapse
|
36
|
ROS-Mediated Enamel Formation Disturbance Characterized by Alternative Cervical Loop Cell Proliferation and Downregulation of RhoA/ROCK in Ameloblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5769679. [DOI: 10.1155/2022/5769679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022]
Abstract
Reactive oxygen stress (ROS) is generally accepted as a signal transducer for coordinating the growth and differentiation of tissues and organs in the oral and maxillofacial region. Although ROS has been confirmed to affect the development of enamel, it is not yet known that the specific mechanism of ROS accumulation induced enamel defects. Given the lack of knowledge of the role of ROS in enamel, the aim of the study is to determine how oxidative stress affects cervical cells and ameloblast cells. Using SOD1 knockout mice, we identified a relationship between ROS fluctuations and abnormal enamel structure with HE staining, micro-CT, and scanning electron microscope. Increased ROS induced by H2O2, certified by the DCFH probe, has resulted in a dual effect on the proliferation and differentiation of cervical cells, indicating a higher tendency to proliferate at low ROS concentrations. Ameloblasts transfected with SOD1 siRNA showed a significant reduction of RhoA and ROCK. This study investigates for the first time that SOD1-mediated ROS accumulation disrupted normal enamel structure through alternative cervical loop cell proliferation and downregulation of RhoA and ROCK in ameloblasts, demonstrating the convoluted role of ROS in monitoring the progress of enamel defects.
Collapse
|
37
|
Tasić D, Opačić M, Kovačević S, Nikolić Kokić A, Dimitrijević M, Nikolić D, Vojnović Milutinović D, Blagojević D, Djordjevic A, Brkljačić J. Effects of Fructose and Stress on Rat Renal Copper Metabolism and Antioxidant Enzymes Function. Int J Mol Sci 2022; 23:ijms23169023. [PMID: 36012287 PMCID: PMC9409054 DOI: 10.3390/ijms23169023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 12/22/2022] Open
Abstract
The effects of a fructose-rich diet and chronic stress on copper metabolism in the kidneys are still understudied. We investigated whether fructose and/or chronic unpredictable stress modulate copper metabolism in a way that affects redox homeostasis, thus contributing to progression of metabolic disturbances in the kidney. We determined protein level of copper transporters, chaperones, and cuproenzymes including cytochrome c oxidase, as well as antioxidant enzymes function in the kidneys of male Wistar rats subjected to 20% liquid fructose supplementation and/or chronic stress. Liquid fructose supplementation increased level of copper chaperone of superoxide dismutase and decreased metallothionein level, while rendering the level of copper importer and copper chaperones involved in copper delivery to mitochondria and trans Golgi network unaffected. Stress had no effect on renal copper metabolism. The activity and expression of renal antioxidant enzymes remained unaltered in all experimental groups. In conclusion, fructose, independently of stress, decreased renal copper level, and modulated renal copper metabolism as to preserve vital cellular function including mitochondrial energy production and antioxidative defense, at the expense of intracellular copper storage.
Collapse
Affiliation(s)
- Danica Tasić
- Department of Biochemistry, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Miloš Opačić
- Department of Life Sciences, Institute for Multidisciplinary Research, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Sanja Kovačević
- Department of Biochemistry, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Aleksandra Nikolić Kokić
- Department of Physiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Milena Dimitrijević
- Department of Life Sciences, Institute for Multidisciplinary Research, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Dušan Nikolić
- Department of Biology and Inland Waters Protection, Institute for Multidisciplinary Research, University of Belgrade, Kneza Višeslava 1, 11030 Belgrade, Serbia
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Duško Blagojević
- Department of Physiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
| | - Jelena Brkljačić
- Department of Biochemistry, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 142 Despot Stefan Blvd, 11060 Belgrade, Serbia
- Correspondence: ; Tel.: +381-11-2078318
| |
Collapse
|
38
|
Li T, Zhou L, Fan M, Chen Z, Yan L, Lu H, Jia M, Wu H, Shan L. Human Umbilical Cord-Derived Mesenchymal Stem Cells Ameliorate Skin Aging of Nude Mice Through Autophagy-Mediated Anti-Senescent Mechanism. Stem Cell Rev Rep 2022; 18:2088-2103. [PMID: 35864432 DOI: 10.1007/s12015-022-10418-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
Skin aging is a currently irreversible process, affected by increased oxidative stress, activated cellular senescence, and lacked regeneration of the dermal layer. Mesenchymal stem cells (MSCs), such as human umbilical cord-derived MSCs (hucMSCs), have pro-regeneration and anti-aging potencies. To explore whether hucMSCs can be used to treat skin aging, this study employed skin-aging model of nude mice to conduct in vivo assays, including biochemical analysis of superoxide dismutase (SOD) and malondialdehyde (MDA), gross observation, histopathological observation, and immunohistochemical analysis. To clarify how hucMSCs work on skin aging, this study employed skin-aging model of human dermal fibroblasts (HDFs) to conduct in vitro assays by applying conditional medium of hucMSCs (CMM), including wound healing assay, senescence staining, flow cytometric oxidative detection, real time PCR, and western blot analysis. The in vivo data demonstrated that hucMSCs dose-dependently removed wrinkles, smoothed skin texture, and increased dermal thickness and collagen production of aged skin by reversing SOD and MDA levels and up-regulating Col-1 and VEGF expressions, indicating anti-oxidative and pro-regenerative effects against skin aging. The in vitro data revealed that hucMSCs significantly reversed the senescence of HDFs by promoting cell migration, inhibiting ROS production, and restoring the overexpressions of oxidative and senescent markers through paracrine mode of action, and the paracrine mechanism was mediated by the inhibition of autophagy. This study provided novel knowledge regarding the anti-aging efficacy and paracrine mechanism of hucMSCs on skin, making hucMSCs-based therapy a promising regime for skin aging treatment.
Collapse
Affiliation(s)
- Ting Li
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengqiang Fan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| | - Haishan Lu
- Department of Dermatology, PLA 903 Hospital, Hangzhou, China
| | - Ming Jia
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiling Wu
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China. .,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China. .,Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China.
| |
Collapse
|
39
|
Bian X, Zhuang X, Zheng Z, Liu S, Liu Z, Song F. Unfolding and aggregation of oxidized metal-deficient superoxide dismutase and isoflavone inhibition based on ion mobility mass spectrometry and ThT fluorescence assay. Arch Biochem Biophys 2022; 727:109306. [DOI: 10.1016/j.abb.2022.109306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/02/2022]
|
40
|
Bernhard P, Bretthauer BA, Brixius SJ, Bügener H, Groh JE, Scherer C, Damjanovic D, Haberstroh J, Trummer G, Benk C, Beyersdorf F, Schilling O, Pooth JS. Serum proteome alterations during conventional and extracorporeal resuscitation in pigs. J Transl Med 2022; 20:238. [PMID: 35606879 PMCID: PMC9125930 DOI: 10.1186/s12967-022-03441-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/13/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Only a small number of patients survive an out-of-hospital cardiac arrest (CA) and can be discharged from hospital alive with a large percentage of these patients retaining neurological impairments. In recent years, extracorporeal cardiopulmonary resuscitation (ECPR) has emerged as a beneficial strategy to optimize cardiac arrest treatment. However, ECPR is still associated with various complications. To reduce these problems, a profound understanding of the underlying mechanisms is required. This study aims to investigate the effects of CA, conventional cardiopulmonary resuscitation (CPR) and ECPR using a whole-body reperfusion protocol (controlled and automated reperfusion of the whole body-CARL) on the serum proteome profiles in a pig model of refractory CA. METHODS N = 7 pigs underwent 5 min of untreated CA followed by 30 min CPR and 120 min perfusion with CARL. Blood samples for proteomic analysis were drawn at baseline, after CPR and at the end of the CARL period. Following albumin-depletion, proteomic analysis was performed using liquid chromatography-tandem mass spectrometry. RESULTS N = 21 serum samples were measured resulting in the identification and quantification of 308-360 proteins per sample and 388 unique proteins in total. The three serum proteome profiles at the investigated time points clustered individually and segregated almost completely when considering a 90% confidence interval. Differential expression analysis showed significant abundance changes in 27 proteins between baseline and after CPR and in 9 proteins after CARL compared to CPR. Significant findings were further validated through a co-abundance cluster analysis corroborating the observed abundance changes. CONCLUSIONS The presented data highlight the impact of systemic ischemia and reperfusion on the entire serum proteome during resuscitation with a special focus on changes regarding haemolysis, coagulation, inflammation, and cell-death processes. Generally, the observed changes contribute to post-ischemic complications. Better understanding of the underlying mechanisms during CA and resuscitation may help to limit these complications and improve therapeutic options.
Collapse
Affiliation(s)
- Patrick Bernhard
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Berit Amelie Bretthauer
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Sam Joé Brixius
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Hannah Bügener
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Johannes Elias Groh
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Christian Scherer
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Domagoj Damjanovic
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Jörg Haberstroh
- Department of Experimental Surgery, Center for Experimental Models and Transgenic Service, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Georg Trummer
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Christoph Benk
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Friedhelm Beyersdorf
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jan-Steffen Pooth
- Department of Cardiovascular Surgery, University Heart Center Freiburg, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| |
Collapse
|
41
|
Trist BG, Genoud S, Roudeau S, Rookyard A, Abdeen A, Cottam V, Hare DJ, White M, Altvater J, Fifita JA, Hogan A, Grima N, Blair IP, Kysenius K, Crouch PJ, Carmona A, Rufin Y, Claverol S, Van Malderen S, Falkenberg G, Paterson DJ, Smith B, Troakes C, Vance C, Shaw CE, Al-Sarraj S, Cordwell S, Halliday G, Ortega R, Double KL. Altered SOD1 maturation and post-translational modification in amyotrophic lateral sclerosis spinal cord. Brain 2022; 145:3108-3130. [PMID: 35512359 PMCID: PMC9473357 DOI: 10.1093/brain/awac165] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Aberrant self-assembly and toxicity of wild-type and mutant superoxide dismutase 1 (SOD1) has been widely examined in silico, in vitro, and in transgenic animal models of amyotrophic lateral sclerosis (ALS). Detailed examination of the protein in disease-affected tissues from ALS patients, however, remains scarce. We employed histological, biochemical and analytical techniques to profile alterations to SOD1 protein deposition, subcellular localization, maturation and post-translational modification in post-mortem spinal cord tissues from ALS cases and controls. Tissues were dissected into ventral and dorsal spinal cord grey matter to assess the specificity of alterations within regions of motor neuron degeneration. We provide evidence of the mislocalization and accumulation of structurally-disordered, immature SOD1 protein conformers in spinal cord motor neurons of SOD1-linked and non-SOD1-linked familial ALS cases, and sporadic ALS cases, compared with control motor neurons. These changes were collectively associated with instability and mismetallation of enzymatically-active SOD1 dimers, as well as alterations to SOD1 post-translational modifications and molecular chaperones governing SOD1 maturation. Atypical changes to SOD1 protein were largely restricted to regions of neurodegeneration in ALS cases, and clearly differentiated all forms of ALS from controls. Substantial heterogeneity in the presence of these changes was also observed between ALS cases. Our data demonstrates that varying forms of SOD1 proteinopathy are a common feature of all forms of ALS, and support the presence of one or more convergent biochemical pathways leading to SOD1 proteinopathy in ALS. The majority of these alterations are specific to regions of neurodegeneration, and may therefore constitute valid targets for therapeutic development.
Collapse
Affiliation(s)
- Benjamin G Trist
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sian Genoud
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Stéphane Roudeau
- Univ. Bordeaux, CNRS, CENBG, UMR 5797, F-33170 Gradignan, France
| | - Alexander Rookyard
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Amr Abdeen
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Veronica Cottam
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Dominic J Hare
- School of Biosciences, The University of Melbourne, Parkville, Victoria 3010, Australia.,Atomic Medicine Initiative, University of Technology Sydney, Broadway, New South Wales 2007, Australia
| | - Melanie White
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jens Altvater
- Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jennifer A Fifita
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Alison Hogan
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Natalie Grima
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Ian P Blair
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Kai Kysenius
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter J Crouch
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Asuncion Carmona
- Univ. Bordeaux, CNRS, CENBG, UMR 5797, F-33170 Gradignan, France
| | - Yann Rufin
- Plateforme Biochimie, University of Bordeaux, France
| | | | - Stijn Van Malderen
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Gerald Falkenberg
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - David J Paterson
- Australian Synchrotron, ANSTO, Clayton, Victoria 3168, Australia
| | - Bradley Smith
- Maurice Wohl Clinical Neuroscience Institute and the Institute of Psychiatry, Psychology and Neuroscience, King's College London, Camberwell, SE5 9RT, London, UK
| | - Claire Troakes
- UK Dementia Research Institute at King's College London, 5 Cutcombe Road, London, SE5 9RT, UK
| | - Caroline Vance
- Maurice Wohl Clinical Neuroscience Institute and the Institute of Psychiatry, Psychology and Neuroscience, King's College London, Camberwell, SE5 9RT, London, UK
| | - Christopher E Shaw
- UK Dementia Research Institute at King's College London, 5 Cutcombe Road, London, SE5 9RT, UK
| | - Safa Al-Sarraj
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AF, London, UK
| | - Stuart Cordwell
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Glenda Halliday
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Richard Ortega
- Univ. Bordeaux, CNRS, CENBG, UMR 5797, F-33170 Gradignan, France
| | - Kay L Double
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
42
|
Skibiel AL, Koh J, Zhu N, Zhu F, Yoo MJ, Laporta J. Carry-over effects of dry period heat stress on the mammary gland proteome and phosphoproteome in the subsequent lactation of dairy cows. Sci Rep 2022; 12:6637. [PMID: 35459770 PMCID: PMC9033811 DOI: 10.1038/s41598-022-10461-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 04/01/2022] [Indexed: 11/15/2022] Open
Abstract
Exposure to heat stress during a cow's dry period disrupts mammary gland remodeling, impairing mammary function and milk production during the subsequent lactation. Yet, proteomic changes in the mammary gland underlying these effects are not yet known. We investigated alterations in the mammary proteome and phosphoproteome during lactation as a result of dry period heat stress using an isobaric tag for relative and absolute quantitation (iTRAQ)-based approach. Cows were cooled (CL; n = 12) with fans and water soakers in a free stall setting or were heat stressed through lack of access to cooling devices (HT; n = 12) during the entire dry period (approximately 46 days). All cows were cooled postpartum. Mammary biopsies were harvested from a subset of cows (n = 4 per treatment) at 14, 42, and 84 days in milk. Overall, 251 proteins and 224 phosphorylated proteins were differentially abundant in the lactating mammary gland of HT compared to CL cows. Top functions of differentially abundant proteins and phosphoproteins affected were related to immune function and inflammation, amino acid metabolism, reactive oxygen species production and metabolism, tissue remodeling, and cell stress response. Patterns of protein expression and phosphorylation are indicative of increased oxidative stress, mammary gland restructuring, and immune dysregulation due to prior exposure to dry period heat stress. This study provides insights into the molecular underpinnings of disrupted mammary function and health during lactation arising from prior exposure to dry period heat stress, which might have led to lower milk yields.
Collapse
Affiliation(s)
- Amy L Skibiel
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Jin Koh
- Interdisciplinary Center for Biotechnology Research, Proteomics and Mass Spectrometry Core, University of Florida, Gainesville, FL, 32611, USA
| | - Ning Zhu
- Interdisciplinary Center for Biotechnology Research, Proteomics and Mass Spectrometry Core, University of Florida, Gainesville, FL, 32611, USA
| | - Fanchao Zhu
- Interdisciplinary Center for Biotechnology Research, Proteomics and Mass Spectrometry Core, University of Florida, Gainesville, FL, 32611, USA
| | - Mi-Jeong Yoo
- Department of Biology, Clarkson University, Potsdam, NY, 13699, USA
| | - Jimena Laporta
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| |
Collapse
|
43
|
Wadood AA, Pu L, Shahzad Q, Waqas M, Yu L, Liao Y, Rehman SU, Chen D, Huang Z, Lu Y. Proteomic analysis identifies potential markers in small white and small yellow follicle development in chickens. Reprod Fertil Dev 2022; 34:516-525. [PMID: 35296374 DOI: 10.1071/rd21184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 12/21/2021] [Indexed: 11/23/2022] Open
Abstract
Extensive knowledge of follicular development is imperative for improving egg production in chickens. The functional role of follicles to produce oocytes (eggs) is well recognised; however, specific markers associated with follicle development have been poorly explored. Therefore, a tandem mass tag based proteomic technique was used to identify the status of the proteome of small white follicles (1-4mm) and small yellow follicles (6-8mm). Analysis of differentially expressed proteins (DEP, Fold Change>1.2, P -value<0.05) demonstrated a total of 92 proteins (n =92), of which 35 (n =35) were upregulated and 57 were downregulated. DEP were further used for gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathways. The GO analysis found that DEP were mainly associated with the RNA metabolic process, cellular component organisation, peptide biosynthetic process and protein folding, thereby suggesting a key role in the follicle development process. Kyoto Encyclopedia of Genes and Genomes enrichment pathway analysis of the DEP substantiated the findings of GO analysis and described that DEP are involved in regulation of the cytoskeleton, carbon metabolism and amino acid biosynthesis. The validation of proteomic data through real-time quantitative polymerase chain reaction suggested HSPA8, HSPA2, SOD1 and FKPB3 as potential markers of small white and small yellow follicle development. This study demonstrates an understanding of proteome dynamics and represents the most comprehensive information on the entire Guangxi Ma chicken follicular proteome.
Collapse
Affiliation(s)
- Armughan Ahmed Wadood
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Liping Pu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Qaisar Shahzad
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Muhammad Waqas
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Lintian Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Yuying Liao
- Guangxi Veterinary Research Institute, Nanning, Guangxi, China
| | - Saif Ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Dongyang Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Zhenwen Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| |
Collapse
|
44
|
Nucera F, Mumby S, Paudel KR, Dharwal V, DI Stefano A, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of oxidative stress in the pathogenesis of COPD. Minerva Med 2022; 113:370-404. [PMID: 35142479 DOI: 10.23736/s0026-4806.22.07972-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic inhalation of cigarette smoke is a prominent cause of chronic obstructive pulmonary disease (COPD) and provides an important source of exogenous oxidants. In addition, several inflammatory and structural cells are a source of endogenous oxidants in the lower airways of COPD patients, even in former smokers. This suggests that oxidants play a key role in the pathogenesis of COPD. This oxidative stress is counterbalanced by the protective effects of the various endogenous antioxidant defenses of the lower airways. A large amount of data from animal models and patients with COPD have shown that both the stable phase of the disease, and during exacerbations, have increased oxidative stress in the lower airways compared with age-matched smokers with normal lung function. Thus, counteracting the increased oxidative stress may produce clinical benefits in COPD patients. Smoking cessation is currently the most effective treatment of COPD patients and reduces oxidative stress in the lower airways. In addition, many drugs used to treat COPD have some antioxidant effects, however, it is still unclear if their clinical efficacy is related to pharmacological modulation of the oxidant/antioxidant balance. Several new antioxidant compounds are in development for the treatment of COPD.
Collapse
Affiliation(s)
- Francesco Nucera
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy -
| | - Sharon Mumby
- Airways Diseases Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, UK
| | - Keshav R Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Vivek Dharwal
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Antonino DI Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Veruno, Novara, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry Scuola Medica Salernitana, University of Salerno, Salerno, Italy
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Ian M Adcock
- Airways Diseases Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, UK
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| |
Collapse
|
45
|
Thauvin M, de Sousa RM, Alves M, Volovitch M, Vriz S, Rampon C. An early Shh-H2O2 reciprocal regulatory interaction controls the regenerative program during zebrafish fin regeneration. J Cell Sci 2022; 135:274206. [PMID: 35107164 DOI: 10.1242/jcs.259664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 11/20/2022] Open
Abstract
Reactive oxygen species (ROS), originally classified as toxic molecules, have attracted increasing interest given their actions in cell signaling. Hydrogen peroxide (H2O2), the major ROS produced by cells, acts as a second messenger to modify redox-sensitive proteins or lipids. After caudal fin amputation, tight spatiotemporal regulation of ROS is required first for wound healing and later to initiate the regenerative program. However, the mechanisms carrying out this sustained ROS production and their integration with signaling pathways are still poorly understood. We focused on the early dialog between H2O2 and Sonic Hedgehog (Shh) during fin regeneration. We demonstrate that H2O2 controls Shh expression and that Shh in turn regulates the H2O2 level via a canonical pathway. Moreover, the means of this tight reciprocal control change during the successive phases of the regenerative program. Dysregulation of the Hedgehog pathway has been implicated in several developmental syndromes, diabetes and cancer. These data support the existence of an early positive crosstalk between Shh and H2O2 that might be more generally involved in various processes paving the way to improve regenerative processes, particularly in vertebrates.
Collapse
Affiliation(s)
- Marion Thauvin
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Rodolphe Matias de Sousa
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Marine Alves
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Université de Paris, Faculty of Sciences, Paris, France
| | - Michel Volovitch
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,École Normale Supérieure, PSL Research University, Department of Biology, Paris, France
| | - Sophie Vriz
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Université de Paris, Faculty of Sciences, Paris, France
| | - Christine Rampon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Université de Paris, Faculty of Sciences, Paris, France
| |
Collapse
|
46
|
Sod1 integrates oxygen availability to redox regulate NADPH production and the thiol redoxome. Proc Natl Acad Sci U S A 2022; 119:2023328119. [PMID: 34969852 PMCID: PMC8740578 DOI: 10.1073/pnas.2023328119] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2021] [Indexed: 12/12/2022] Open
Abstract
Cu/Zn superoxide dismutase (Sod1) is a key antioxidant enzyme, and its importance is underscored by the fact that its ablation in cell and animal models results in oxidative stress; metabolic defects; and reductions in cell proliferation, viability, and lifespan. Curiously, Sod1 detoxifies superoxide radicals (O2•−) in a manner that produces an oxidant as byproduct, hydrogen peroxide (H2O2). While much is known about the necessity of scavenging O2•−, it is less clear what the physiological roles of Sod1-derived H2O2 are. We discovered that Sod1-derived H2O2 plays an important role in antioxidant defense by stimulating the production of NADPH, a vital cellular reductant required for reactive oxygen species scavenging enzymes, as well as redox regulating a large network of enzymes. Cu/Zn superoxide dismutase (Sod1) is a highly conserved and abundant antioxidant enzyme that detoxifies superoxide (O2•−) by catalyzing its conversion to dioxygen (O2) and hydrogen peroxide (H2O2). Using Saccharomyces cerevisiae and mammalian cells, we discovered that a major aspect of the antioxidant function of Sod1 is to integrate O2 availability to promote NADPH production. The mechanism involves Sod1-derived H2O2 oxidatively inactivating the glycolytic enzyme, GAPDH, which in turn reroutes carbohydrate flux to the oxidative phase of the pentose phosphate pathway (oxPPP) to generate NADPH. The aerobic oxidation of GAPDH is dependent on and rate-limited by Sod1. Thus, Sod1 senses O2 via O2•− to balance glycolytic and oxPPP flux, through control of GAPDH activity, for adaptation to life in air. Importantly, this mechanism for Sod1 antioxidant activity requires the bulk of cellular Sod1, unlike for its role in protection against O2•− toxicity, which only requires <1% of total Sod1. Using mass spectrometry, we identified proteome-wide targets of Sod1-dependent redox signaling, including numerous metabolic enzymes. Altogether, Sod1-derived H2O2 is important for antioxidant defense and a master regulator of metabolism and the thiol redoxome.
Collapse
|
47
|
Lutsenko S. Dynamic and cell-specific transport networks for intracellular copper ions. J Cell Sci 2021; 134:272704. [PMID: 34734631 DOI: 10.1242/jcs.240523] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Copper (Cu) homeostasis is essential for the development and function of many organisms. In humans, Cu misbalance causes serious pathologies and has been observed in a growing number of diseases. This Review focuses on mammalian Cu(I) transporters and highlights recent studies on regulation of intracellular Cu fluxes. Cu is used by essential metabolic enzymes for their activity. These enzymes are located in various intracellular compartments and outside cells. When cells differentiate, or their metabolic state is otherwise altered, the need for Cu in different cell compartments change, and Cu has to be redistributed to accommodate these changes. The Cu transporters SLC31A1 (CTR1), SLC31A2 (CTR2), ATP7A and ATP7B regulate Cu content in cellular compartments and maintain Cu homeostasis. Increasing numbers of regulatory proteins have been shown to contribute to multifaceted regulation of these Cu transporters. It is becoming abundantly clear that the Cu transport networks are dynamic and cell specific. The comparison of the Cu transport machinery in the liver and intestine illustrates the distinct composition and dissimilar regulatory response of their Cu transporters to changing Cu levels.
Collapse
Affiliation(s)
- Svetlana Lutsenko
- Johns Hopkins Medical Institutes, Department of Physiology, Baltimore, MD 21205, USA
| |
Collapse
|
48
|
Klichkhanov NK, Nikitina ER, Shihamirova ZM, Astaeva MD, Chalabov SI, Krivchenko AI. Erythrocytes of Little Ground Squirrels Undergo Reversible Oxidative Stress During Arousal From Hibernation. Front Physiol 2021; 12:730657. [PMID: 34690805 PMCID: PMC8529035 DOI: 10.3389/fphys.2021.730657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022] Open
Abstract
The hibernation of small mammals is characterized by long torpor bouts alternating with short periods of arousal. During arousal, due to a significant increase in oxygen consumption, tissue perfusion, and the launch of thermogenesis in cells, a large amount of reactive oxygen species (ROS) and nitrogen (RNS) can be formed, which can trigger oxidative stress in cells. To estimate this possibility, we studied the intensity of free-radical processes in the red blood cells (RBCs) of little ground squirrels (LGS; Spermophilus pygmaeus) in the dynamics of arousal from hibernation. We found that in the torpid state, the degree of generation of ROS and RNS (8.3%, p>0.09; 20.7%, p<0.001, respectively), the degree of oxidative modification of membrane lipids and RBC proteins is at a low level (47%, p<0.001; 82.7%, p<0.001, respectively) compared to the summer control. At the same time, the activity of superoxide dismutase (SOD) and catalase (CAT) in RBC is significantly reduced (32.8%, p<0.001; 22.2%, p<0.001, respectively), but not the level of glutathione (GSH). In the torpid state, SOD is activated by exogenous GSH in concentration-dependent manner, which indicates reversible enzyme inhibition. During the arousal of ground squirrels, when the body temperature reaches 25°C, RBCs are exposed oxidative stress. This is confirmed by the maximum increase in the level of uric acid (25.4%, p<0.001) in plasma, a marker of oxidative modification of lipids [thiobarbituric acid reactive substances (TBARS); 82%, p < 0.001] and proteins (carbonyl groups; 499%, p < 0.001) in RBC membranes, as well as the decrease in the level of GSH (19.7%, p < 0.001) in erythrocytes relative to the torpid state and activity of SOD and CAT in erythrocytes to values at the Tb 20°C. After full recovery of body temperature, the level of GSH increases, the ratio of SOD/CAT is restored, which significantly reduces the degree of oxidative damage of lipids and proteins of RBC membranes. Thus, the oxidative stress detected at Tb 25°C was transient and physiologically regulated.
Collapse
Affiliation(s)
| | - Elena R Nikitina
- Laboratory of Comparative Physiology of Respiration, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Maria D Astaeva
- Department of Biochemistry, Dagestan State University, Makhachkala, Russia
| | - Shamil I Chalabov
- Department of Biochemistry, Dagestan State University, Makhachkala, Russia.,Laboratory of Comparative Physiology of Respiration, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Aleksandr I Krivchenko
- Laboratory of Comparative Physiology of Respiration, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
49
|
Computer analysis of the relation between hydrogen bond stability in SOD1 mutants and the survival time of amyotrophic lateral sclerosis patients. J Mol Graph Model 2021; 110:108026. [PMID: 34653813 DOI: 10.1016/j.jmgm.2021.108026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/06/2021] [Accepted: 09/04/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Mutations in the SOD1 protein can lead to the death of motor neurons, which, in turn, causes an incurable disease called amyotrophic lateral sclerosis (ALS). At the same time, the mechanism of the onset and development of this disease is not fully understood and is often contradictory. METHODS Accelerated Molecular Dynamics as implemented in the OpenMM library, principal component analysis, regression analysis, random forest method. RESULTS The stability of hydrogen bonds in 72 mutants of the SOD1 protein was calculated. Principal component analysis was carried out. Based on ten principal components acting as predictors, a multiple linear regression model was constructed. An analysis of the correlation of these ten principal components with the initial values of the stability of hydrogen bonds made it possible to characterize the contribution of known structurally and functionally important sites in the SOD1 to the scatter of ALS patients' survival time. CONCLUSION Such an analysis made it possible to put forward hypotheses about the relationship between the stabilizing and destabilizing effects of mutations in different structurally and functionally important regions of SOD1 with the patients's survival time.
Collapse
|
50
|
Manochkumar J, Doss CGP, El-Seedi HR, Efferth T, Ramamoorthy S. The neuroprotective potential of carotenoids in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153676. [PMID: 34339943 DOI: 10.1016/j.phymed.2021.153676] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/26/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Despite advances in research on neurodegenerative diseases, the pathogenesis and treatment response of neurodegenerative diseases remain unclear. Recent studies revealed a significant role of carotenoids to treat neurodegenerative diseases. The aim of this study was to systematically review the neuroprotective potential of carotenoids in vivo and in vitro and the molecular mechanisms and pathological factors contributing to major neurodegenerative diseases (Alzheimer's disease, Huntington's disease, Parkinson's disease, amyotrophic lateral sclerosis, and stroke). HYPOTHESIS Carotenoids as therapeutic molecules to target neurodegenerative diseases. RESULTS Aggregation of toxic proteins, mitochondrial dysfunction, oxidative stress, the excitotoxic pathway, and neuroinflammation were the major pathological factors contributing to the progression of neurodegenerative diseases. Furthermore, in vitro and in vivo studies supported the beneficiary role of carotenoids, namely lycopene, β-carotene, crocin, crocetin, lutein, fucoxanthin and astaxanthin in alleviating disease progression. These carotenoids provide neuroprotection by inhibition of neuro-inflammation, microglial activation, excitotoxic pathway, modulation of autophagy, attenuation of oxidative damage and activation of defensive antioxidant enzymes. Additionally, studies conducted on humans also demonstrated that dietary intake of carotenoids lowers the risk of neurodegenerative diseases. CONCLUSION Carotenoids may be used as drugs to prevent and treat neurodegenerative diseases. Although, the in vitro and in vivo results are encouraging, further well conducted clinical studies on humans are required to conclude about the full potential of neurodegenerative diseases.
Collapse
Affiliation(s)
- Janani Manochkumar
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - C George Priya Doss
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Hesham R El-Seedi
- Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Box 574, SE-75 123 Uppsala, Sweden; Department of Chemistry, Faculty of Science, Menoufia University, 32512 Shebin El-Koom, Egypt
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Germany
| | - Siva Ramamoorthy
- School of Bio Sciences and Technology, VIT University, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|