1
|
Fraiha-Pegado J, de Paula VJR, Alotaibi T, Forlenza O, Hajek T. Trace lithium levels in drinking water and risk of dementia: a systematic review. Int J Bipolar Disord 2024; 12:32. [PMID: 39212809 PMCID: PMC11364728 DOI: 10.1186/s40345-024-00348-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Since its debut in 1949, lithium (Li) has been regarded as a gold standard therapy for mood stabilization. Neuroprotective effects of Li have been replicated across many different paradigms ranging from tissue cultures to human studies. This has generated interest in potentially repurposing this drug. However, the optimal dosage required for neuroprotective effects remains unclear and may be different than the doses needed for treatment of bipolar disorders. Recent studies on trace-Li levels in the water suggest that Li, could slow cognitive decline and prevent dementia with long-term use even at very low doses. The current review aims to synthesize the data on the topic and challenge the conventional high-dose paradigm. RESULTS We systematically reviewed five available studies, which reported associations between trace-Li in water and incidence or mortality from dementia. Association between trace-Li levels and a lower risk or mortality from dementia were observed at concentrations of Li in drinking water as low as 0.002 mg/L and 0.056 mg/L. Meanwhile, levels below 0.002 mg/L did not elicit this effect. Although three of the five studies found dementia protective properties of Li in both sexes, a single study including lower Li levels (0.002 mg/l) found such association only in women. CONCLUSION: The reviewed evidence shows that trace-Li levels in the water are sufficient to lower the incidence or mortality from dementia. Considering the lack of options for the prevention or treatment of dementia, we should not ignore these findings. Future trials of Li should focus on long term use of low or even micro doses of Li in the prevention or treatment of dementia.
Collapse
Affiliation(s)
- Julia Fraiha-Pegado
- Department of Psychiatry, Dalhousie University, 909 Veteran's Memorial Lane, Halifax, Canada
| | - Vanessa J Rodrigues de Paula
- Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, HCFMUSP-Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Tariq Alotaibi
- Department of Psychiatry, Dalhousie University, 909 Veteran's Memorial Lane, Halifax, Canada
- Department of Medical Specialities, College of Medicine, Majmmah University, Majmmah City, Saudi Arabia
| | - Orestes Forlenza
- Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, HCFMUSP-Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Tomas Hajek
- Department of Psychiatry, Dalhousie University, 909 Veteran's Memorial Lane, Halifax, Canada.
| |
Collapse
|
2
|
Shen Y, Zhao M, Zhao P, Meng L, Zhang Y, Zhang G, Taishi Y, Sun L. Molecular mechanisms and therapeutic potential of lithium in Alzheimer's disease: repurposing an old class of drugs. Front Pharmacol 2024; 15:1408462. [PMID: 39055498 PMCID: PMC11269163 DOI: 10.3389/fphar.2024.1408462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline and memory loss. Despite advances in understanding the pathophysiological mechanisms of AD, effective treatments remain scarce. Lithium salts, recognized as mood stabilizers in bipolar disorder, have been extensively studied for their neuroprotective effects. Several studies indicate that lithium may be a disease-modifying agent in the treatment of AD. Lithium's neuroprotective properties in AD by acting on multiple neuropathological targets, such as reducing amyloid deposition and tau phosphorylation, enhancing autophagy, neurogenesis, and synaptic plasticity, regulating cholinergic and glucose metabolism, inhibiting neuroinflammation, oxidative stress, and apoptosis, while preserving mitochondrial function. Clinical trials have demonstrated that lithium therapy can improve cognitive function in patients with AD. In particular, meta-analyses have shown that lithium may be a more effective and safer treatment than the recently FDA-approved aducanumab for improving cognitive function in patients with AD. The affordability and therapeutic efficacy of lithium have prompted a reassessment of its use. However, the use of lithium may lead to potential side effects and safety issues, which may limit its clinical application. Currently, several new lithium formulations are undergoing clinical trials to improve safety and efficacy. This review focuses on lithium's mechanism of action in treating AD, highlighting the latest advances in preclinical studies and clinical trials. It also explores the side effects of lithium therapy and coping strategies, offering a potential therapeutic strategy for patients with AD.
Collapse
Affiliation(s)
- Yanxin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Meng Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Panpan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Lingjie Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Yan Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Yezi Taishi
- Department of Cadre Ward, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
3
|
Shoghi E, Safari T, Parsi-Moud A, Mirzaei I, Rad NS, Chahkandi M. Effects of moderate intensity training and lithium on spatial learning and memory in a rat model: The role of SIRT3 and PGC1-α expression levels and brain-derived neurotropic factor. Exp Gerontol 2024; 191:112442. [PMID: 38663491 DOI: 10.1016/j.exger.2024.112442] [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: 02/27/2024] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
In this study we investigated the potential synergistic effects of moderate interval training (MIT) and lithium on spatial learning and memory. Forty-two male Wistar males were classified into six groups including I: Control, II: 10 mg/kg/day IP lithium (Li10), III: MIT, IV: Li10 + MIT, V: 40 mg/kg/day IP lithium (Li40), and VI: Li40 + MIT. Then, the rats underwent Morris Water Maze (MWM) test to assess their spatial memory and learning ability. Brain-derived neurotrophic factor (BDNF) density was measured by enzyme-linked immunosorbent assay (ELISA), and the expression of PGC1 and SIRT3 were assessed via qRT-PCR. The results show that MIT improves both memory and spatial learning; but lithium alone, does not cause this. Additionally, those exposed to a combination of exercise and lithium also had improved spatial learning and memory. Finally, we observed a positive role of BDNF protein, and PGC1 gene on the effects of exercise and lithium.
Collapse
Affiliation(s)
- Elham Shoghi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Tahereh Safari
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran; Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Abolfazl Parsi-Moud
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran; Student Research Committee, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ilia Mirzaei
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran; Student Research Committee, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Nahid Sepehri Rad
- Infectious Diseases and Tropical Medicine Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohadeseh Chahkandi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
4
|
Singulani MP, Ferreira AFF, Figueroa PS, Cuyul-Vásquez I, Talib LL, Britto LR, Forlenza OV. Lithium and disease modification: A systematic review and meta-analysis in Alzheimer's and Parkinson's disease. Ageing Res Rev 2024; 95:102231. [PMID: 38364914 DOI: 10.1016/j.arr.2024.102231] [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: 07/19/2023] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
The role of lithium as a possible therapeutic strategy for neurodegenerative diseases has generated scientific interest. We systematically reviewed and meta-analyzed pre-clinical and clinical studies that evidenced the neuroprotective effects of lithium in Alzheimer's (AD) and Parkinson's disease (PD). We followed the PRISMA guidelines and performed the systematic literature search using PubMed, EMBASE, Web of Science, and Cochrane Library. A total of 32 articles were identified. Twenty-nine studies were performed in animal models and 3 studies were performed on human samples of AD. A total of 17 preclinical studies were included in the meta-analysis. Our analysis showed that lithium treatment has neuroprotective effects in diseases. Lithium treatment reduced amyloid-β and tau levels and significantly improved cognitive behavior in animal models of AD. Lithium increased the tyrosine hydroxylase levels and improved motor behavior in the PD model. Despite fewer clinical studies on these aspects, we evidenced the positive effects of lithium in AD patients. This study lends further support to the idea of lithium's therapeutic potential in neurodegenerative diseases.
Collapse
Affiliation(s)
- Monique Patricio Singulani
- Laboratory of Neuroscience LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil; Centro de Neurociências Translacionais (CNT), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| | - Ana Flávia Fernandes Ferreira
- Laboratory of Cellular Neurobiology, Department of Physiology and Biophysics, Instituto de Ciências Biomédicas da Universidade de São Paulo (USP), São Paulo, Brazil
| | | | - Iván Cuyul-Vásquez
- Departamento de Procesos Terapéuticos, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco, Chile; Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Leda Leme Talib
- Laboratory of Neuroscience LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil; Centro de Neurociências Translacionais (CNT), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| | - Luiz Roberto Britto
- Laboratory of Cellular Neurobiology, Department of Physiology and Biophysics, Instituto de Ciências Biomédicas da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Orestes Vicente Forlenza
- Laboratory of Neuroscience LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, Brazil; Centro de Neurociências Translacionais (CNT), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil; Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBioN), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil.
| |
Collapse
|
5
|
Themoteo RM, De Paula VJR, Rocha NKR, Brentani H, Forlenza OV. Lithium Prevents Telomere Shortening in Cortical Neurons in Amyloid-Beta Induced Toxicity. NEUROSCI 2023; 4:1-8. [PMID: 39484296 PMCID: PMC11523687 DOI: 10.3390/neurosci4010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND There is consistent evidence of the potential benefits of lithium attenuating mechanisms of neurodegeneration, including those related to the pathophysiology of Alzheimer's disease (AD), and facilitating neurotrophic and protective responses, including maintenance of telomere length. The aim was to investigate the protective effect of the pre-treatment with lithium on amyloid-beta (Aβ)-induced toxicity and telomere length in neurons. METHODS Cortical neurons were treated with lithium chloride at therapeutic and subtherapeutic concentrations (2 mM, 0.2 mM and 0.02 mM) for seven days. Amyloid toxicity was induced 24 h before the end of lithium treatment. RESULTS Lithium resulted in 120% (2 mM), 180% (0.2 mM) and 140% (0.02 mM) increments in telomere length as compared to untreated controls. Incubation with Aβ1-42 was associated with significant reductions in MTT uptake (33%) and telomere length (83%) as compared to controls. CONCLUSIONS Lithium prevented loss of culture viability and telomere shortening in neuronal cultures challenged with Aβ fibrils.
Collapse
Affiliation(s)
- Rafael M. Themoteo
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Vanessa J. R. De Paula
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
- Laboratory of Psychobiology (LIM-23), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Nicole K. R. Rocha
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
- Laboratory of Psychobiology (LIM-23), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Helena Brentani
- Laboratory of Psychobiology (LIM-23), Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05508-090, SP, Brazil
| |
Collapse
|
6
|
Ding H, Zhong Y, Liu N, Wu H, Xu H, Wu Y, Liu G, Yuan S, Zhou Q, Wang C. Panic disorder aging characteristics: The role of telomerase reverse transcriptase gene and brain function. Front Aging Neurosci 2022; 14:835963. [PMID: 35992589 PMCID: PMC9389410 DOI: 10.3389/fnagi.2022.835963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/13/2022] [Indexed: 11/30/2022] Open
Abstract
Panic disorder (PD) causes serious functional damage and disability and accelerates the process of individual aging. The pathological basis of PD is the same as that of age-related diseases, which is proposed as a new viewpoint in recent years. Memory decline and social functional impairment are common manifestations of accelerated aging in PD. The function of telomerase reverse transcriptase (TERT) and telomere length (TL) is abnormal in patients with aging and PD. However, the molecular mechanism behind remains unclear. The purpose of this study was to explore the relationship between TERT gene expression (including DNA methylation) and the changes in PD aging characteristics (memory and social function). By TERT gene knockout mice, we found that loss of TERT attenuated the acquisition of recent fear memory during contextual fear conditioning. This study reported that a significantly lower methylation level of human TERT (hTERT) gene was detected in PD patients compared with healthy control and particularly decreased CpG methylation in the promoter region of hTERT was associated with the clinical characteristics in PD. Regional homogeneity (ReHo) analysis showed that the methylation of hTERT (cg1295648) influenced social function of PD patients through moderating the function of the left postcentral gyrus (PCG). This indicates that the hTERT gene may play an important role in the pathological basis of PD aging and may become a biological marker for evaluating PD aging. These findings provide multidimensional evidence for the underlying genetic and pathological mechanisms of PD.
Collapse
Affiliation(s)
- Huachen Ding
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Yuan Zhong
- School of Psychology, Nanjing Normal University, Nanjing, China
| | - Na Liu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, China
| | - Huiqin Wu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Huazhen Xu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Yun Wu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
- School of Psychology, Nanjing Normal University, Nanjing, China
- Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, China
| | - Gang Liu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
- Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, China
| | - Shiting Yuan
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Qigang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Qigang Zhou,
| | - Chun Wang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
- School of Psychology, Nanjing Normal University, Nanjing, China
- Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, China
- *Correspondence: Chun Wang,
| |
Collapse
|
7
|
De-Paula VJ, Forlenza OV. Lithium modulates multiple tau kinases with distinct effects in cortical and hippocampal neurons according to concentration ranges. Naunyn Schmiedebergs Arch Pharmacol 2021; 395:105-113. [PMID: 34751792 DOI: 10.1007/s00210-021-02171-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/19/2021] [Indexed: 11/26/2022]
Abstract
The hyperphosphorylation of tau is a central mechanism in the pathogenesis of Alzheimer's disease (AD). Lithium is a potent inhibitor of glycogen synthase kinase-3beta (GSK3β), the most important tau kinase in neurons, and may also affect tau phosphorylation by modifying the expression and/or activity of other kinases, such as protein kinase A (PKA), Akt (PKB), and calcium calmodulin kinase-II (CaMKII). The aim of the present study is to determine the effect of chronic lithium treatment on the protein expression of tau and its major kinases in cortical and hippocampal neurons, at distinct working concentrations. Primary cultures of cortical and hippocampal neurons were treated with sub-therapeutic (0.02 mM and 0.2 mM) and therapeutic (2 mM) concentrations of lithium for 7 days. Protein expression of tau and tau-kinases was determined by immunoblotting. An indirect estimate of GSK3β activity was determined by the GSK3β ratio (rGSKβ). Statistically significant increments in the protein expression of tau and CaMKII were observed both in cortical and hippocampal neurons treated with subtherapeutic doses of lithium. GSK3β activity was increased in cortical, but decreased in hippocampal neurons. Distinct patterns of changes in the expression of the remaining tau tau-kinases were observed: in cortical neurons, lithium treatment was associated with consistent decrements in Akt and PKA, whereas hippocampal neurons displayed increased protein expression of Akt and decreased PKA. Our results suggest that chronic lithium treatment may yield distinct biological effects depending on the concentration range, with regional specificity. We further suggest that hippocampal neurons may be more sensitive to the effect of lithium, presenting with changes in the expression of tau-related proteins at subtherapeutic doses, which may not be mirrored by the effects observed in cortical neurons.
Collapse
Affiliation(s)
- V J De-Paula
- Laboratório de Neurociências (LIM-27), Departamento E Instituto de Psiquiatria, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
- Laboratório de Psicobiologia (LIM-23), Instituto de Psiquiatria, Hospital das Clinicas da Faculdade de Medicina da USP, Rua Dr. Ovídio Pires de Campos 785, São Paulo, SP, 05403-903, Brazil.
| | - O V Forlenza
- Laboratório de Neurociências (LIM-27), Departamento E Instituto de Psiquiatria, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
8
|
Singulani MP, De Paula VJR, Forlenza OV. Mitochondrial dysfunction in Alzheimer's disease: Therapeutic implications of lithium. Neurosci Lett 2021; 760:136078. [PMID: 34161823 DOI: 10.1016/j.neulet.2021.136078] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/12/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases, characterized by the accumulation of abnormal tau proteins within neurons and amyloid plaques in the brain parenchyma, which leads to progressive loss of neurons in the brain. While the detailed mechanism of the pathogenesis of AD is still unknown, evidence suggests that mitochondrial dysfunction likely plays a fundamental role in the pathogenesis of this disease. Due to the relevance of mitochondrial alterations in AD, recent works have suggested the therapeutic potential of mitochondrial-targeted lithium. Lithium has been shown to possess neuroprotective and neurotrophic properties that could also be related to the upregulation of mitochondrial function. In the current work, we perform a comprehensive investigation of the significance of mitochondrial dysfunction in AD and pharmacological treatment with lithium as imperative in this pathology, through a brief review of the major findings on the effects of lithium as a therapeutic approach targeting mitochondria in the context of AD.
Collapse
Affiliation(s)
- Monique P Singulani
- Laboratory of Neurosciences - LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vanessa J R De Paula
- Laboratory of Neurosciences - LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Orestes V Forlenza
- Laboratory of Neurosciences - LIM27, Departamento e Instituto de Psiquiatria, Hospital das Clínicas HCFMUSP, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
9
|
Mini-review: The anti-aging effects of lithium in bipolar disorder. Neurosci Lett 2021; 759:136051. [PMID: 34139318 DOI: 10.1016/j.neulet.2021.136051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 11/22/2022]
Abstract
The medical use of lithium has grown since its initial introduction in the 1800s as a treatment for gout. Today, the divalent cation remains as the pharmacological gold standard in treatment of bipolar disorder (BD) with strong mood stabilizing effects. Lithium has demonstrated efficacy in the treatment of acute affective episodes, in the reduction of affective episode recurrence, and in significantly decreasing the risk of suicide in patients. BD has been consistently associated with clinical signs of accelerated aging, including increased rates of age-related diseases such as cardiovascular diseases, malignancies, and diabetes mellitus. This clinical scenario parallels accelerated aging mechanisms observed on a molecular basis, with studies reporting shortened telomeres, increased oxidative stress, and accelerated epigenetic aging in patients with BD compared to controls. Lithium has proved useful as a potential agent in slowing down this accelerated aging process in BD, potentially reversing effects induced by the disorder. This mini-review summarizes findings of anti-aging mechanisms associated with lithium use and provides a discussion of the clinical implications and perspectives of this evolving field. Despite many promising results, more studies are warranted in order to elucidate the exact mechanism by which lithium may act as an anti-aging agent and the extent to which these mechanisms are relevant to its mood stabilizing properties in BD.
Collapse
|
10
|
Gavia-García G, Rosado-Pérez J, Arista-Ugalde TL, Aguiñiga-Sánchez I, Santiago-Osorio E, Mendoza-Núñez VM. Telomere Length and Oxidative Stress and Its Relation with Metabolic Syndrome Components in the Aging. BIOLOGY 2021; 10:253. [PMID: 33804844 PMCID: PMC8063797 DOI: 10.3390/biology10040253] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/20/2022]
Abstract
A great amount of scientific evidence supports that Oxidative Stress (OxS) can contribute to telomeric attrition and also plays an important role in the development of certain age-related diseases, among them the metabolic syndrome (MetS), which is characterised by clinical and biochemical alterations such as obesity, dyslipidaemia, arterial hypertension, hyperglycaemia, and insulin resistance, all of which are considered as risk factors for type 2 diabetes mellitus (T2DM) and cardiovascular diseases, which are associated in turn with an increase of OxS. In this sense, we review scientific evidence that supports the association between OxS with telomere length (TL) dynamics and the relationship with MetS components in aging. It was analysed whether each MetS component affects the telomere length separately or if they all affect it together. Likewise, this review provides a summary of the structure and function of telomeres and telomerase, the mechanisms of telomeric DNA repair, how telomere length may influence the fate of cells or be linked to inflammation and the development of age-related diseases, and finally, how the lifestyles can affect telomere length.
Collapse
Affiliation(s)
- Graciela Gavia-García
- Research Unit on Gerontology, FES Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.G.-G.); (J.R.-P.); (T.L.A.-U.)
| | - Juana Rosado-Pérez
- Research Unit on Gerontology, FES Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.G.-G.); (J.R.-P.); (T.L.A.-U.)
| | - Taide Laurita Arista-Ugalde
- Research Unit on Gerontology, FES Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.G.-G.); (J.R.-P.); (T.L.A.-U.)
| | - Itzen Aguiñiga-Sánchez
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (I.A.-S.); (E.S.-O.)
| | - Edelmiro Santiago-Osorio
- Hematopoiesis and Leukemia Laboratory, Research Unit on Cell Differentiation and Cancer, FES Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (I.A.-S.); (E.S.-O.)
| | - Víctor Manuel Mendoza-Núñez
- Research Unit on Gerontology, FES Zaragoza, National Autonomous University of Mexico, Mexico City 09230, Mexico; (G.G.-G.); (J.R.-P.); (T.L.A.-U.)
| |
Collapse
|
11
|
De-Paula VJ, Dos Santos CCC, Luque MCA, Ali TM, Kalil JE, Forlenza OV, Cunha-Neto E. Acute and chronic lithium treatment increases Wnt/β-catenin transcripts in cortical and hippocampal tissue at therapeutic concentrations in mice. Metab Brain Dis 2021; 36:193-197. [PMID: 33170418 DOI: 10.1007/s11011-020-00638-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 10/27/2020] [Indexed: 12/27/2022]
Abstract
Lithium activates Wnt/β-catenin signaling leading to stabilization of free cytosolic β-catenin. The aim of the present study is to evaluate the in vivo effect of acute and chronic lithium treatment on the expression of β-catenin target genes, addressing its transcripts HIG2, Bcl-xL, Cyclin D1, c-myc, in cortical and hippocampal tissue from adult mice. Lithium doses were established to yield therapeutic working concentrations. In acute treatment, mice received a 300µL of a 350 mg/kg solution of LiCl by gavage, and were euthanized after 2 h, 6 h and 12 h. To determine the effect of chronic treatment, animals were continuously fed either with chow supplemented with 2 g/kg Li2CO3, or regular chow (controls), being euthanized after 30 days. All animals had access to drinking water and 0.9% saline ad libitum. After acute and chronic treatments samples of peripheral blood were obtained from the tail vein for each animal, and serum concentrations of lithium were determined. All transcripts were up-regulated in cortical and hippocampal tissues of lithium-treated mice, both under acute and chronic treatments. There was a positive correlation between serum lithium concentrations and the increment in the expression of all transcripts. This effect was observed in all time points of the acute treatment (i.e., 2, 6 and 12 hours) and also after 30 days. We conclude that Wnt/β-catenin transcriptional response (HIG2, Bcl-xL, Cyclin D1 and c-myc) is up-regulated in the mouse brain in response to acute and chronic lithium treatment at therapeutic concentrations.
Collapse
Affiliation(s)
- Vanessa J De-Paula
- Laboratório de Psicobiologia (LIM23), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil.
- Laboratorio de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil.
- Laboratory of Neuroscience, Department and Institute of Psychiatry, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil.
| | - Carla Cristine C Dos Santos
- Laboratório de Imunologia Clínica e Alergia (LIM60), Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
| | - Maria Carolina A Luque
- Laboratório de Imunologia Clínica e Alergia (LIM60), Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
- Laboratorio de Imunologia, Instituto do Coração (InCor), Faculty of Medicine, Universidade de Sao Paulo, SP, Sao Paulo, Brasil
| | - Taccyana M Ali
- Laboratório de Imunologia Clínica e Alergia (LIM60), Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
- Laboratorio de Imunologia, Instituto do Coração (InCor), Faculty of Medicine, Universidade de Sao Paulo, SP, Sao Paulo, Brasil
| | - Jorge E Kalil
- Laboratorio de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
- Laboratorio de Imunologia, Instituto do Coração (InCor), Faculty of Medicine, Universidade de Sao Paulo, SP, Sao Paulo, Brasil
- Instituto de Investigação em Imunologia (iii)-INCT, São Paulo, Brazil
| | | | - Edecio Cunha-Neto
- Laboratório de Imunologia Clínica e Alergia (LIM60), Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brasil
- Laboratorio de Imunologia, Instituto do Coração (InCor), Faculty of Medicine, Universidade de Sao Paulo, SP, Sao Paulo, Brasil
- Instituto de Investigação em Imunologia (iii)-INCT, São Paulo, Brazil
| |
Collapse
|
12
|
Rocha NKR, Themoteo R, Brentani H, Forlenza OV, De Paula VDJR. Neuronal-Glial Interaction in a Triple-Transgenic Mouse Model of Alzheimer's Disease: Gene Ontology and Lithium Pathways. Front Neurosci 2020; 14:579984. [PMID: 33335468 PMCID: PMC7737403 DOI: 10.3389/fnins.2020.579984] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Neuronal-glial interactions are critical for brain homeostasis, and disruption of this process may lead to excessive glial activation and inadequate pro-inflammatory responses. Abnormalities in neuronal-glial interactions have been reported in the pathophysiology of Alzheimer’s disease (AD), where lithium has been shown to exert neuroprotective effects, including the up-regulation of cytoprotective proteins. In the present study, we characterize by Gene Ontology (GO) the signaling pathways related to neuronal-glial interactions in response to lithium in a triple-transgenic mouse model of AD (3×-TgAD). Mice were treated for 8 months with lithium carbonate (Li) supplemented to chow, using two dose ranges to yield subtherapeutic working concentrations (Li1, 1.0 g/kg; and Li2, 2.0 g/kg of chow), or with standard chow (Li0). The hippocampi were removed and analyzed by proteomics. A neuronal-glial interaction network was created by a systematic literature search, and the selected genes were submitted to STRING, a functional network to analyze protein interactions. Proteomics data and neuronal-glial interactomes were compared by GO using ClueGo (Cytoscape plugin) with p ≤ 0.05. The proportional effects of neuron-glia interactions were determined on three GO domains: (i) biological process; (ii) cellular component; and (iii) molecular function. The gene ontology of this enriched network of genes was further stratified according to lithium treatments, with statistically significant effects observed in the Li2 group (as compared to controls) for the GO domains biological process and cellular component. In the former, there was an even distribution of the interactions occurring at the following functions: “positive regulation of protein localization to membrane,” “regulation of protein localization to cell periphery,” “oligodendrocyte differentiation,” and “regulation of protein localization to plasma membrane.” In cellular component, interactions were also balanced for “myelin sheath” and “rough endoplasmic reticulum.” We conclude that neuronal-glial interactions are implicated in the neuroprotective response mediated by lithium in the hippocampus of AD-transgenic mice. The effect of lithium on homeostatic pathways mediated by the interaction between neurons and glial cells are implicated in membrane permeability, protein synthesis and DNA repair, which may be relevant for the survival of nerve cells amidst AD pathology.
Collapse
Affiliation(s)
- Nicole Kemberly R Rocha
- Laboratório de Psicobiologia (LIM23), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Themoteo
- Laboratorio de Neurociencias (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Helena Brentani
- Laboratório de Psicobiologia (LIM23), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Orestes V Forlenza
- Laboratorio de Neurociencias (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Vanessa De Jesus Rodrigues De Paula
- Laboratório de Psicobiologia (LIM23), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Laboratorio de Neurociencias (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Martínez-González K, Islas-Hernández A, Martínez-Ezquerro JD, Bermúdez-Rattoni F, Garcia-delaTorre P. Telomere length and oxidative stress variations in a murine model of Alzheimer's disease progression. Eur J Neurosci 2020; 52:4863-4874. [PMID: 32594585 DOI: 10.1111/ejn.14877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and ageing is its major risk factor. Changes in telomere length have been associated with ageing and some degenerative diseases. Our aim was to explore some of the molecular changes caused by the progression of AD in a transgenic murine model (3xTg-AD; B6; 129-Psen1 <tm1Mpm> Tg (APPSwe, tauP301L) 1Lfa). Telomere length was assessed by qPCR in both brain tissue and peripheral blood cells and compared between three age groups: 5, 9 and 13 months. In addition, a possible effect of oxidative stress on telomere length and AD progression was explored. Shorter telomeres were found in blood cells of older transgenic mice compared to younger and wild-type mice but no changes in telomere length in the hippocampus. An increase in oxidative stress with age was found for all strains, but no correlation was found between oxidative stress and shorter telomere length for transgenic mice. Telomere length and oxidative stress are affected by AD progression in the 3xTg-AD murine model. Changes in blood cells are more noticeable than changes in brain tissue, suggesting that systemic changes can be detected early in the disease in this murine model.
Collapse
Affiliation(s)
- Katia Martínez-González
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, CDMX, Coyoacán, México
| | - Azul Islas-Hernández
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, CDMX, Coyoacán, México
| | - José Darío Martínez-Ezquerro
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México City, México
| | - Federico Bermúdez-Rattoni
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Paola Garcia-delaTorre
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| |
Collapse
|
14
|
Pisanu C, Congiu D, Manchia M, Caria P, Cocco C, Dettori T, Frau DV, Manca E, Meloni A, Nieddu M, Noli B, Pinna F, Robledo R, Sogos V, Ferri GL, Carpiniello B, Vanni R, Bocchetta A, Severino G, Ardau R, Chillotti C, Zompo MD, Squassina A. Differences in telomere length between patients with bipolar disorder and controls are influenced by lithium treatment. Pharmacogenomics 2020; 21:533-540. [DOI: 10.2217/pgs-2020-0028] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: To assess the role of lithium treatment in the relationship between bipolar disorder (BD) and leukocyte telomere length (LTL). Materials & methods: We compared LTL between 131 patients with BD, with or without a history of lithium treatment, and 336 controls. We tested the association between genetically determined LTL and BD in two large genome-wide association datasets. Results: Patients with BD with a history lithium treatment showed longer LTL compared with never-treated patients (p = 0.015), and similar LTL compared with controls. Patients never treated with lithium showed shorter LTL compared with controls (p = 0.029). Mendelian randomization analysis showed no association between BD and genetically determined LTL. Conclusion: Our data support previous findings showing that long-term lithium treatment might protect against telomere shortening.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Donatella Congiu
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Mirko Manchia
- Unit of Psychiatry, Department of Public Health, Clinical & Molecular Medicine, University of Cagliari, Cagliari, 09100, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Paola Caria
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Cristina Cocco
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Tinuccia Dettori
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Daniela Virginia Frau
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Elias Manca
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Anna Meloni
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Mariella Nieddu
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Barbara Noli
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Federica Pinna
- Unit of Psychiatry, Department of Public Health, Clinical & Molecular Medicine, University of Cagliari, Cagliari, 09100, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Renato Robledo
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Valeria Sogos
- Department of Biomedical Sciences, Section of Cytomorphology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Gian Luca Ferri
- Department of Biomedical Sciences, NEF Laboratory, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Bernardo Carpiniello
- Unit of Psychiatry, Department of Public Health, Clinical & Molecular Medicine, University of Cagliari, Cagliari, 09100, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Roberta Vanni
- Department of Biomedical Sciences, Unit of Biology & Genetics, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Alberto Bocchetta
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Giovanni Severino
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Maria Del Zompo
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
- Unit of Clinical Pharmacology, University Hospital Agency of Cagliari, Cagliari, 09100, Italy
| | - Alessio Squassina
- Department of Biomedical Science, Section of Neuroscience & Clinical Pharmacology, University of Cagliari, Monserrato, Cagliari, 09042, Italy
| |
Collapse
|
15
|
Lundberg M, Millischer V, Backlund L, Martinsson L, Stenvinkel P, Sellgren CM, Lavebratt C, Schalling M. Lithium and the Interplay Between Telomeres and Mitochondria in Bipolar Disorder. Front Psychiatry 2020; 11:586083. [PMID: 33132941 PMCID: PMC7553080 DOI: 10.3389/fpsyt.2020.586083] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 01/06/2023] Open
Abstract
Bipolar disorder is a severe psychiatric disorder which affects more than 1% of the world's population and is a leading cause of disability among young people. For the past 50 years, lithium has been the drug of choice for maintenance treatment of bipolar disorder due to its potent ability to prevent both manic and depressive episodes as well as suicide. However, though lithium has been associated with a multitude of effects within different cellular pathways and biological systems, its specific mechanism of action in stabilizing mood remains largely elusive. Mitochondrial dysfunction and telomere shortening have been implicated in both the pathophysiology of bipolar disorder and as targets of lithium treatment. Interestingly, it has in recent years become clear that these phenomena are intimately linked, partly through reactive oxygen species signaling and the subcellular translocation and non-canonical actions of telomerase reverse transcriptase. In this review, we integrate the current understanding of mitochondrial dysfunction, oxidative stress and telomere shortening in bipolar disorder with documented effects of lithium. Moreover, we propose that lithium's mechanism of action is intimately connected with the interdependent regulation of mitochondrial bioenergetics and telomere maintenance.
Collapse
Affiliation(s)
- Martin Lundberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lena Backlund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lina Martinsson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Healthcare Services, Region Stockholm, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Carl M Sellgren
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Healthcare Services, Region Stockholm, Stockholm, Sweden.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Fessel J. Prevention of Alzheimer's disease by treating mild cognitive impairment with combinations chosen from eight available drugs. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:780-788. [PMID: 31763432 PMCID: PMC6861553 DOI: 10.1016/j.trci.2019.09.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several hundred clinical trials of initially promising drugs have failed to produce meaningful clinical improvement of Alzheimer's disease (AD), which is probably because there are at least 25 biochemical pathways known to be aberrant that underpin the disease, and unless there is a single drug that addresses all or most of them, even promising drugs if given alone are unlikely to succeed. Because so many pathways are potentially at fault, it is quite possible that no treatment might succeed. However, because amnestic mild cognitive impairment (aMCI) often precedes AD and, assuming that those with aMCI who progress to AD commence with insufficient risk factors for AD but accrue them later, then it is likely that fewer pathways need addressing in aMCI than in AD to either prevent progression of aMCI to AD or effect its reversion. Published reports show that eight drugs, that is, dantrolene, erythropoietin, lithium, memantine, minocycline, piracetam, riluzole, and silymarin, address many of the pathways underlying MCI and AD. Among those eight drugs, combinations between either two or three of them have combined nonoverlapping actions that benefit enough of the approximately 25 pathways at fault so that their convergent efficacy has the potential to prevent aMCI from progressing to AD. The combinations should be subjected to a clinical trial in persons with aMCI to establish their safety and efficacy.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California San Francisco, San Francisco, CA
| |
Collapse
|
17
|
Pisanu C, Tsermpini EE, Skokou M, Kordou Z, Gourzis P, Assimakopoulos K, Congiu D, Meloni A, Balasopoulos D, Patrinos GP, Squassina A. Leukocyte telomere length is reduced in patients with major depressive disorder. Drug Dev Res 2019; 81:268-273. [DOI: 10.1002/ddr.21612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical PharmacologyUniversity of Cagliari Cagliari Italy
| | | | - Maria Skokou
- Psychiatric Clinic, Patras General Hospital Patras Greece
| | - Zoe Kordou
- Department of PharmacyUniversity of Patras School of Health Sciences Patras Greece
| | - Philippos Gourzis
- Department of MedicineUniversity of Patras School of Health Sciences Patras Greece
| | | | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical PharmacologyUniversity of Cagliari Cagliari Italy
| | - Anna Meloni
- Department of Biomedical Sciences, Section of Neuroscience and Clinical PharmacologyUniversity of Cagliari Cagliari Italy
| | | | - George P. Patrinos
- Department of PharmacyUniversity of Patras School of Health Sciences Patras Greece
- Department of PathologyUnited Arab Emirates University, College of Medicine and Health Sciences Al‐Ain UAE
- Zayed Center of Health SciencesUnited Arab Emirates University Al‐Ain UAE
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical PharmacologyUniversity of Cagliari Cagliari Italy
| |
Collapse
|
18
|
Hampel H, Lista S, Mango D, Nisticò R, Perry G, Avila J, Hernandez F, Geerts H, Vergallo A. Lithium as a Treatment for Alzheimer’s Disease: The Systems Pharmacology Perspective. J Alzheimers Dis 2019; 69:615-629. [DOI: 10.3233/jad-190197] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Harald Hampel
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, F-75013, Paris, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, F-75013, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institute of Memory and Alzheimer’s Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Dalila Mango
- Laboratory of Neuropharmacology, European Brain Research Institute, Rita Levi-Montalcini Foundation, Rome, Italy
| | - Robert Nisticò
- Laboratory of Neuropharmacology, European Brain Research Institute, Rita Levi-Montalcini Foundation, Rome, Italy
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - George Perry
- College of Sciences, One UTSA Circle, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jesus Avila
- Centro de Biologia Molecular “Severo Ochoa”, Consejo Superior de Investigaciones, Cientificas, Universidad Autonoma de Madrid, C/ Nicolas Cabrera, 1. Campus de Cantoblanco, 28049, Madrid, Spain
- Networking Research Center on Neurodegenerative, Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Felix Hernandez
- Centro de Biologia Molecular “Severo Ochoa”, Consejo Superior de Investigaciones, Cientificas, Universidad Autonoma de Madrid, C/ Nicolas Cabrera, 1. Campus de Cantoblanco, 28049, Madrid, Spain
- Networking Research Center on Neurodegenerative, Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Hugo Geerts
- In silico Biosciences, Computational Neuropharmacology, Berwyn, PA, USA
| | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l’hôpital, F-75013, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institute of Memory and Alzheimer’s Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | | |
Collapse
|
19
|
Squassina A, Pisanu C, Vanni R. Mood Disorders, Accelerated Aging, and Inflammation: Is the Link Hidden in Telomeres? Cells 2019; 8:cells8010052. [PMID: 30650526 PMCID: PMC6356466 DOI: 10.3390/cells8010052] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 12/26/2022] Open
Abstract
Mood disorders are associated with an increased risk of aging-related diseases, which greatly contribute to the excess morbidity and mortality observed in affected individuals. Clinical and molecular findings also suggest that mood disorders might be characterized by a permanent state of low-grade inflammation. At the cellular level, aging translates into telomeres shortening. Intriguingly, inflammation and telomere shortening show a bidirectional association: a pro-inflammatory state seems to contribute to aging and telomere dysfunction, and telomere attrition is able to induce low-grade inflammation. Several independent studies have reported shorter telomere length and increased levels of circulating inflammatory cytokines in mood disorders, suggesting a complex interplay between altered inflammatory–immune responses and telomere dynamics in the etiopathogenesis of these disorders. In this review, we critically discuss studies investigating the role of telomere attrition and inflammation in the pathogenesis and course of mood disorders, and in pharmacological treatments with psychotropic medications.
Collapse
Affiliation(s)
- Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato Cagliari, Italy.
- Department of Psychiatry, Dalhousie University, Halifax, NS B3H 2E2, Canada.
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato Cagliari, Italy.
- Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, 752 39 Uppsala, Sweden.
| | - Roberta Vanni
- Department of Biomedical Sciences, Unit of Biology and Genetics, University of Cagliari, 09042 Monserrato Cagliari, Italy.
| |
Collapse
|