1
|
Duan C, Zhao Y, Xiao Y, Hou Y, Gong W, Zhang H, Wang Y, Nie X. Lithium with environmentally relevant concentrations interferes with mitochondrial function, antioxidant response, and autophagy processes in Daphnia magna, leading to changes in life-history traits and behavior. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137420. [PMID: 39893979 DOI: 10.1016/j.jhazmat.2025.137420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/09/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
With the increasing production and use of lithium-based products, concerns over lithium pollution in aquatic ecosystems are increasing, whereas research on its toxicity mechanisms in aquatic organisms remains limited. The main objective of the present study was to explore the effects of environmentally relevant concentrations of lithium exposure on the life-history strategy, behavior, antioxidant system, and autophagy process of Daphnia magna. Acute (24-96 h) and chronic (21 days) exposure experiments under three lithium treatments (low: 8.34 μg/L, medium: 83.44 μg/L, and high: 834.41 μg/L) were conducted. The results indicated that exposure to medium and high lithium concentrations led to eye and tail deformities in D. magna. Furthermore, developmental and reproductive parameters such as body length, total neonates per female, and average neonates per time were negatively influenced. Lithium also interfered with energy metabolism to cause the decreasing swimming speed and the reduction in the swimming range. In addition, lithium exposure affected the expression of gsk-3β, further disrupting the dynamic balance of mitochondrial fission, fusion, and regeneration, which caused ROS accumulation and induced oxidative stress. D. magna attenuated the stress by activating the FoxO/SESN and Nrf2/Keap1 pathways, synergistically enhancing downstream antioxidant enzymes expression. Concurrently, D. magna also mitigated oxidative stress and mitochondrial damage by promoting autophagy and inhibiting apoptosis. In summary, lithium harmed the physiological and biochemical functions of D. magna through multiple mechanisms, suggesting that environmental lithium pollution may pose a potential threat to aquatic organisms.
Collapse
Affiliation(s)
- Chunni Duan
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yuanyuan Xiao
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yingshi Hou
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Weibo Gong
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Huiyu Zhang
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
2
|
Li J, Sun S, Li Y, Tian M, Li X, Ren S, Huang Z, Wang Y, Du S. Nrf2 signaling pathway studies in Drosophila melanogaster: parallel roles in human health and insect environmental responses. Xenobiotica 2025; 55:85-98. [PMID: 39932394 DOI: 10.1080/00498254.2025.2465239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/25/2025]
Abstract
The Nrf2 signalling pathway is crucial for cellular defense against oxidative stress and xenobiotic toxicity, highlighting its importance in both human health and environmental responses.This review focuses on the dual role of Drosophila melanogaster in Nrf2 research: we utilised the PubMed database to collect and summarised research articles on fruit fly Nrf2 studies published in the past decade, using keywords such as 'Nrf2', 'CncC', and 'Drosophila'.We found that Drosophila melanogaster, as a classical model organism for studying human diseases such as neurodegenerative disorders, cancers, and diabetes, and as an insect model for investigating xenobiotic responses and pesticide resistance, is particularly well-suited for exploring the diverse and complex functions of Nrf2 pathway.Additionally, Natural products such as curcumin and quercetin can modulate Nrf2 activity for cytoprotection. Utilising D. melanogaster's genetic tools and short life cycles, researchers can discover new therapeutics and study their mechanisms.This twofold exploration not only advances our understanding of Nrf2 in human health but also provides insights into pest control strategies through enhanced insect resistance mechanisms. Continued research in this area is essential for developing innovative treatments and effective pest management approaches.
Collapse
Affiliation(s)
- Jingyi Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Shushen Sun
- Department of Gastroenterology, Tianjin University Jinnan Hospital (Tianjin Jinnan Hospital), Tianjin, China
| | - Ying Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Mengzhe Tian
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xinyi Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Suxia Ren
- School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Zengyi Huang
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Shaoshan Du
- Department of Gastroenterology, Tianjin University Jinnan Hospital (Tianjin Jinnan Hospital), Tianjin, China
| |
Collapse
|
3
|
Teh YM, Mualif SA, Mohd Noh NI, Lim SK. The Potential of Naturally Derived Compounds for Treating Chronic Kidney Disease: A Review of Autophagy and Cellular Senescence. Int J Mol Sci 2024; 26:3. [PMID: 39795863 PMCID: PMC11719669 DOI: 10.3390/ijms26010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic kidney disease (CKD) is characterized by irreversible progressive worsening of kidney function leading to kidney failure. CKD is viewed as a clinical model of premature aging and to date, there is no treatment to reverse kidney damage. The well-established treatment for CKD aims to control factors that may aggravate kidney progression and to provide kidney protection effects to delay the progression of kidney disease. As an alternative, Traditional Chinese Medicine (TCM) has been shown to have fewer adverse effects for CKD patients. However, there is a lack of clinical and molecular studies investigating the mechanisms by which natural products used in TCM can improve CKD. In recent years, autophagy and cellular senescence have been identified as key contributors to aging and age-related diseases. Exploring the potential of natural products in TCM to target these processes in CKD patients could slow disease progression. A better understanding of the characteristics of these natural products and their effects on autophagy and cellular senescence through clinical studies, coupled with the use of these products as complementary therapy alongside mainstream treatment, may maximize therapeutic benefits and minimize adverse effects for CKD patients. While promising, there is currently a lack of thorough research on the potential synergistic effects of these natural products. This review examines the use of natural products in TCM as an alternative treatment for CKD and discusses their active ingredients in terms of renoprotection, autophagy, and cellular senescence.
Collapse
Affiliation(s)
- Yoong Mond Teh
- Department of Biomedical Engineering and Health Science, Faculty of Electrical Engineering, University Technology Malaysia (UTM), Johor Bahru 81310, Malaysia; (Y.M.T.); (S.A.M.)
| | - Siti Aisyah Mualif
- Department of Biomedical Engineering and Health Science, Faculty of Electrical Engineering, University Technology Malaysia (UTM), Johor Bahru 81310, Malaysia; (Y.M.T.); (S.A.M.)
| | - Nur Izzati Mohd Noh
- Department of Biosciences, Faculty of Science, University Technology Malaysia (UTM), Johor Bahru 81310, Malaysia;
| | - Soo Kun Lim
- Department of Medicine, Faculty of Medicine, University of Malaysia (UM), Kuala Lumpur 59100, Malaysia
| |
Collapse
|
4
|
Zhao Y, Duan C, Xiao Y, Gong W, Wang Y, Zhang H, Ku P, Nie X. Water acidification aggravates lithium-induced toxicity represented by energy supply, oxidative stress, and cell fate in Daphnia magna neonates. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177143. [PMID: 39490820 DOI: 10.1016/j.scitotenv.2024.177143] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/01/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Lithium is extensively utilized in industrial energy production, particularly in lithium-ion batteries, and in pharmaceuticals for the treating clinical mood disorders. Consequently, lithium is frequently detected in various environmental matrices. It has been reported to cause a range of toxic effects on aquatic organisms including oxidative stress, neurological disorders, and reproductive suppression. Water acidification is a global issue with numerous negative impacts on aquatic organisms. It can alter the physio-chemical properties and bioavailability of metal ions. The acidic leaching process during lithium battery treatment and global water acidification both suggest that lithium contamination often occurs in acidic environments. In the present study, Daphnia magna neonates were exposed to four treatments (control, lithium alone, low pH, and combined) to investigate whether an acidic environment exacerbates the toxic effects of lithium on aquatic organisms and to explore potential toxic action mechanisms. The results indicated that low pH posed a significant threat to the growth and reproduction of D. magna. When exposed to both lithium and low pH, there was increased lithium accumulation and an energy trade-off response, leading to increased energy allocation to reproduction and reduced energy for growth. Lithium exposure stimulated D. magna activity, while low pH inhibited it, suggesting that an imbalance in energy consumption and supply. Combined exposure to lithium and low pH resulted in severe oxidative stress due to mitochondrial dysfunction, under-utilization of energy substances, and increased ionic homeostasis disturbances. Consequently, the exposed organism altered apoptosis and autophagy processes to maintain homeostasis. The present study demonstrated that lithium and water acidification posed a population-level threat to D. magna, and their combined exposure significantly largely exacerbated the toxic effects.
Collapse
Affiliation(s)
- Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Chunni Duan
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yuanyuan Xiao
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Weibo Gong
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou 510632, China; Guangdong Laboratory Animals Monitoring Institute, Guangzhou 510663, China
| | - Huiyu Zhang
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Peijia Ku
- Environmental Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
5
|
Zhao Q, Huang Y, Fu N, Cui C, Peng X, Kang H, Xiao J, Ke G. Podocyte senescence: from molecular mechanisms to therapeutics. Ren Fail 2024; 46:2398712. [PMID: 39248407 PMCID: PMC11385655 DOI: 10.1080/0886022x.2024.2398712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
As an important component of the glomerular filtration membrane, the state of the podocytes is closely related to kidney function, they are also key cells involved in aging and play a central role in the damage caused by renal aging. Therefore, understanding the aging process of podocytes will allow us to understand their susceptibility to injury and identify targeted protective mechanisms. In fact, the process of physiological aging itself can induce podocyte senescence. Pathological stresses, such as oxidative stress, mitochondrial damage, secretion of senescence-associated secretory phenotype, reduced autophagy, oncogene activation, altered transcription factors, DNA damage response, and other factors, play a crucial role in inducing premature senescence and accelerating aging. Senescence-associated-β-galactosidase (SA-β-gal) is a marker of aging, and β-hydroxybutyric acid treatment can reduce SA-β-gal activity to alleviate cellular senescence and damage. In addition, CCAAT/enhancer-binding protein-α, transforming growth factor-β signaling, glycogen synthase kinase-3β, cycle-dependent kinase, programmed cell death protein 1, and plasminogen activator inhibitor-1 are closely related to aging. The absence or elevation of these factors can affect aging through different mechanisms. Podocyte injury is not an independent process, and injured podocytes interact with the surrounding epithelial cells or other kidney cells to mediate the injury or loss of podocytes. In this review, we discuss the manifestations, molecular mechanisms, biomarkers, and therapeutic drugs for podocyte senescence. We included elamipretide, lithium, calorie restriction, rapamycin; and emerging treatment strategies, such as gene and immune therapies. More importantly, we summarize how podocyte interact with other kidney cells.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongzhang Huang
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ningying Fu
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Caixia Cui
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xuan Peng
- Department of Nephrology, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu, China
| | - Haiyan Kang
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Xiao
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guibao Ke
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Jans K, Jöckel T, von Frieling J, Ipharraguerre IR, Roeder T, Lüersen K, Rimbach G. Lithium affects sodium balance but not intestinal microbiota - studies in Drosophila melanogaster. J Trace Elem Med Biol 2024; 86:127548. [PMID: 39442469 DOI: 10.1016/j.jtemb.2024.127548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND The trace element lithium (Li) is known for its therapeutic mood-stabilizing application in humans, but also for its various bioactivities, which have been uncovered in model organisms. According to the literature, Li may interfere with the homeostasis of other minerals in mammals, namely sodium, calcium and magnesium. In addition, Li was found to influence the composition and diversity of the intestinal microbiota in vertebrates, an observation that may be related to the many bioactivities of Li. METHODS Based on these previous findings, we employed the model organism Drosophila melanogaster to decipher whether Li exhibits similar bioactivities in invertebrates. First, we examined the influence of increasing dietary Li supply (0 -100 mM LiCl) on the status of Li and ten other minerals via Inductively coupled plasma - mass spectrometry (ICP-MS) in heads and remaining body parts of the three wildtype strains w1118, Oregon-R-C and Canton-S. In addition, we investigated the potential impact of Li feeding (0, 0.1, 1 mM LiCl) on the total bacterial load, α- and β-diversity via real-time quantitative polymerase chain reaction (RT q-PCR) and 16S rDNA sequencing in the intestines of female w1118. RESULTS Our observations revealed that Li accumulates linearly in both sexes and all body parts of the three Drosophila strains as the dietary Li supply increases. While the status of most elements remained unchanged, the sodium levels of the fly also correlated positively with the Li content of the diet. The intestinal microbiota, however, remained largely unaffected by Li feeding in terms of both, bacterial load and diversity. CONCLUSION These findings support the hypothesis that elevating the Li supply affects sodium homeostasis in Drosophila, a finding coherent with observations in mammals. Furthermore, our data opposes a possible involvement of the bacterial intestinal colonization in the bioactivity of Li in Drosophila.
Collapse
Affiliation(s)
- Katharina Jans
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany.
| | - Tobias Jöckel
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany
| | - Jakob von Frieling
- Division of Molecular Physiology, Institute of Zoology, University of Kiel, Kiel D-24118, Germany
| | - Ignacio R Ipharraguerre
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, University of Kiel, Kiel D-24118, Germany
| | - Kai Lüersen
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany
| | - Gerald Rimbach
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany
| |
Collapse
|
7
|
Mutz J, Wong WLE, Powell TR, Young AH, Dawe GS, Lewis CM. The duration of lithium use and biological ageing: telomere length, frailty, metabolomic age and all-cause mortality. GeroScience 2024; 46:5981-5994. [PMID: 38539016 PMCID: PMC11493902 DOI: 10.1007/s11357-024-01142-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/17/2024] [Indexed: 10/23/2024] Open
Abstract
Lithium is an established first-line treatment for bipolar disorder. Beyond its therapeutic effect as a mood stabiliser, lithium exhibits potential anti-ageing effects. This study aimed to examine the relationship between the duration of lithium use, biological ageing and mortality. The UK Biobank is an observational study of middle-aged and older adults. We tested associations between the duration of lithium use (number of prescriptions, total duration of use and duration of the first prescription period) and telomere length, frailty, metabolomic age (MileAge) delta, pulse rate and all-cause mortality. Five hundred ninety-one individuals (mean age = 57.49 years; 55% females) had been prescribed lithium. There was no evidence that the number of prescriptions (β = - 0.022, 95% CI - 0.081 to 0.037, p = 0.47), the total duration of use (β = - 0.005, 95% CI - 0.023 to 0.013, p = 0.57) or the duration of the first prescription period (β = - 0.018, 95% CI - 0.051 to 0.015, p = 0.29) correlated with telomere length. There was also no evidence that the duration of lithium use correlated with frailty or MileAge delta. However, a higher prescription count and a longer duration of use was associated with a lower pulse rate. The duration of lithium use did not predict all-cause mortality. We observed no evidence of associations between the duration of lithium use and biological ageing markers, including telomere length. Our findings suggest that the potential anti-ageing effects of lithium do not differ by the duration of use.
Collapse
Affiliation(s)
- Julian Mutz
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Memory Lane, London, UK.
| | - Win Lee Edwin Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Timothy R Powell
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Memory Lane, London, UK
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- South London & Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, London, UK
| | - Gavin S Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Cathryn M Lewis
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Memory Lane, London, UK
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
8
|
Zhang Y, Li Y, Ren T, Duan JA, Xiao P. Promising tools into oxidative stress: A review of non-rodent model organisms. Redox Biol 2024; 77:103402. [PMID: 39437623 PMCID: PMC11532775 DOI: 10.1016/j.redox.2024.103402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
Oxidative stress is a crucial concept in redox biology, and significant progress has been made in recent years. Excessive levels of reactive oxygen species (ROS) can lead to oxidative damage, heightening vulnerability to various diseases. By contrast, ROS maintained within a moderate range plays a role in regulating normal physiological metabolism. Choosing suitable animal models in a complex research context is critical for enhancing research efficacy. While rodents are frequently utilized in medical experiments, they pose challenges such as high costs and ethical considerations. Alternatively, non-rodent model organisms like zebrafish, Drosophila, and C. elegans offer promising avenues into oxidative stress research. These organisms boast advantages such as their small size, high reproduction rate, availability for live imaging, and ease of gene manipulation. This review highlights advancements in the detection of oxidative stress using non-rodent models. The oxidative homeostasis regulatory pathway, Kelch-like ECH-associated protein 1-Nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2), is systematically reviewed alongside multiple regulation of Nrf2-centered pathways in different organisms. Ultimately, this review conducts a comprehensive comparative analysis of different model organisms and further explores the combination of novel techniques with non-rodents. This review aims to summarize state-of-the-art findings in oxidative stress research using non-rodents and to delineate future directions.
Collapse
Affiliation(s)
- Yuhao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yun Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tianyi Ren
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
9
|
Dos Santos E, Cochemé HM. Pharmacology of Aging: Drosophila as a Tool to Validate Drug Targets for Healthy Lifespan. AGING BIOLOGY 2024; 2:20240034. [PMID: 39346601 PMCID: PMC7616647 DOI: 10.59368/agingbio.20240034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Finding effective therapies to manage age-related conditions is an emerging public health challenge. Although disease-targeted treatments are important, a preventive approach focused on aging can be more efficient. Pharmacological targeting of aging-related processes can extend lifespan and improve health in animal models. However, drug development and translation are particularly challenging in geroscience. Preclinical studies have survival as a major endpoint for drug screening, which requires years of research in mammalian models. Shorter-lived invertebrates can be exploited to accelerate this process. In particular, the fruit fly Drosophila melanogaster allows the validation of new drug targets using precise genetic tools and proof-of-concept experiments on drugs impacting conserved aging processes. Screening for clinically approved drugs that act on aging-related targets may further accelerate translation and create new tools for aging research. To date, 31 drugs used in clinical practice have been shown to extend the lifespan of flies. Here, we describe recent advances in the pharmacology of aging, focusing on Drosophila as a tool to repurpose these drugs and study age-related processes.
Collapse
Affiliation(s)
- Eliano Dos Santos
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Helena M Cochemé
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
10
|
Feng YF, Zhang Y, Yang RJ, Li SQ, Liu XJ, Han C, Xing YF, Yang JX. Ecotoxicological assessment, oxidative response, and enzyme activity disorder of the rotifer Brachionus asplanchnoidis exposed to a toxic cocktail of spent lithium-ion battery leachate. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135050. [PMID: 38954852 DOI: 10.1016/j.jhazmat.2024.135050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
Spent lithium-ion batteries (LIBs) have emerged as a major source of waste due to their low recovery rate. The physical disposal of spent LIBs can lead to the leaching of their contents into the surrounding environment. While it is widely agreed that hazardous substances such as nickel and cobalt in the leachate can pose a threat to the environment and human health, the overall composition and toxicity of LIB leachate remain unclear. In this study, a chemical analysis of leachate from spent LIBs was conducted to identify its primary constituents. The ecotoxicological parameters of the model organism, rotifer Brachionus asplanchnoidis, were assessed to elucidate the toxicity of the LIB leachate. Subsequent experiments elucidated the impacts of the LIB leachate and its representative components on the malondialdehyde (MDA) level, antioxidant capacity, and enzyme activity of B. asplanchnoidis. The results indicate that both the LIB leachate and its components are harmful to individual rotifers due to the adverse effects of stress-induced disturbances in biochemical indicators, posing a threat to population development. The intensified poisoning phenomenon under combined stress suggests the presence of complex synergistic effects among the components of LIB leachate. Due to the likely environmental and biological hazards, LIBs should be strictly managed after disposal. Additionally, more economical and eco-friendly recycling and treatment technologies need to be developed and commercialized.
Collapse
Affiliation(s)
- Yi-Fan Feng
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| | - Yu Zhang
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| | - Run-Jia Yang
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| | - Si-Qi Li
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| | - Xiao-Jie Liu
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| | - Cui Han
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| | - Yi-Fu Xing
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| | - Jia-Xin Yang
- School of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province 210023, PR China.
| |
Collapse
|
11
|
Au WH, Miller-Fleming L, Sanchez-Martinez A, Lee JA, Twyning MJ, Prag HA, Raik L, Allen SP, Shaw PJ, Ferraiuolo L, Mortiboys H, Whitworth AJ. Activation of the Keap1/Nrf2 pathway suppresses mitochondrial dysfunction, oxidative stress, and motor phenotypes in C9orf72 ALS/FTD models. Life Sci Alliance 2024; 7:e202402853. [PMID: 38906677 PMCID: PMC11192839 DOI: 10.26508/lsa.202402853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/23/2024] Open
Abstract
Mitochondrial dysfunction is a common feature of C9orf72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD); however, it remains unclear whether this is a cause or consequence of the pathogenic process. Analysing multiple aspects of mitochondrial biology across several Drosophila models of C9orf72-ALS/FTD, we found morphology, oxidative stress, and mitophagy are commonly affected, which correlated with progressive loss of locomotor performance. Notably, only genetic manipulations that reversed the oxidative stress levels were also able to rescue C9orf72 locomotor deficits, supporting a causative link between mitochondrial dysfunction, oxidative stress, and behavioural phenotypes. Targeting the key antioxidant Keap1/Nrf2 pathway, we found that genetic reduction of Keap1 or pharmacological inhibition by dimethyl fumarate significantly rescued the C9orf72-related oxidative stress and motor deficits. Finally, mitochondrial ROS levels were also elevated in C9orf72 patient-derived iNeurons and were effectively suppressed by dimethyl fumarate treatment. These results indicate that mitochondrial oxidative stress is an important mechanistic contributor to C9orf72 pathogenesis, affecting multiple aspects of mitochondrial function and turnover. Targeting the Keap1/Nrf2 signalling pathway to combat oxidative stress represents a therapeutic strategy for C9orf72-related ALS/FTD.
Collapse
Affiliation(s)
- Wing Hei Au
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | | | - James Ak Lee
- Sheffield Institute for Translational Neuroscience (SITraN), School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | | | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Laura Raik
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Scott P Allen
- Sheffield Institute for Translational Neuroscience (SITraN), School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), School of Medicine and Population Health, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | | |
Collapse
|
12
|
Godoy JA, Mira RG, Inestrosa NC. Intracellular effects of lithium in aging neurons. Ageing Res Rev 2024; 99:102396. [PMID: 38942199 DOI: 10.1016/j.arr.2024.102396] [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: 04/15/2024] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Lithium therapy received approval during the 1970s, and it has been used for its antidepressant, antimanic, and anti-suicidal effects for acute and long-term prophylaxis and treatment of bipolar disorder (BPD). These properties have been well established; however, the molecular and cellular mechanisms remain controversial. In the past few years, many studies demonstrated that at the cellular level, lithium acts as a regulator of neurogenesis, aging, and Ca2+ homeostasis. At the molecular level, lithium modulates aging by inhibiting glycogen synthase kinase-3β (GSK-3β), and the phosphatidylinositol (PI) cycle; latter, lithium specifically inhibits inositol production, acting as a non-competitive inhibitor of inositol monophosphatase (IMPase). Mitochondria and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) have been related to lithium activity, and its regulation is mediated by GSK-3β degradation and inhibition. Lithium also impacts Ca2+ homeostasis in the mitochondria modulating the function of the lithium-permeable mitochondrial Na+-Ca2+exchanger (NCLX), affecting Ca2+ efflux from the mitochondrial matrix to the endoplasmic reticulum (ER). A close relationship between the protease Omi, GSK-3β, and PGC-1α has also been established. The purpose of this review is to summarize some of the intracellular mechanisms related to lithium activity and how, through them, neuronal aging could be controlled.
Collapse
Affiliation(s)
- Juan A Godoy
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo G Mira
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
13
|
Berner K, Oz N, Kaya A, Acharjee A, Berner J. mTORC1 activation in presumed classical monocytes: observed correlation with human size variation and neuropsychiatric disease. Aging (Albany NY) 2024; 16:11134-11150. [PMID: 39068671 PMCID: PMC11315394 DOI: 10.18632/aging.206033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Gain of function disturbances in nutrient sensing are likely the largest component in human age-related disease. Mammalian target of rapamycin complex 1 (mTORC1) activity affects health span and longevity. The drugs ketamine and rapamycin are effective against chronic pain and depression, and both affect mTORC1 activity. Our objective was to measure phosphorylated p70S6K, a marker for mTORC1 activity, in individuals with psychiatric disease to determine whether phosphorylated p70S6K could predict medication response. METHODS Twenty-seven females provided blood samples in which p70S6K and phosphorylated p70S6K were analyzed. Chart review gathered biometric measurements, clinical phenotypes, and medication response. Questionnaires assessed anxiety, depression, autism traits, and mitochondrial dysfunction, to determine neuropsychiatric disease profiles. Univariate and multivariate statistical analyses were used to identify predictors of medication response. RESULTS mTORC1 activity correlated highly with both classical biometrics (height, macrocephaly, pupil distance) and specific neuropsychiatric disease profiles (anxiety and autism). Across all cases, phosphorylated p70S6K was the best predictor for ketamine response, and also the best predictor for rapamycin response in a single instance. CONCLUSIONS The data illustrate the importance of mTORC1 activity in both observable body structure and medication response. This report suggests that a simple assay may allow cost-effective prediction of medication response.
Collapse
Affiliation(s)
- Karl Berner
- Woodinville Psychiatric Associates, Woodinville, WA 98072, USA
| | - Naci Oz
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
- Life Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Alaattin Kaya
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Animesh Acharjee
- Institute of Cancer and Genomics Sciences, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- MRC Health Data Research UK (HDR UK), London, UK
| | - Jon Berner
- Woodinville Psychiatric Associates, Woodinville, WA 98072, USA
| |
Collapse
|
14
|
Jans K, Lüersen K, von Frieling J, Roeder T, Rimbach G. Dietary sucrose determines the regulatory activity of lithium on gene expression and lifespan in Drosophila melanogaster. Aging (Albany NY) 2024; 16:9309-9333. [PMID: 38862239 PMCID: PMC11210232 DOI: 10.18632/aging.205933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/10/2024] [Indexed: 06/13/2024]
Abstract
The amount of dietary sugars and the administration of lithium both impact the lifespan of the fruit fly Drosophila melanogaster. It is noteworthy that lithium is attributed with insulin-like activity as it stimulates protein kinase B/Akt and suppresses the activity of glycogen synthase kinase-3 (GSK-3). However, its interaction with dietary sugar has largely remained unexplored. Therefore, we investigated the effects of lithium supplementation on known lithium-sensitive parameters in fruit flies, such as lifespan, body composition, GSK-3 phosphorylation, and the transcriptome, while varying the dietary sugar concentration. For all these parameters, we observed that the efficacy of lithium was significantly influenced by the sucrose content in the diet. Overall, we found that lithium was most effective in enhancing longevity and altering body composition when added to a low-sucrose diet. Whole-body RNA sequencing revealed a remarkably similar transcriptional response when either increasing dietary sucrose from 1% to 10% or adding 1 mM LiCl to a 1% sucrose diet, characterized by a substantial overlap of nearly 500 differentially expressed genes. Hence, dietary sugar supply is suggested as a key factor in understanding lithium bioactivity, which could hold relevance for its therapeutic applications.
Collapse
Affiliation(s)
- Katharina Jans
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany
| | - Kai Lüersen
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany
| | - Jakob von Frieling
- Division of Molecular Physiology, Institute of Zoology, University of Kiel, Kiel D-24118, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, University of Kiel, Kiel D-24118, Germany
| | - Gerald Rimbach
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel D-24118, Germany
| |
Collapse
|
15
|
Chen M, Fang Y, Ge Y, Qiu S, Dworkin L, Gong R. The redox-sensitive GSK3β is a key regulator of glomerular podocyte injury in type 2 diabetic kidney disease. Redox Biol 2024; 72:103127. [PMID: 38527400 PMCID: PMC10979123 DOI: 10.1016/j.redox.2024.103127] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/06/2024] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
Emerging evidence suggests that GSK3β, a redox-sensitive transducer downstream of insulin signaling, acts as a convergent point for myriad pathways implicated in kidney injury, repair, and regeneration. However, its role in diabetic kidney disease remains controversial. In cultured glomerular podocytes, exposure to a milieu of type 2 diabetes elicited prominent signs of podocyte injury and degeneration, marked by loss of homeostatic marker proteins like synaptopodin, actin cytoskeleton disruption, oxidative stress, apoptosis, and stress-induced premature senescence, as shown by increased staining for senescence-associated β-galactosidase activity, amplified formation of γH2AX foci, and elevated expression of mediators of senescence signaling, like p21 and p16INK4A. These degenerative changes coincided with GSK3β hyperactivity, as evidenced by GSK3β overexpression and reduced inhibitory phosphorylation of GSK3β, and were averted by tideglusib, a highly-selective small molecule inhibitor of GSK3β. In agreement, post-hoc analysis of a publicly-available glomerular transcriptomics dataset from patients with type 2 diabetic nephropathy revealed that the curated diabetic nephropathy-related gene set was enriched in high GSK3β expression group. Mechanistically, GSK3β-modulated nuclear factor Nrf2 signaling is involved in diabetic podocytopathy, because GSK3β knockdown reinforced Nrf2 antioxidant response and suppressed oxidative stress, resulting in an improvement in podocyte injury and senescence. Conversely, ectopic expression of the constitutively active mutant of GSK3β impaired Nrf2 antioxidant response and augmented oxidative stress, culminating in an exacerbated diabetic podocyte injury and senescence. Moreover, IRS-1 was found to be a cognate substrate of GSK3β for phosphorylation at IRS-1S332, which negatively regulates IRS-1 activity. GSK3β hyperactivity promoted IRS-1 phosphorylation, denoting a desensitized insulin signaling. Consistently, in vivo in db/db mice with diabetic nephropathy, GSK3β was hyperactive in glomerular podocytes, associated with IRS-1 hyperphosphorylation, impaired Nrf2 response and premature senescence. Our finding suggests that GSK3β is likely a novel therapeutic target for treating type 2 diabetic glomerular injury.
Collapse
Affiliation(s)
- Mengxuan Chen
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Yudong Fang
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Yan Ge
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Shuhao Qiu
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Lance Dworkin
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA
| | - Rujun Gong
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA; Center for Diabetes and Endocrine Research, University of Toledo Medical Center, Toledo, OH, USA.
| |
Collapse
|
16
|
Chen PH, Lee TW, Liu SH, Huynh TV, Chung CC, Yeh YH, Kao YH, Chen YJ. Lithium downregulates phosphorylated acetyl‑CoA carboxylase 2 and attenuates mitochondrial fatty acid utilization and oxidative stress in cardiomyocytes. Exp Ther Med 2024; 27:126. [PMID: 38414784 PMCID: PMC10895620 DOI: 10.3892/etm.2024.12413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/11/2024] [Indexed: 02/29/2024] Open
Abstract
Acetyl-CoA carboxylase 2 plays a crucial role in regulating mitochondrial fatty acid oxidation in cardiomyocytes. Lithium, a monovalent cation known for its cardioprotective potential, has been investigated for its influence on mitochondrial bioenergetics. The present study explored whether lithium modulated acetyl-CoA carboxylase 2 and mitochondrial fatty acid metabolism in cardiomyocytes and the potential therapeutic applications of lithium in alleviating metabolic stress. Mitochondrial bioenergetic function, fatty acid oxidation, reactive oxygen species production, membrane potential and the expression of proteins involved in fatty acid metabolism in H9c2 cardiomyocytes treated with LiCl for 48 h was measured by using a Seahorse extracellular flux analyzer, fluorescence microscopy and western blotting. Small interfering RNA against glucose transporter type 4 was transfected into H9c2 cardiomyocytes for 48 h to induce metabolic stress mimicking insulin resistance. The results revealed that LiCl at a concentration of 0.3 mM (but not at a concentration of 0.1 or 1.0 mM) upregulated the expression of phosphorylated (p-)glycogen synthase kinase-3 beta and downregulated the expression of p-acetyl-CoA carboxylase 2 but did not affect the expression of adenosine monophosphate-activated protein kinase or calcineurin. Cotreatment with TWS119 (8 µM) and LiCl (0.3 mM) downregulated p-acetyl-CoA carboxylase 2 expression to a similar extent as did treatment with TWS119 (8 µM) alone. Moreover, LiCl (0.3 mM) inhibited mitochondrial fatty acid oxidation, improved coupling efficiency and the cellular respiratory control ratio, hindered reactive oxygen species production and proton leakage and restored mitochondrial membrane potential in glucose transporter type 4 knockdown-H9c2 cardiomyocytes. These findings suggested that low therapeutic levels of lithium can downregulate p-acetyl-CoA carboxylase 2, thus reducing mitochondrial fatty acid oxidation and oxidative stress in cardiomyocytes.
Collapse
Affiliation(s)
- Pao-Huan Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Psychiatry, Taipei Medical University Hospital, Taipei 11031, Taiwan, R.O.C
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| | - Shuen-Hsin Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan, R.O.C
| | - Tin Van Huynh
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Interventional Cardiology, Thong Nhat Hospital, Ho Chi Minh City 700000, Vietnam
| | - Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| | - Yung-Hsin Yeh
- Division of Cardiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan, R.O.C
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan, R.O.C
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan, R.O.C
| |
Collapse
|
17
|
Jans K, Lüersen K, von Frieling J, Roeder T, Rimbach G. Dietary lithium stimulates female fecundity in Drosophila melanogaster. Biofactors 2024; 50:326-346. [PMID: 37706424 DOI: 10.1002/biof.2007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/28/2023] [Indexed: 09/15/2023]
Abstract
The trace element lithium exerts a versatile bioactivity in humans, to some extend overlapping with in vivo findings in the model organism Drosophila melanogaster. A potentially essential function of lithium in reproduction has been suggested since the 1980s and multiple studies have since been published postulating a regulatory role of lithium in female gametogenesis. However, the impact of lithium on fruit fly egg production has not been at the center of attention to date. In the present study, we report that dietary lithium (0.1-5.0 mM LiCl) substantially improved life time egg production in D. melanogaster w1118 females, with a maximum increase of plus 45% when supplementing 1.0 mM LiCl. This phenomenon was not observed in the insulin receptor mutant InRE19, indicating a potential involvement of insulin-like signaling in the lithium-mediated fecundity boost. Analysis of the whole-body and ovarian transcriptome revealed that dietary lithium affects the mRNA levels of genes encoding proteins related to processes of follicular maturation. To the best of our knowledge, this is the first report on dietary lithium acting as an in vivo fecundity stimulant in D. melanogaster, further supporting the suggested benefit of the trace element in female reproduction.
Collapse
Affiliation(s)
- Katharina Jans
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Kai Lüersen
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Jakob von Frieling
- Division of Molecular Physiology, Institute of Zoology, University of Kiel, Kiel, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, University of Kiel, Kiel, Germany
| | - Gerald Rimbach
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
18
|
Chen W, Zhang P, Ye L, Yao J, Wang Z, Liu J, Qin X, Wang Z. Concentration-dependent effects of lithium on Daphnia magna: Life-history profiles and integrated biomarker response implementation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169866. [PMID: 38190914 DOI: 10.1016/j.scitotenv.2023.169866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/02/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024]
Abstract
The growing use of lithium (Li) in industrial and energy applications and increasing demand worldwide has inevitably resulted in its wide dispersal, representing a significant threat to aquatic systems. Unfortunately, as a ubiquitous emerging contaminant, the comprehensive toxicological information regarding Li at multifarious levels is limited. To diminish this gap, this work was focused to explore Li-induced cascading effects on Daphnia magna as a key species in freshwater ecosystems. Specifically, the organisms were chronically exposed to gradient Li concentrations with emphasis on characterizing life-history traits from individual to population scale, primarily as observed by a markedly concentration-dependent decrease along exposure gradients. In parallel, a robust set of biomarkers relating to energy reserves, antioxidant and biotransformation enzymes, cellular damage, ionoregulation and neurotoxicity were assayed for further understanding potential underlying mechanisms. As a result, biomarker alterations were characterized by significant decreases in energy storage and enzymatic profiles of antioxidant and biotransformation systems, not only triggering an imbalance between reactive oxygen species (ROS) generation and elimination under Li exposure, but compromising the fecundity fitness of phenotypical costs. In contrast, malondialdehyde (MDA) levels were remarkably enhanced as a consequence of inefficient antioxidant and biotransformation capacity leading to lipid peroxidation (LPO). Additionally, Li exerted a dose-dependent biphasic effect on the activities of superoxide dismutase (SOD), Na+,K+-ATPase and acetylcholinesterase (AChE) by interfering with inherent balance. In terms of responsive patterns and dose-effect trends, the integrated biomarker response indices (IBRv2) and star plots were consistent with the differences in biomarker profiles, not only presenting comprehensively biological effects in a visualized form, but signaling the importance of progressive induced changes in an integrative way. Overall, these findings highlighted the need for elucidating Li-produced impacts from a comprehensive perspective, providing valuable insights into better understanding the toxicity of Li in relation to aquatic ecosystem functioning and ecological relevance.
Collapse
Affiliation(s)
- Wenqing Chen
- Jiangxi Provincial Key Laboratory of Environmental Pollution Prevention and Control in Mining and Metallurgy, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China; School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China
| | - Pengze Zhang
- Jiangxi Provincial Key Laboratory of Environmental Pollution Prevention and Control in Mining and Metallurgy, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China; School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China
| | - Li Ye
- Jiangxi Provincial Key Laboratory of Environmental Pollution Prevention and Control in Mining and Metallurgy, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China; School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China
| | - Junqiang Yao
- Jiangxi Provincial Key Laboratory of Environmental Pollution Prevention and Control in Mining and Metallurgy, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China; School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China
| | - Zaosheng Wang
- Jiangxi Provincial Key Laboratory of Environmental Pollution Prevention and Control in Mining and Metallurgy, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China; School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China.
| | - Jun Liu
- Jiangxi Provincial Key Laboratory of Environmental Pollution Prevention and Control in Mining and Metallurgy, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China; School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China
| | - Xiaohai Qin
- School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China
| | - Zhaoru Wang
- School of Resource and Environment Engineering, Jiangxi University of Science and Technology, 156 Kejia Boulevard, Ganzhou, Jiangxi 341000, China
| |
Collapse
|
19
|
Durante LDS, Hollmann G, Nazari EM. Impact of exposure to glyphosate-based herbicide on morphological and physiological parameters in embryonic and larval development of zebrafish. ENVIRONMENTAL TOXICOLOGY 2024; 39:1822-1835. [PMID: 38083805 DOI: 10.1002/tox.24024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 07/20/2023] [Accepted: 10/31/2023] [Indexed: 02/08/2024]
Abstract
Glyphosate-based herbicides (GBH) have been commonly used in agriculture to inhibit weed growth and increase yields. However, due to the high solubility of these herbicides in water, they can reach aquatic environments, by infiltration, erosion, and/or lixiviation, affecting non target organisms. Thus, this study aimed to characterize the toxicity of GBH Roundup WG® (RWG®) during the embryonic and larval development of Danio rerio. Embryos (3 hours post fertilization, hpf-until hatching) and larvae (3 days post fertilization, dpf to 6 dpf) were exposed to concentrations of 0.065 and 6.5 mg L-1 . They were evaluated for survival, hatching, spontaneous movements, heartbeat, morphology, and morphometry by in vivo photographs in microscope, cell proliferation and apoptosis by immunohistochemistry, and exploratory behavior and phototropism by video recording. Our results showed an increase in embryo and larvae mortality in those exposed to 0.065 mg L-1 , as well as a reduction in spontaneous embryo movements. The larval heartbeats showed a decrease at 4 dpf in the group exposed to 0.065 mg L-1 and an increase at 5 and 6 dpf in both exposed groups. Cell proliferation was reduced in both groups exposed in embryos and only in the 0.065 mg L-1 group in larvae, while cell death increased in embryos exposed to 6.5 mg L-1 . These results demonstrated the toxic effect of low concentrations of the herbicide RWG® during embryonic and larval development of non target organisms, as well as the importance of constantly reviewing acceptable limits for exposure in natural environments.
Collapse
Affiliation(s)
- Laíse da Silva Durante
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Programa de Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina - UFSC, Florianópolis, Brazil
| | - Gabriela Hollmann
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Programa de Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina - UFSC, Florianópolis, Brazil
| | - Evelise Maria Nazari
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Programa de Pós-Graduação em Biologia Celular e do Desenvolvimento, Universidade Federal de Santa Catarina - UFSC, Florianópolis, Brazil
| |
Collapse
|
20
|
Tziortzouda P, Steyaert J, Scheveneels W, Sicart A, Stoklund Dittlau K, Barbosa Correia AM, Burg T, Pal A, Hermann A, Van Damme P, Moens TG, Van Den Bosch L. PP2A and GSK3 act as modifiers of FUS-ALS by modulating mitochondrial transport. Acta Neuropathol 2024; 147:41. [PMID: 38363426 PMCID: PMC10873455 DOI: 10.1007/s00401-024-02689-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/22/2023] [Accepted: 01/11/2024] [Indexed: 02/17/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease which currently lacks effective treatments. Mutations in the RNA-binding protein FUS are a common cause of familial ALS, accounting for around 4% of the cases. Understanding the mechanisms by which mutant FUS becomes toxic to neurons can provide insight into the pathogenesis of both familial and sporadic ALS. We have previously observed that overexpression of wild-type or ALS-mutant FUS in Drosophila motor neurons is toxic, which allowed us to screen for novel genetic modifiers of the disease. Using a genome-wide screening approach, we identified Protein Phosphatase 2A (PP2A) and Glycogen Synthase Kinase 3 (GSK3) as novel modifiers of FUS-ALS. Loss of function or pharmacological inhibition of either protein rescued FUS-associated lethality in Drosophila. Consistent with a conserved role in disease pathogenesis, pharmacological inhibition of both proteins rescued disease-relevant phenotypes, including mitochondrial trafficking defects and neuromuscular junction failure, in patient iPSC-derived spinal motor neurons (iPSC-sMNs). In FUS-ALS flies, mice, and human iPSC-sMNs, we observed reduced GSK3 inhibitory phosphorylation, suggesting that FUS dysfunction results in GSK3 hyperactivity. Furthermore, we found that PP2A acts upstream of GSK3, affecting its inhibitory phosphorylation. GSK3 has previously been linked to kinesin-1 hyperphosphorylation. We observed this in both flies and iPSC-sMNs, and we rescued this hyperphosphorylation by inhibiting GSK3 or PP2A. Moreover, increasing the level of kinesin-1 expression in our Drosophila model strongly rescued toxicity, confirming the relevance of kinesin-1 hyperphosphorylation. Our data provide in vivo evidence that PP2A and GSK3 are disease modifiers, and reveal an unexplored mechanistic link between PP2A, GSK3, and kinesin-1, that may be central to the pathogenesis of FUS-ALS and sporadic forms of the disease.
Collapse
Affiliation(s)
- Paraskevi Tziortzouda
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
| | - Jolien Steyaert
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
| | - Wendy Scheveneels
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
| | - Adria Sicart
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
| | - Katarina Stoklund Dittlau
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
| | - Adriana Margarida Barbosa Correia
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
- Instituto Superior Técnico-Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Thibaut Burg
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
| | - Arun Pal
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden Rossendorf, 01328, Dresden, Germany
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, 01307, Dresden, Germany
| | - Andreas Hermann
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, 01307, Dresden, Germany
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, 18147, Rostock, Germany
- Deutsches Zentrum Fur Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147, Rostock, Germany
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Thomas G Moens
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium.
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, PB 602, 3000, Leuven, Belgium.
| |
Collapse
|
21
|
Calabrese EJ, Nascarella M, Pressman P, Hayes AW, Dhawan G, Kapoor R, Calabrese V, Agathokleous E. Hormesis determines lifespan. Ageing Res Rev 2024; 94:102181. [PMID: 38182079 DOI: 10.1016/j.arr.2023.102181] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/27/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
This paper addresses how long lifespan can be extended via multiple interventions, such as dietary supplements [e.g., curcumin, resveratrol, sulforaphane, complex phytochemical mixtures (e.g., Moringa, Rhodiola)], pharmaceutical agents (e.g., metformin), caloric restriction, intermittent fasting, exercise and other activities. This evaluation was framed within the context of hormesis, a biphasic dose response with specific quantitative features describing the limits of biological/phenotypic plasticity for integrative biological endpoints (e.g., cell proliferation, memory, fecundity, growth, tissue repair, stem cell population expansion/differentiation, longevity). Evaluation of several hundred lifespan extending agents using yeast, nematode (Caenorhabditis elegans), multiple insect and other invertebrate and vertebrate models (e.g., fish, rodents), revealed they responded in a manner [average (mean/median) and maximum lifespans] consistent with the quantitative features [i.e., 30-60% greater at maximum (Hormesis Rule)] of the hormetic dose response. These lifespan extension features were independent of biological model, inducing agent, endpoints measured and mechanism. These findings indicate that hormesis describes the capacity to extend life via numerous agents and activities and that the magnitude of lifespan extension is modest, in the percentage, not fold, range. These findings have important implications for human aging, genetic diseases/environmental stresses and lifespan extension, as well as public health practices and long-term societal resource planning.
Collapse
Affiliation(s)
- Edward J Calabrese
- School of Public Health and Health Sciences; University of Massachusetts, Morrill I - Room N344, Amherst, MA 01003, USA.
| | - Marc Nascarella
- Mass College of Pharmacy and Health Sciences University; School of Arts and Sciences, 179 Longwood Avenue, Boston, MA 02115, USA
| | - Peter Pressman
- University of Maine, 5728 Fernald Hall, Room 201, Orono, ME 04469, USA
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management; College of Public Health; University of South Florida, Tampa, FL, USA
| | - Gaurav Dhawan
- Sri Guru Ram Das (SGRD) University of Health Sciences, Amritsar, India
| | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, CT, USA
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine University of Catania, Via Santa Sofia 97, Catania 95123, Italy
| | - Evgenios Agathokleous
- School of Ecology and Applied Meteorology; Nanjing University of Information Science & Technology; Nanjing 210044, China
| |
Collapse
|
22
|
Diniz BS, Seitz-Holland J, Sehgal R, Kasamoto J, Higgins-Chen AT, Lenze E. Geroscience-Centric Perspective for Geriatric Psychiatry: Integrating Aging Biology With Geriatric Mental Health Research. Am J Geriatr Psychiatry 2024; 32:1-16. [PMID: 37845116 PMCID: PMC10841054 DOI: 10.1016/j.jagp.2023.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/30/2023] [Accepted: 09/14/2023] [Indexed: 10/18/2023]
Abstract
The geroscience hypothesis asserts that physiological aging is caused by a small number of biological pathways. Despite the explosion of geroscience research over the past couple of decades, the research on how serious mental illnesses (SMI) affects the biological aging processes is still in its infancy. In this review, we aim to provide a critical appraisal of the emerging literature focusing on how we measure biological aging systematically, and in the brain and how SMIs affect biological aging measures in older adults. We will also review recent developments in the field of cellular senescence and potential targets for interventions for SMIs in older adults, based on the geroscience hypothesis.
Collapse
Affiliation(s)
- Breno S Diniz
- UConn Center on Aging & Department of Psychiatry (BSD), School of Medicine, University of Connecticut Health Center, Farmington, CT.
| | - Johanna Seitz-Holland
- Department of Psychiatry (JSH), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Psychiatry (JSH), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Raghav Sehgal
- Program in Computational Biology and Bioinformatics (RS, JK), Yale University, New Haven, CT
| | - Jessica Kasamoto
- Program in Computational Biology and Bioinformatics (RS, JK), Yale University, New Haven, CT
| | - Albert T Higgins-Chen
- Department of Psychiatry (ATHC), Yale University School of Medicine, New Haven, CT; Department of Pathology (ATHC), Yale University School of Medicine, New Haven, CT
| | - Eric Lenze
- Department of Psychiatry (EL), School of Medicine, Washington University at St. Louis, St. Louis, MO
| |
Collapse
|
23
|
Bharat V, Durairaj AS, Vanhauwaert R, Li L, Muir CM, Chandra S, Kwak CS, Le Guen Y, Nandakishore P, Hsieh CH, Rensi SE, Altman RB, Greicius MD, Feng L, Wang X. A mitochondrial inside-out iron-calcium signal reveals drug targets for Parkinson's disease. Cell Rep 2023; 42:113544. [PMID: 38060381 PMCID: PMC10804639 DOI: 10.1016/j.celrep.2023.113544] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/11/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023] Open
Abstract
Dysregulated iron or Ca2+ homeostasis has been reported in Parkinson's disease (PD) models. Here, we discover a connection between these two metals at the mitochondria. Elevation of iron levels causes inward mitochondrial Ca2+ overflow, through an interaction of Fe2+ with mitochondrial calcium uniporter (MCU). In PD neurons, iron accumulation-triggered Ca2+ influx across the mitochondrial surface leads to spatially confined Ca2+ elevation at the outer mitochondrial membrane, which is subsequently sensed by Miro1, a Ca2+-binding protein. A Miro1 blood test distinguishes PD patients from controls and responds to drug treatment. Miro1-based drug screens in PD cells discover Food and Drug Administration-approved T-type Ca2+-channel blockers. Human genetic analysis reveals enrichment of rare variants in T-type Ca2+-channel subtypes associated with PD status. Our results identify a molecular mechanism in PD pathophysiology and drug targets and candidates coupled with a convenient stratification method.
Collapse
Affiliation(s)
- Vinita Bharat
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aarooran S Durairaj
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Roeland Vanhauwaert
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Li Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Colin M Muir
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Graduate Program of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sujyoti Chandra
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chulhwan S Kwak
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Institut du Cerveau - Paris Brain Institute - ICM, 75013 Paris, France
| | | | - Chung-Han Hsieh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefano E Rensi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Russ B Altman
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liang Feng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinnan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
24
|
Janssens GE, Grevendonk L, Schomakers BV, Perez RZ, van Weeghel M, Schrauwen P, Hoeks J, Houtkooper RH. A metabolomic signature of decelerated physiological aging in human plasma. GeroScience 2023; 45:3147-3164. [PMID: 37259015 PMCID: PMC10643795 DOI: 10.1007/s11357-023-00827-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/13/2023] [Indexed: 06/02/2023] Open
Abstract
The degenerative processes that occur during aging increase the risk of disease and impaired health. Meanwhile, interventions that target aging to promote healthy longevity are gaining interest, both academically and in the public. While nutritional and physical interventions exist, efficacy is often difficult to determine. It is therefore imperative that an aging score measuring the biological aging process is available to the wider public. However, simple, interpret, and accessible biological aging scores are lacking. Here, we developed PhysiAge, a physiological aging score based on five accessible parameters that have influence on or reflect the aging process: (1) average daily step count, (2) blood glucose, (3) systolic blood pressure, (4) sex, and (5) age. Here, we found that compared to calendar age alone, PhysiAge better predicts mortality, as well as established muscle aging markers such as decrease in NAD+ levels, increase in oxidative stress, and decline in physical functioning. In order to demonstrate the usefulness of PhysiAge in identifying relevant factors associated with decelerated aging, we calculated PhysiAges for a cohort of aged individuals and obtained mass spectrometry-based blood plasma metabolomic profiles for each individual. Here, we identified a metabolic signature of decelerated aging, which included components of the TCA cycle, including malate, citrate, and isocitrate. Higher abundance of these metabolites was associated with decelerated aging, in line with supplementation studies in model organisms. PhysiAge represents an accessible way for people to track and intervene in their aging trajectories, and identifies a metabolic signature of decelerated aging in human blood plasma, which can be further studied for its causal involvement in human aging.
Collapse
Affiliation(s)
- Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.
| | - Lotte Grevendonk
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6200 MD, Maastricht, The Netherlands
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
| | - Bauke V Schomakers
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ruben Zapata Perez
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Faculty of Health Sciences, UCAM - Universidad Católica de Murcia, 30107, Murcia, Spain
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6200 MD, Maastricht, The Netherlands
- TI Food and Nutrition, PO Box 557, 6700 AN, Wageningen, The Netherlands
| | - Joris Hoeks
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Pun FW, Leung GHD, Leung HW, Rice J, Schmauck‐Medina T, Lautrup S, Long X, Liu BHM, Wong CW, Ozerov IV, Aliper A, Ren F, Rosenberg AJ, Agrawal N, Izumchenko E, Fang EF, Zhavoronkov A. A comprehensive AI-driven analysis of large-scale omic datasets reveals novel dual-purpose targets for the treatment of cancer and aging. Aging Cell 2023; 22:e14017. [PMID: 37888486 PMCID: PMC10726874 DOI: 10.1111/acel.14017] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
As aging and tumorigenesis are tightly interconnected biological processes, targeting their common underlying driving pathways may induce dual-purpose anti-aging and anti-cancer effects. Our transcriptomic analyses of 16,740 healthy samples demonstrated tissue-specific age-associated gene expression, with most tumor suppressor genes downregulated during aging. Furthermore, a large-scale pan-cancer analysis of 11 solid tumor types (11,303 cases and 4431 control samples) revealed that many cellular processes, such as protein localization, DNA replication, DNA repair, cell cycle, and RNA metabolism, were upregulated in cancer but downregulated in healthy aging tissues, whereas pathways regulating cellular senescence were upregulated in both aging and cancer. Common cancer targets were identified by the AI-driven target discovery platform-PandaOmics. Age-associated cancer targets were selected and further classified into four groups based on their reported roles in lifespan. Among the 51 identified age-associated cancer targets with anti-aging experimental evidence, 22 were proposed as dual-purpose targets for anti-aging and anti-cancer treatment with the same therapeutic direction. Among age-associated cancer targets without known lifespan-regulating activity, 23 genes were selected based on predicted dual-purpose properties. Knockdown of histone demethylase KDM1A, one of these unexplored candidates, significantly extended lifespan in Caenorhabditis elegans. Given KDM1A's anti-cancer activities reported in both preclinical and clinical studies, our findings propose KDM1A as a promising dual-purpose target. This is the first study utilizing an innovative AI-driven approach to identify dual-purpose target candidates for anti-aging and anti-cancer treatment, supporting the value of AI-assisted target identification for drug discovery.
Collapse
Affiliation(s)
| | | | | | - Jared Rice
- Department of Clinical Molecular BiologyUniversity of Oslo and Akershus University HospitalLørenskogNorway
| | - Tomas Schmauck‐Medina
- Department of Clinical Molecular BiologyUniversity of Oslo and Akershus University HospitalLørenskogNorway
| | - Sofie Lautrup
- Department of Clinical Molecular BiologyUniversity of Oslo and Akershus University HospitalLørenskogNorway
| | - Xi Long
- Insilico Medicine Hong Kong Ltd.Hong KongChina
| | | | | | | | - Alex Aliper
- Insilico Medicine AI Ltd.Masdar CityUnited Arab Emirates
| | - Feng Ren
- Insilico Medicine Shanghai Ltd.ShanghaiChina
| | - Ari J. Rosenberg
- Department of Medicine, Section of Hematology and OncologyUniversity of ChicagoChicagoIllinoisUSA
| | - Nishant Agrawal
- Department of SurgeryUniversity of ChicagoChicagoIllinoisUSA
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and OncologyUniversity of ChicagoChicagoIllinoisUSA
| | - Evandro F. Fang
- Department of Clinical Molecular BiologyUniversity of Oslo and Akershus University HospitalLørenskogNorway
- The Norwegian Centre On Healthy Ageing (NO‐Age)OsloNorway
| | - Alex Zhavoronkov
- Insilico Medicine Hong Kong Ltd.Hong KongChina
- Insilico Medicine AI Ltd.Masdar CityUnited Arab Emirates
- Buck Institute for Research on AgingNovatoCaliforniaUSA
| |
Collapse
|
26
|
Erdem B, Arslan OC, Sevin S, Gozen AG, Agosto-Rivera JL, Giray T, Alemdar H. Effects of lithium on locomotor activity and circadian rhythm of honey bees. Sci Rep 2023; 13:19861. [PMID: 37963948 PMCID: PMC10646147 DOI: 10.1038/s41598-023-46777-7] [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: 05/18/2023] [Accepted: 11/04/2023] [Indexed: 11/16/2023] Open
Abstract
Lithium has been considered a potential acaricidal agent against the honey bee (Apis mellifera) parasite Varroa. It is known that lithium suppresses elevated activity and regulates circadian rhythms and light response when administered to humans as a primary therapeutic chemical for bipolar disorder and to other bipolar syndrome model organisms, given the crucial role of timing in the bee's foraging activity and the alternating sunlight vs dark colony environment bees are exposed, we explored the influence of lithium on locomotor activity (LMA) and circadian rhythm of honey bees. We conducted acute and chronic lithium administration experiments, altering light conditions and lithium doses to assess LMA and circadian rhythm changes. We fed bees one time 10 μl sucrose solution with 0, 50, 150, and 450 mM LiCl in the acute application experiment and 0, 1, 5, and 10 mmol/kg LiCl ad libitum in bee candy in the chronic application experiment. Both acute and chronic lithium treatments significantly decreased the induced LMA under constant light. Chronic lithium treatment disrupted circadian rhythmicity in constant darkness. The circadian period was lengthened by lithium treatment under constant light. We discuss the results in the context of Varroa control and lithium's effect on bipolar disorder.
Collapse
Affiliation(s)
- Babur Erdem
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey.
- Center for Robotics and Artificial Intelligence (ROMER), Middle East Technical University, Ankara, Turkey.
| | - Okan Can Arslan
- Center for Robotics and Artificial Intelligence (ROMER), Middle East Technical University, Ankara, Turkey
| | - Sedat Sevin
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ankara University, Ankara, Turkey
| | - Ayse Gul Gozen
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | | | - Tugrul Giray
- Department of Biology, University of Puerto Rico, Rio Piedras, Puerto Rico
| | - Hande Alemdar
- Department of Computer Engineering, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
27
|
Srivastava V, Gross E. Mitophagy-promoting agents and their ability to promote healthy-aging. Biochem Soc Trans 2023; 51:1811-1846. [PMID: 37650304 PMCID: PMC10657188 DOI: 10.1042/bst20221363] [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: 04/13/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023]
Abstract
The removal of damaged mitochondrial components through a process called mitochondrial autophagy (mitophagy) is essential for the proper function of the mitochondrial network. Hence, mitophagy is vital for the health of all aerobic animals, including humans. Unfortunately, mitophagy declines with age. Many age-associated diseases, including Alzheimer's and Parkinson's, are characterized by the accumulation of damaged mitochondria and oxidative damage. Therefore, activating the mitophagy process with small molecules is an emerging strategy for treating multiple aging diseases. Recent studies have identified natural and synthetic compounds that promote mitophagy and lifespan. This article aims to summarize the existing knowledge about these substances. For readers' convenience, the knowledge is presented in a table that indicates the chemical data of each substance and its effect on lifespan. The impact on healthspan and the molecular mechanism is reported if known. The article explores the potential of utilizing a combination of mitophagy-inducing drugs within a therapeutic framework and addresses the associated challenges of this strategy. Finally, we discuss the process that balances mitophagy, i.e. mitochondrial biogenesis. In this process, new mitochondrial components are generated to replace the ones cleared by mitophagy. Furthermore, some mitophagy-inducing substances activate biogenesis (e.g. resveratrol and metformin). Finally, we discuss the possibility of combining mitophagy and biogenesis enhancers for future treatment. In conclusion, this article provides an up-to-date source of information about natural and synthetic substances that activate mitophagy and, hopefully, stimulates new hypotheses and studies that promote healthy human aging worldwide.
Collapse
Affiliation(s)
- Vijigisha Srivastava
- Faculty of Medicine, IMRIC Department of Biochemistry and Molecular Biology, The Hebrew University of Jerusalem, PO Box 12271, Jerusalem, Israel
| | - Einav Gross
- Faculty of Medicine, IMRIC Department of Biochemistry and Molecular Biology, The Hebrew University of Jerusalem, PO Box 12271, Jerusalem, Israel
| |
Collapse
|
28
|
Parkhitko AA, Filine E, Tatar M. Combinatorial interventions in aging. NATURE AGING 2023; 3:1187-1200. [PMID: 37783817 PMCID: PMC11194689 DOI: 10.1038/s43587-023-00489-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/15/2023] [Indexed: 10/04/2023]
Abstract
Insight on the underlying mechanisms of aging will advance our ability to extend healthspan, treat age-related pathology and improve quality of life. Multiple genetic and pharmacological manipulations extend longevity in different species, yet monotherapy may be relatively inefficient, and we have limited data on the effect of combined interventions. Here we summarize interactions between age-related pathways and discuss strategies to simultaneously retard these in different organisms. In some cases, combined manipulations additively increase their impact on common hallmarks of aging and lifespan, suggesting they quantitatively participate within the same pathway. In other cases, interactions affect different hallmarks, suggesting their joint manipulation may independently maximize their effects on lifespan and healthy aging. While most interaction studies have been conducted with invertebrates and show varying levels of translatability, the conservation of pro-longevity pathways offers an opportunity to identify 'druggable' targets relevant to multiple human age-associated pathologies.
Collapse
Affiliation(s)
- Andrey A Parkhitko
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA.
| | - Elizabeth Filine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Marc Tatar
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI, USA.
| |
Collapse
|
29
|
Deline ML, Straub J, Patel M, Subba P, Grashei M, van Heijster FHA, Pirkwieser P, Somoza V, Livingstone JD, Beazely M, Kendall B, Gingras MJP, Leonenko Z, Höschen C, Harrington G, Kuellmer K, Bian W, Schilling F, Fisher MPA, Helgeson ME, Fromme T. Lithium isotopes differentially modify mitochondrial amorphous calcium phosphate cluster size distribution and calcium capacity. Front Physiol 2023; 14:1200119. [PMID: 37781224 PMCID: PMC10540846 DOI: 10.3389/fphys.2023.1200119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Lithium is commonly prescribed as a mood stabilizer in a variety of mental health conditions, yet its molecular mode of action is incompletely understood. Many cellular events associated with lithium appear tied to mitochondrial function. Further, recent evidence suggests that lithium bioactivities are isotope specific. Here we focus on lithium effects related to mitochondrial calcium handling. Lithium protected against calcium-induced permeability transition and decreased the calcium capacity of liver mitochondria at a clinically relevant concentration. In contrast, brain mitochondrial calcium capacity was increased by lithium. Surprisingly, 7Li acted more potently than 6Li on calcium capacity, yet 6Li was more effective at delaying permeability transition. The size distribution of amorphous calcium phosphate colloids formed in vitro was differentially affected by lithium isotopes, providing a mechanistic basis for the observed isotope specific effects on mitochondrial calcium handling. This work highlights a need to better understand how mitochondrial calcium stores are structurally regulated and provides key considerations for future formulations of lithium-based therapeutics.
Collapse
Affiliation(s)
- Marshall L. Deline
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Joshua Straub
- Department of Physics, University of California, Santa Barbara, CA, United States
| | - Manisha Patel
- Department of Physics, University of California, Santa Barbara, CA, United States
| | - Pratigya Subba
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Martin Grashei
- Department of Nuclear Medicine, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Frits H. A. van Heijster
- Department of Nuclear Medicine, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Philip Pirkwieser
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Veronika Somoza
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chair of Nutritional Systems Biology, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | | | - Michael Beazely
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Brian Kendall
- Department of Earth and Environmental Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Michel J. P. Gingras
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
- CIFAR, MaRS Centre, Toronto, ON, Canada
| | - Zoya Leonenko
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Carmen Höschen
- Chair of Soil Science, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Gertraud Harrington
- Chair of Soil Science, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Katharina Kuellmer
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Wangqing Bian
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthew P. A. Fisher
- Department of Physics, University of California, Santa Barbara, CA, United States
| | - Matthew E. Helgeson
- Department of Chemical Engineering, University of California, Santa Barbara, CA, United States
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- EKFZ—Else Kröner-Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| |
Collapse
|
30
|
Kwok WTH, Kwak HA, Andreazza AC. N-acetylcysteine modulates rotenone-induced mitochondrial Complex I dysfunction in THP-1 cells. Mitochondrion 2023; 72:1-10. [PMID: 37419232 DOI: 10.1016/j.mito.2023.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/12/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Mitochondrial Complex I dysfunction and oxidative stress have been part of the pathophysiology of several diseases ranging from mitochondrial disease to chronic diseases such as diabetes, mood disorders and Parkinson's Disease. Nonetheless, to investigate the potential of mitochondria-targeted therapeutic strategies for these conditions, there is a need further our understanding on how cells respond and adapt in the presence of Complex I dysfunction. In this study, we used low doses of rotenone, a classical inhibitor of mitochondrial complex I, to mimic peripheral mitochondrial dysfunction in THP-1 cells, a human monocytic cell line, and explored the effects of N-acetylcysteine on preventing this rotenone-induced mitochondrial dysfunction. Our results show that in THP-1 cells, rotenone exposure led to increases in mitochondrial superoxide, levels of cell-free mitochondrial DNA, and protein levels of the NDUFS7 subunit. N-acetylcysteine (NAC) pre-treatment ameliorated the rotenone-induced increase of cell-free mitochondrial DNA and NDUFS7 protein levels, but not mitochondrial superoxide. Furthermore, rotenone exposure did not affect protein levels of the NDUFV1 subunit but induced NDUFV1 glutathionylation. In summary, NAC may help to mitigate the effects of rotenone on Complex I and preserve the normal function of mitochondria in THP-1 cells.
Collapse
Affiliation(s)
- Winston Tse-Hou Kwok
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Haejin Angela Kwak
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Ana Cristina Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Mitochondrial Innovation Initiative, Toronto, ON, Canada.
| |
Collapse
|
31
|
Han S, Wang Q, Song Y, Pang M, Ren C, Wang J, Guan D, Xu W, Li F, Wang F, Zhou X, Fernández-Hernando C, Zhang H, Wu D, Ye Z. Lithium ameliorates Niemann-Pick C1 disease phenotypes by impeding STING/SREBP2 activation. iScience 2023; 26:106613. [PMID: 37128603 PMCID: PMC10148154 DOI: 10.1016/j.isci.2023.106613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/18/2022] [Accepted: 03/31/2023] [Indexed: 05/03/2023] Open
Abstract
Niemann-Pick disease type C (NP-C) is a genetic lysosomal disorder associated with progressive neurodegenerative phenotypes. Its therapeutic options are very limited. Here, we show that lithium treatment improves ataxia and feeding phenotypes, attenuates cerebellar inflammation and degeneration, and extends survival in Npc1 mouse models. In addition, lithium suppresses STING activation, SREBP2 processing to its mature form and the expression of the target genes in the Npc1 mice and in Npc1-deficient fibroblasts. Lithium impedes STING/SREBP2 transport from the ER to the Golgi, a step required for STING activation and SREBP2 processing, probably by lowering cytosolic calcium concentrations. This effect of lithium on STING/SREBP2 transport provides a mechanistic explanation for lithium's effects on Npc1 mice. Thus, this study reveals a potential therapeutic option for NP-C patients as well as a strategy to reduce active STING/SREBP2 pathway.
Collapse
Affiliation(s)
- Shiqian Han
- Department of Tropical Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qijun Wang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520, USA
- Departments of Pharmacology, Yale University School of Medicine, New Haven, CT06520, USA
- Shanghai Institute of Immunology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yongfeng Song
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520, USA
- Departments of Pharmacology, Yale University School of Medicine, New Haven, CT06520, USA
| | - Mao Pang
- Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing 400044, China
| | - Chunguang Ren
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520, USA
- Departments of Pharmacology, Yale University School of Medicine, New Haven, CT06520, USA
| | - Jing Wang
- Department of Tropical Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Dongwei Guan
- Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing 400044, China
| | - Wei Xu
- Biostatistics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Fangyong Li
- Biostatistics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Fengchao Wang
- Institute of Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xinyuan Zhou
- Department of Immunology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520, USA
- Comparative Medicine and Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Huiwen Zhang
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dianqing Wu
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520, USA
- Departments of Pharmacology, Yale University School of Medicine, New Haven, CT06520, USA
| | - Zhijia Ye
- Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing 400044, China
| |
Collapse
|
32
|
Wang R, Ma B, Shi K, Wu F, Zhou C. Effects of lithium on aggression in Drosophila. Neuropsychopharmacology 2023; 48:754-763. [PMID: 36253547 PMCID: PMC10066353 DOI: 10.1038/s41386-022-01475-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/15/2022] [Accepted: 10/05/2022] [Indexed: 11/08/2022]
Abstract
Lithium is a common medication used to treat mania and bipolar disorder, but the mechanisms by which lithium stabilizes mood and modifies aggression are still not fully understood. Here we found that acute but not chronic lithium significantly suppresses aggression without affecting locomotion in Drosophila melanogaster. Male flies treated with acute lithium are also less competitive than control males in establishing dominance. We also provided evidence that glycogen synthase kinase-3 (GSK-3), a well-known target of lithium, plays an important role in the anti-aggressive effect of lithium in Drosophila. Our genetic data showed that acute knockdown of GSK-3 in neurons can mimic the inhibitory effect of acute lithium on aggression, while specific overexpression of GSK-3 in a subset of P1 neurons profoundly promotes aggression which can be partially rescued by acute lithium application. Thus, these findings revealed the inhibitory effect of lithium on aggression in Drosophila and laid a groundwork for using Drosophila as a powerful model to investigate the mechanisms by which lithium reduces aggression.
Collapse
Affiliation(s)
- Rencong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Baoxu Ma
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Kai Shi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Fengming Wu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100101, Beijing, China.
| | - Chuan Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100101, Beijing, China.
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
33
|
Calabrese EJ, Pressman P, Hayes AW, Dhawan G, Kapoor R, Agathokleous E, Calabrese V. Lithium and hormesis: Enhancement of adaptive responses and biological performance via hormetic mechanisms. J Trace Elem Med Biol 2023; 78:127156. [PMID: 36958112 DOI: 10.1016/j.jtemb.2023.127156] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Biomedical and consumer interest in the health-promoting properties of pure single entities of known or unknown chemical constituents and mixtures has never been greater. Since its "rediscovery" in the 1950s, lithium is an example of such a constituent that represents an array of scientific and public health challenges and medical potentials that may now be understood best when seen through the lens of the dose-response paradigm known as hormesis. The present paper represents the first review of the capacity of lithium to induce hormetic dose responses in a broad range of biological models, organ systems, and endpoints. Of significance is that the numerous hormetic findings occur with extensive concentration/dose response evaluations with the optimal dosing being similar across multiple organ systems. The particular focus of these hormetic dose-response findings was targeted to research with a broad spectrum of stem cell types and neuroprotective effects. These findings suggest that lithium may have critically valuable systemic effects with respect to those therapeutically treated with lithium as well as for exposures that may be achieved via dietary intervention.
Collapse
Affiliation(s)
- Edward J Calabrese
- Environmental Health Sciences Division, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA.
| | - Peter Pressman
- Saba University School of Medicine, Caribbean, the Netherlands
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management College of Public Health, University of South Florida, Tampa, FL, USA
| | | | - Rachna Kapoor
- Saint Francis Hospital and Medical Center; Hartford, CT, USA
| | - Evgenios Agathokleous
- School of Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing 210044, China
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences; School of Medicine University of Catania, Via Santa Sofia 97, Catania 95123, Italy
| |
Collapse
|
34
|
Bian L, Nguyen VT, Tamaoki J, Endo Y, Dong G, Sato A, Kobayashi M. Genetic hyperactivation of Nrf2 causes larval lethality in Keap1a and Keap1b-double-knockout zebrafish. Redox Biol 2023; 62:102673. [PMID: 36934645 PMCID: PMC10031532 DOI: 10.1016/j.redox.2023.102673] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/15/2023] Open
Abstract
The Keap1-Nrf2 pathway is an evolutionarily conserved mechanism that protects cells from oxidative stress and electrophiles. Keap1 is a repressor of Nrf2 in normal cellular conditions but also a stress sensor for Nrf2 activation. Interestingly, fish and amphibians have two Keap1s (Keap1a and Keap1b), of which Keap1b is the ortholog of mammalian Keap1. Keap1a, on the other hand, is a gene found only in fish and amphibians, having been lost during the evolution to amniotes. We have previously shown that keap1b-knockout zebrafish have increased Nrf2 activity and reduced response to certain Nrf2-activating compounds but that they grow normally to adulthood. This may be because the remaining keap1a suppresses the hyperactivation of Nrf2, which is responsible for the post-natal lethality of Keap1-knockout mice. In this study, we analyzed keap1a;keap1b-double-knockout zebrafish to test this hypothesis. We found that keap1a;keap1b-double-knockout zebrafish, like Keap1-knockout mice, showed eating defects and were lethal within a week of hatching. Genetic introduction of the Nrf2 mutation rescued both the eating defects and the larval lethality, indicating that Nrf2 hyperactivation is the cause. However, unlike Keap1-knockout mice, keap1a;keap1b-double-knockout zebrafish showed no physical blockage of the food pathway; moreover, the cause of death was not directly related to eating defects. RNA-sequencing analysis revealed that keap1a;keap1b-double-knockout larvae showed extraordinarily high expression of known Nrf2-target genes as well as decreased expression of visual cycle genes. Finally, trigonelline or brusatol partially rescued the lethality of keap1a;keap1b-double-knockout larvae, suggesting that they can serve as an in vivo evaluation system for Nrf2-inhibiting compounds.
Collapse
Affiliation(s)
- Lixuan Bian
- Department of Molecular and Developmental Biology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Vu Thanh Nguyen
- Department of Molecular and Developmental Biology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan; Division of Aquaculture Biotechnology, Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Junya Tamaoki
- Department of Molecular and Developmental Biology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Yuka Endo
- Department of Molecular and Developmental Biology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Guilin Dong
- Department of Molecular and Developmental Biology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Ayaka Sato
- Department of Molecular and Developmental Biology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan
| | - Makoto Kobayashi
- Department of Molecular and Developmental Biology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan.
| |
Collapse
|
35
|
Compound combinations targeting longevity: Challenges and perspectives. Ageing Res Rev 2023; 85:101851. [PMID: 36642188 DOI: 10.1016/j.arr.2023.101851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/05/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Aging is one of the world's greatest concerns, requiring urgent, effective, large-scale interventions to decrease the number of late-life chronic diseases and improve human healthspan. Anti-aging drug therapy is one of the most promising strategies to combat the effects of aging. However, most geroprotective compounds are known to successfully affect only a few aging-related targets. Given this, there is a great biological rationale for the use of combinations of anti-aging interventions. In this review, we characterize the various types of compound combinations used to modulate lifespan, discuss the existing evidence on their role in life extension, and present some key points about current challenges and future prospects for the development of combination drug anti-aging therapy.
Collapse
|
36
|
Araldi E, Jutzeler CR, Ristow M. Lithium treatment extends human lifespan: findings from the UK Biobank. Aging (Albany NY) 2023; 15:421-440. [PMID: 36640269 PMCID: PMC9925675 DOI: 10.18632/aging.204476] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/27/2022] [Indexed: 01/13/2023]
Abstract
Lithium is a nutritional trace element that is also used pharmacologically for the management of bipolar and related psychiatric disorders. Recent studies have shown that lithium supplementation can extend health and lifespan in different animal models. Moreover, nutritional lithium uptake from drinking water was repeatedly found to be positively correlated with human longevity. By analyzing a large observational aging cohort (UK Biobank, n = 501,461 individuals) along with prescription data derived from the National Health Services (NHS), we here find therapeutic supplementation of lithium linked to decreased mortality (p = 0.0017) of individuals diagnosed with affective disorders. Subsequent multivariate survival analyses reveal lithium to be the strongest factor in regards to increased survival effects (hazard ratio = 0.274 [0.119-0.634 CI 95%, p = 0.0023]), corresponding to 3.641 times lower (95% CI 1.577-8.407) chances of dying at a given age for lithium users compared to users of other anti-psychotic drugs. While these results may further support the use of lithium as a geroprotective supplement, it should be noted that doses applied within the UK Biobank/NHS setting require close supervision by qualified medical professionals.
Collapse
Affiliation(s)
- Elisa Araldi
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zürich, Schwerzenbach CH-8603, Switzerland
- Center for Thrombosis and Hemostasis and Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz D-55131, Germany
| | - Catherine R. Jutzeler
- Biomedical Data Science Lab, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zürich, Zürich CH-8008, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zürich, Schwerzenbach CH-8603, Switzerland
- Institute of Experimental Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin D-10117, Germany
| |
Collapse
|
37
|
Castillo-Quan JI, Steinbaugh MJ, Fernández-Cárdenas LP, Pohl NK, Wu Z, Zhu F, Moroz N, Teixeira V, Bland MS, Lehrbach NJ, Moronetti L, Teufl M, Blackwell TK. An antisteatosis response regulated by oleic acid through lipid droplet-mediated ERAD enhancement. SCIENCE ADVANCES 2023; 9:eadc8917. [PMID: 36598980 PMCID: PMC9812393 DOI: 10.1126/sciadv.adc8917] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 11/23/2022] [Indexed: 05/19/2023]
Abstract
Although excessive lipid accumulation is a hallmark of obesity-related pathologies, some lipids are beneficial. Oleic acid (OA), the most abundant monounsaturated fatty acid (FA), promotes health and longevity. Here, we show that OA benefits Caenorhabditis elegans by activating the endoplasmic reticulum (ER)-resident transcription factor SKN-1A (Nrf1/NFE2L1) in a lipid homeostasis response. SKN-1A/Nrf1 is cleared from the ER by the ER-associated degradation (ERAD) machinery and stabilized when proteasome activity is low and canonically maintains proteasome homeostasis. Unexpectedly, OA increases nuclear SKN-1A levels independently of proteasome activity, through lipid droplet-dependent enhancement of ERAD. In turn, SKN-1A reduces steatosis by reshaping the lipid metabolism transcriptome and mediates longevity from OA provided through endogenous accumulation, reduced H3K4 trimethylation, or dietary supplementation. Our findings reveal an unexpected mechanism of FA signal transduction, as well as a lipid homeostasis pathway that provides strategies for opposing steatosis and aging, and may mediate some benefits of the OA-rich Mediterranean diet.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Michael J. Steinbaugh
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Laura Paulette Fernández-Cárdenas
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Nancy K. Pohl
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Ziyun Wu
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Feimei Zhu
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Natalie Moroz
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Biology Department, Emmanuel College, Boston, MA, USA
| | - Veronica Teixeira
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Monet S. Bland
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Nicolas J. Lehrbach
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Lorenza Moronetti
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Magdalena Teufl
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - T. Keith Blackwell
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Corresponding author.
| |
Collapse
|
38
|
Hamstra SI, Roy BD, Tiidus P, MacNeil AJ, Klentrou P, MacPherson RE, Fajardo VA. Beyond its Psychiatric Use: The Benefits of Low-dose Lithium Supplementation. Curr Neuropharmacol 2023; 21:891-910. [PMID: 35236261 PMCID: PMC10227915 DOI: 10.2174/1570159x20666220302151224] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/16/2022] [Accepted: 02/10/2022] [Indexed: 11/22/2022] Open
Abstract
Lithium is most well-known for its mood-stabilizing effects in the treatment of bipolar disorder. Due to its narrow therapeutic window (0.5-1.2 mM serum concentration), there is a stigma associated with lithium treatment and the adverse effects that can occur at therapeutic doses. However, several studies have indicated that doses of lithium under the predetermined therapeutic dose used in bipolar disorder treatment may have beneficial effects not only in the brain but across the body. Currently, literature shows that low-dose lithium (≤0.5 mM) may be beneficial for cardiovascular, musculoskeletal, metabolic, and cognitive function, as well as inflammatory and antioxidant processes of the aging body. There is also some evidence of low-dose lithium exerting a similar and sometimes synergistic effect on these systems. This review summarizes these findings with a focus on low-dose lithium's potential benefits on the aging process and age-related diseases of these systems, such as cardiovascular disease, osteoporosis, sarcopenia, obesity and type 2 diabetes, Alzheimer's disease, and the chronic low-grade inflammatory state known as inflammaging. Although lithium's actions have been widely studied in the brain, the study of the potential benefits of lithium, particularly at a low dose, is still relatively novel. Therefore, this review aims to provide possible mechanistic insights for future research in this field.
Collapse
Affiliation(s)
- Sophie I. Hamstra
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Brian D. Roy
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Peter Tiidus
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada
| | - Adam J. MacNeil
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Panagiota Klentrou
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Rebecca E.K. MacPherson
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
- Centre for Neurosciences, Brock University, St. Catharines, Ontario, Canada
| | - Val A. Fajardo
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
- Centre for Neurosciences, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
39
|
Hurvitz N, Elkhateeb N, Sigawi T, Rinsky-Halivni L, Ilan Y. Improving the effectiveness of anti-aging modalities by using the constrained disorder principle-based management algorithms. FRONTIERS IN AGING 2022; 3:1044038. [PMID: 36589143 PMCID: PMC9795077 DOI: 10.3389/fragi.2022.1044038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
Aging is a complex biological process with multifactorial nature underlined by genetic, environmental, and social factors. In the present paper, we review several mechanisms of aging and the pre-clinically and clinically studied anti-aging therapies. Variability characterizes biological processes from the genome to cellular organelles, biochemical processes, and whole organs' function. Aging is associated with alterations in the degrees of variability and complexity of systems. The constrained disorder principle defines living organisms based on their inherent disorder within arbitrary boundaries and defines aging as having a lower variability or moving outside the boundaries of variability. We focus on associations between variability and hallmarks of aging and discuss the roles of disorder and variability of systems in the pathogenesis of aging. The paper presents the concept of implementing the constrained disease principle-based second-generation artificial intelligence systems for improving anti-aging modalities. The platform uses constrained noise to enhance systems' efficiency and slow the aging process. Described is the potential use of second-generation artificial intelligence systems in patients with chronic disease and its implications for the aged population.
Collapse
Affiliation(s)
- Noa Hurvitz
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Narmine Elkhateeb
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Tal Sigawi
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Lilah Rinsky-Halivni
- Braun School of Public Health, Hebrew University of Jerusalem, Jerusalem, Israel,Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Yaron Ilan
- Faculty of Medicine, Hebrew University and Department of Medicine, Hadassah Medical Center, Jerusalem, Israel,*Correspondence: Yaron Ilan,
| |
Collapse
|
40
|
Mathis BJ, Kato H, Hiramatsu Y. Induction of Cardiac Pathology: Endogenous versus Exogenous Nrf2 Upregulation. Cells 2022; 11:cells11233855. [PMID: 36497112 PMCID: PMC9736027 DOI: 10.3390/cells11233855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of the endogenous antioxidant response to reactive oxygen species as well as a controller of Phase II detoxification in response to xenobiotics. This amenity to specific external manipulation exploits the binding affinity of Nrf2 for its constitutive repressor and degradation facilitator Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1). Derived from both natural and synthesized origins, these compounds have been extensively tested without definitive beneficial results. Unfortunately, multiple terminated trials have shown a negative side to Nrf2 with regard to cardiac pathologies while animal-based studies have demonstrated cardiomyocyte hypertrophy and heart failure after chronic Nrf2 upregulation. Putatively based on autophagic control of Nrf2 activity-modulating upstream factors, new evidence of miRNA involvement has added complexity to this mechanism. What follows is an extensive survey of Nrf2-regulating exogenous compounds that may promote cardiomyopathy, clinical trial evidence, and a comparison to exercise-induced factors that also upregulate Nrf2 while preventing cardiac pathologies.
Collapse
Affiliation(s)
- Bryan J. Mathis
- International Medical Center, University of Tsukuba Hospital, Tsukuba 305-8576, Ibaraki, Japan
- Correspondence: ; Tel.: +81-29-853-3004
| | - Hideyuki Kato
- Department of Cardiovascular Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Yuji Hiramatsu
- International Medical Center, University of Tsukuba Hospital, Tsukuba 305-8576, Ibaraki, Japan
| |
Collapse
|
41
|
Hottin C, Perron M, Roger JE. GSK3 Is a Central Player in Retinal Degenerative Diseases but a Challenging Therapeutic Target. Cells 2022; 11:cells11182898. [PMID: 36139472 PMCID: PMC9496697 DOI: 10.3390/cells11182898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. Although recent works strongly suggest that inhibiting GSK3 might also be a promising therapeutic approach for retinal degenerative diseases, its full potential is still under-evaluated. In this review, we summarize the literature on the role of GSK3 on the main cellular functions reported as deregulated during retinal degeneration, such as glucose homeostasis which is critical for photoreceptor survival, or oxidative stress, a major component of retinal degeneration. We also discuss the interest in targeting GSK3 for its beneficial effects on inflammation, for reducing neovascularization that occurs in some retinal dystrophies, or for cell-based therapy by enhancing Müller glia cell proliferation in diseased retina. Together, although GSK3 inhibitors hold promise as therapeutic agents, we highlight the complexity of targeting such a multitasked kinase and the need to increase our knowledge of the impact of reducing GSK3 activity on these multiple cellular pathways and biological processes.
Collapse
Affiliation(s)
- Catherine Hottin
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
42
|
Baek M, Jang W, Kim C. Dual Oxidase, a Hydrogen-Peroxide-Producing Enzyme, Regulates Neuronal Oxidative Damage and Animal Lifespan in Drosophila melanogaster. Cells 2022; 11:cells11132059. [PMID: 35805145 PMCID: PMC9265666 DOI: 10.3390/cells11132059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Reducing the oxidative stress in neurons extends lifespan in Drosophila melanogaster, highlighting the crucial role of neuronal oxidative damage in lifespan determination. However, the source of the reactive oxygen species (ROS) that provoke oxidative stress in neurons is not clearly defined. Here, we identify dual oxidase (duox), a calcium-activated ROS-producing enzyme, as a lifespan determinant. Due to the lethality of duox homozygous mutants, we employed a duox heterozygote that exhibited normal appearance and movement. We found that duox heterozygous male flies, which were isogenized with control flies, demonstrated extended lifespan. Neuronal knockdown experiments further suggested that duox is crucial to oxidative stress in neurons. Our findings suggest duox to be a source of neuronal oxidative stress associated with animal lifespan.
Collapse
|
43
|
An in vivo drug repurposing screen and transcriptional analyses reveals the serotonin pathway and GSK3 as major therapeutic targets for NGLY1 deficiency. PLoS Genet 2022; 18:e1010228. [PMID: 35653343 PMCID: PMC9162339 DOI: 10.1371/journal.pgen.1010228] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/02/2022] [Indexed: 11/19/2022] Open
Abstract
NGLY1 deficiency, a rare disease with no effective treatment, is caused by autosomal recessive, loss-of-function mutations in the N-glycanase 1 (NGLY1) gene and is characterized by global developmental delay, hypotonia, alacrima, and seizures. We used a Drosophila model of NGLY1 deficiency to conduct an in vivo, unbiased, small molecule, repurposing screen of FDA-approved drugs to identify therapeutic compounds. Seventeen molecules partially rescued lethality in a patient-specific NGLY1 deficiency model, including multiple serotonin and dopamine modulators. Exclusive dNGLY1 expression in serotonin and dopamine neurons, in an otherwise dNGLY1 deficient fly, was sufficient to partially rescue lethality. Further, genetic modifier and transcriptomic data supports the importance of serotonin signaling in NGLY1 deficiency. Connectivity Map analysis identified glycogen synthase kinase 3 (GSK3) inhibition as a potential therapeutic mechanism for NGLY1 deficiency, which we experimentally validated with TWS119, lithium, and GSK3 knockdown. Strikingly, GSK3 inhibitors and a serotonin modulator rescued size defects in dNGLY1 deficient larvae upon proteasome inhibition, suggesting that these compounds act through NRF1, a transcription factor that is regulated by NGLY1 and regulates proteasome expression. This study reveals the importance of the serotonin pathway in NGLY1 deficiency, and serotonin modulators or GSK3 inhibitors may be effective therapeutics for this rare disease. NGLY1 deficiency is a rare disease with no effective treatment. We conducted a drug repurposing screen and used the Connectivity Map, a transcriptional-based computational approach, to identify compounds that may serve as therapeutics for NGLY1 deficient individuals. The drug repurposing screen identified FDA-approved compounds acting through the serotonin and dopamine pathway that partially rescued lethality in an NGLY1 deficiency fly model. We also found that expressing dNGLY1 (the Drosophila ortholog of NGLY1) exclusively in serotonin neurons, in an otherwise dNGLY1 deficient fly, partially rescued lethality. These data indicate the importance of the serotonin and dopamine systems in NGLY1 deficiency. The Connectivity Map analyses found GSK3 inhibitors as potential therapeutic compounds, which were validated in vivo in the fly. Furthermore, knockdown of sgg (the Drosophila ortholog of GSK3) partially rescued lethality in dNGLY1 deficient flies, suggesting GSK3 as a therapeutic target for NGLY1 deficiency. Taken together, this work identifies therapeutic strategies for NGLY1 deficiency.
Collapse
|
44
|
Editorial for the Special Issue “Adaptation, Aging, and Cell Death in Yeast Stress Response: Models, Mechanisms and Applications”. Microorganisms 2022; 10:microorganisms10061126. [PMID: 35744644 PMCID: PMC9230367 DOI: 10.3390/microorganisms10061126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 01/27/2023] Open
|
45
|
Lithium chloride sensitivity connects the activity of PEX11 and RIM20 to the translation of PGM2 and other mRNAs with structured 5’-UTRs. Mol Cell Biochem 2022; 477:2643-2656. [DOI: 10.1007/s11010-022-04466-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
|
46
|
Liu YJ, McIntyre RL, Janssens GE. Considerations Regarding Public Use of Longevity Interventions. FRONTIERS IN AGING 2022; 3:903049. [PMID: 35821857 PMCID: PMC9261328 DOI: 10.3389/fragi.2022.903049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022]
Abstract
Public attention and interest for longevity interventions are growing. These can include dietary interventions such as intermittent fasting, physical interventions such as various exercise regimens, or through supplementation of nutraceuticals or administration of pharmaceutics. However, it is unlikely that most interventions identified in model organisms will translate to humans, or that every intervention will benefit each person equally. In the worst case, even detrimental health effects may occur. Therefore, identifying longevity interventions using human data and tracking the aging process in people is of paramount importance as we look towards longevity interventions for the public. In this work, we illustrate how to identify candidate longevity interventions using population data in humans, an approach we have recently employed. We consider metformin as a case-study for potential confounders that influence effectiveness of a longevity intervention, such as lifestyle, sex, genetics, age of administration and the microbiome. Indeed, metformin, like most other longevity interventions, may end up only benefitting a subgroup of individuals. Fortunately, technologies have emerged for tracking the rate of 'biological' aging in individuals, which greatly aids in assessing effectiveness. Recently, we have demonstrated that even wearable devices, accessible to everyone, can be used for this purpose. We therefore propose how to use such approaches to test interventions in the general population. In summary, we advocate that 1) not all interventions will be beneficial for each individual and therefore 2) it is imperative that individuals track their own aging rates to assess healthy aging interventions.
Collapse
Affiliation(s)
| | | | - Georges E. Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
47
|
Spuch C, López-García M, Rivera-Baltanás T, Cabrera-Alvargonzález JJ, Gadh S, Rodrigues-Amorim D, Álvarez-Estévez T, Mora A, Iglesias-Martínez-Almeida M, Freiría-Martínez L, Pérez-Rodríguez M, Pérez-González A, López-Domínguez A, Longueira-Suarez MR, Sousa-Domínguez A, Araújo-Ameijeiras A, Mosquera-Rodríguez D, Crespo M, Vila-Fernández D, Regueiro B, Olivares JM. Efficacy and Safety of Lithium Treatment in SARS-CoV-2 Infected Patients. Front Pharmacol 2022; 13:850583. [PMID: 35496309 PMCID: PMC9046673 DOI: 10.3389/fphar.2022.850583] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/14/2022] [Indexed: 12/15/2022] Open
Abstract
At the beginning of the pandemic, we observed that lithium carbonate had a positive effect on the recovery of severely ill patients with COVID-19. Lithium is able to inhibit the replication of several types of viruses, some of which are similar to the SARS-CoV-2 virus, increase the immune response and reduce inflammation by preventing or reducing the cytokine storm. Previously, we published an article with data from six patients with severe COVID-19 infection, where we proposed that lithium carbonate could be used as a potential treatment for COVID-19. Now, we set out to conduct a randomized clinical trial number EudraCT 2020–002008–37 to evaluate the efficacy and safety of lithium treatment in patients infected with severe SARS-CoV-2. We showed that lithium was able to reduce the number of days of hospital and intensive care unit admission as well as the risk of death, reduces inflammatory cytokine levels by preventing cytokine storms, and also reduced the long COVID syndromes. We propose that lithium carbonate can be used to reduce the severity of COVID-19.
Collapse
Affiliation(s)
- Carlos Spuch
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- *Correspondence: Carlos Spuch,
| | - Marta López-García
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- Department of Psychiatry, Hospital Álvaro Cunqueiro, SERGAS, Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
| | - J. J Cabrera-Alvargonzález
- Microbiology and Infectology Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Sudhir Gadh
- US Navy Medical Corps Commander, Medical Director at Educational Alliance, Medical Director at Rejuvenation Health, New York, NY, United States
| | - Daniela Rodrigues-Amorim
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Norfolk, United States
| | - Tania Álvarez-Estévez
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- Department of Psychiatry, Hospital Álvaro Cunqueiro, SERGAS, Vigo, Spain
| | - Almudena Mora
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
| | - Marta Iglesias-Martínez-Almeida
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- Universidade de Vigo, Vigo, Spain
| | - Luis Freiría-Martínez
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- Universidade de Vigo, Vigo, Spain
| | - Maite Pérez-Rodríguez
- Infectious Diseases Unit and Virology & Pathogenesis Group, Department of Internal Medicine, Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Alexandre Pérez-González
- Infectious Diseases Unit and Virology & Pathogenesis Group, Department of Internal Medicine, Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Ana López-Domínguez
- Infectious Diseases Unit and Virology & Pathogenesis Group, Department of Internal Medicine, Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - María Rebeca Longueira-Suarez
- Infectious Diseases Unit and Virology & Pathogenesis Group, Department of Internal Medicine, Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Adrián Sousa-Domínguez
- Infectious Diseases Unit and Virology & Pathogenesis Group, Department of Internal Medicine, Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Alejandro Araújo-Ameijeiras
- Infectious Diseases Unit and Virology & Pathogenesis Group, Department of Internal Medicine, Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - David Mosquera-Rodríguez
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- Intensive Care Unit, Critical Care and Emergency Department, Hospital Álvaro Cunqueiro, SERGAS, Vigo, Spain
| | - Manuel Crespo
- Microbiology and Infectology Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Dolores Vila-Fernández
- Intensive Care Unit, Critical Care and Emergency Department, Hospital Álvaro Cunqueiro, SERGAS, Vigo, Spain
| | - Benito Regueiro
- Microbiology and Infectology Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Microbiology Department, Hospital Álvaro Cunqueiro, SERGAS, Vigo, Spain
- Microbiology and Parasitology Department Medicine and Odontology, Universidade de Santiago, Santiago de Compostela, Spain
| | - Jose Manuel Olivares
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute (IIS-Galicia Sur), SERGAS-UVIGO, CIBERSAM, Vigo, Spain
- Department of Psychiatry, Hospital Álvaro Cunqueiro, SERGAS, Vigo, Spain
| |
Collapse
|
48
|
Moskalev A, Guvatova Z, Lopes IDA, Beckett CW, Kennedy BK, De Magalhaes JP, Makarov AA. Targeting aging mechanisms: pharmacological perspectives. Trends Endocrinol Metab 2022; 33:266-280. [PMID: 35183431 DOI: 10.1016/j.tem.2022.01.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/12/2022]
Abstract
Geroprotectors slow down aging and promote healthy longevity in model animals. Although hundreds of compounds have been shown to extend the life of laboratory model organisms, clinical studies on potential geroprotectors are exceedingly rare, especially in healthy elders. This review aims to classify potential geroprotectors based on the mechanisms by which they influence aging. These pharmacological interventions can be classified into the following groups: those that prevent oxidation; proteostasis regulators; suppressors of genomic instability; epigenetic drugs; those that preserve mitochondrial function; inhibitors of aging-associated signaling pathways; hormetins; senolytics/senostatics; anti-inflammatory drugs; antifibrotic agents; neurotrophic factors; factors preventing the impairment of barrier function; immunomodulators; and prebiotics, metabiotics, and enterosorbents.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; Institute of Biology of the Federal Research Center of Komi Science Center, Ural Branch of the Russian Academy of Sciences, 28 Kommunisticheskaya Street, Syktyvkar 167982, Russia.
| | - Zulfiya Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Ines De Almeida Lopes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Charles W Beckett
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Brian K Kennedy
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore; Singapore Institute of Clinical Sciences, A*STAR, Singapore
| | - Joao Pedro De Magalhaes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK.
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.
| |
Collapse
|
49
|
Reith P, Braam S, Welkenhuysen N, Lecinski S, Shepherd J, MacDonald C, Leake MC, Hohmann S, Shashkova S, Cvijovic M. The Effect of Lithium on the Budding Yeast Saccharomyces cerevisiae upon Stress Adaptation. Microorganisms 2022; 10:590. [PMID: 35336166 PMCID: PMC8953283 DOI: 10.3390/microorganisms10030590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Lithium salts are used in the treatment of mood disorders, cancer, and Alzheimer's disease. It has been shown to prolong life span in several phyla; however, not yet in budding yeast. In our study, we investigate the influence of lithium on yeast cells' viability by characterizing protein aggregate formation, cell volume, and molecular crowding in the context of stress adaptation. While our data suggest a concentration-dependent growth inhibition caused by LiCl, we show an extended long-term survival rate as an effect of lithium addition upon glucose deprivation. We show that caloric restriction mitigates the negative impact of LiCl on cellular survival. Therefore, we suggest that lithium could affect glucose metabolism upon caloric restriction, which could explain the extended long-term survival observed in our study. We find furthermore that lithium chloride did not affect an immediate salt-induced Hsp104-dependent aggregate formation but cellular adaptation to H2O2 and acute glucose starvation. We presume that different salt types and concentrations interfere with effective Hsp104 recruitment or its ATP-dependent disaggregase activity as a response to salt stress. This work provides novel details of Li+ effect on live eukaryotic cells which may also be applicable in further research on the treatment of cancer, Alzheimer's, or other age-related diseases in humans.
Collapse
Affiliation(s)
- Patrick Reith
- Department of Mathematical Sciences, University of Gothenburg, 412 96 Gothenburg, Sweden; (P.R.); (S.B.); (N.W.)
- Department of Mathematical Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden;
| | - Svenja Braam
- Department of Mathematical Sciences, University of Gothenburg, 412 96 Gothenburg, Sweden; (P.R.); (S.B.); (N.W.)
- Department of Mathematical Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Niek Welkenhuysen
- Department of Mathematical Sciences, University of Gothenburg, 412 96 Gothenburg, Sweden; (P.R.); (S.B.); (N.W.)
- Department of Mathematical Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Sarah Lecinski
- Department of Physics, University of York, York YO10 5DD, UK; (S.L.); (J.S.); (M.C.L.)
| | - Jack Shepherd
- Department of Physics, University of York, York YO10 5DD, UK; (S.L.); (J.S.); (M.C.L.)
- Department of Biology, University of York, York YO10 5DD, UK;
| | - Chris MacDonald
- Department of Biology, University of York, York YO10 5DD, UK;
| | - Mark C. Leake
- Department of Physics, University of York, York YO10 5DD, UK; (S.L.); (J.S.); (M.C.L.)
- Department of Biology, University of York, York YO10 5DD, UK;
| | - Stefan Hohmann
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden;
| | - Sviatlana Shashkova
- Department of Mathematical Sciences, University of Gothenburg, 412 96 Gothenburg, Sweden; (P.R.); (S.B.); (N.W.)
- Department of Mathematical Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Marija Cvijovic
- Department of Mathematical Sciences, University of Gothenburg, 412 96 Gothenburg, Sweden; (P.R.); (S.B.); (N.W.)
- Department of Mathematical Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| |
Collapse
|
50
|
Low-Dose Delta-9-Tetrahydrocannabinol as Beneficial Treatment for Aged APP/PS1 Mice. Int J Mol Sci 2022; 23:ijms23052757. [PMID: 35269905 PMCID: PMC8910894 DOI: 10.3390/ijms23052757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/15/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Studies on the effective and safe therapeutic dosage of delta-9-tetrahydrocannabinol (THC) for the treatment of Alzheimer’s disease (AD) have been sparse due to the concern about THC’s psychotropic activity. The present study focused on demonstrating the beneficial effect of low-dose THC treatment in preclinical AD models. The effect of THC on amyloid-β (Aβ) production was examined in N2a/AβPPswe cells. An in vivo study was conducted in aged APP/PS1 transgenic mice that received an intraperitoneal injection of THC at 0.02 and 0.2 mg/kg every other day for three months. The in vitro study showed that THC inhibited Aβ aggregation within a safe dose range. Results of the radial arm water maze (RAWM) test demonstrated that treatment with 0.02 and 0.2 mg/kg of THC for three months significantly improved the spatial learning performance of aged APP/PS1 mice in a dose-dependent manner. Results of protein analyses revealed that low-dose THC treatment significantly decreased the expression of Aβ oligomers, phospho-tau and total tau, and increased the expression of Aβ monomers and phospho-GSK-3β (Ser9) in the THC-treated brain tissues. In conclusion, treatment with THC at 0.2 and 0.02 mg/kg improved the spatial learning of aged APP/PS1 mice, suggesting low-dose THC is a safe and effective treatment for AD.
Collapse
|