1
|
Tan Y, Tang W, Xiao W, Huang R, Li X, Peng W, Yan K, Cao Y, Zeng Y, Kang J. lncRNA-associated ceRNA network revealing the potential regulatory roles of ferroptosis and immune infiltration in Alzheimer's disease. Front Aging Neurosci 2023; 15:1105690. [PMID: 36875702 PMCID: PMC9979855 DOI: 10.3389/fnagi.2023.1105690] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/05/2023] [Indexed: 02/18/2023] Open
Abstract
Background Alzheimer's disease (AD) is the most common form of dementia characterized by a prominent cognitive deterioration of sufficient magnitude to impair daily living. Increasing studies indicate that non-coding RNAs (ncRNAs) are involved in ferroptosis and AD progression. However, the role of ferroptosis-related ncRNAs in AD remains unexplored. Methods We obtained the intersection of differentially expressed genes in GSE5281 (brain tissue expression profile of patients with AD) from the GEO database and ferroptosis-related genes (FRGs) from the ferrDb database. Least absolute shrinkage and selection operator model along with weighted gene co-expression network analysis screened for FRGs highly associated with AD. Results A total of five FRGs were identified and further validated in GSE29378 (area under the curve = 0.877, 95% confidence interval = 0.794-0.960). A competing endogenous RNA (ceRNA) network of ferroptosis-related hub genes (EPT1, KLHL24, LRRFIP1, CXCL2 and CD44) was subsequently constructed to explore the regulatory mechanism between hub genes, lncRNAs and miRNAs. Finally, CIBERSORT algorithms were used to unravel the immune cell infiltration landscape in AD and normal samples. M1 macrophages and mast cells were more infiltrated whereas memory B cells were less infiltrated in AD samples than in normal samples. Spearman's correlation analysis revealed that LRRFIP1 was positively correlated with M1 macrophages (r = -0.340, P < 0.001) whereas ferroptosis-related lncRNAs were negatively correlated with immune cells, wherein miR7-3HG correlated with M1 macrophages and NIFK-AS1, EMX2OS and VAC14-AS1 correlated with memory B cells (|r| > 0.3, P < 0.001). Conclusion We constructed a novel ferroptosis-related signature model including mRNAs, miRNAs and lncRNAs, and characterized its association with immune infiltration in AD. The model provides novel ideas for the pathologic mechanism elucidation and targeted therapy development of AD.
Collapse
Affiliation(s)
- Yejun Tan
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Wang Tang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wenbiao Xiao
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Roujie Huang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xin Li
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Kuipo Yan
- Department of Cardiology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, China
| | - Yuan Cao
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yi Zeng
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jin Kang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Fišar Z. Linking the Amyloid, Tau, and Mitochondrial Hypotheses of Alzheimer's Disease and Identifying Promising Drug Targets. Biomolecules 2022; 12:1676. [PMID: 36421690 PMCID: PMC9687482 DOI: 10.3390/biom12111676] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/23/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023] Open
Abstract
Damage or loss of brain cells and impaired neurochemistry, neurogenesis, and synaptic and nonsynaptic plasticity of the brain lead to dementia in neurodegenerative diseases, such as Alzheimer's disease (AD). Injury to synapses and neurons and accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles are considered the main morphological and neuropathological features of AD. Age, genetic and epigenetic factors, environmental stressors, and lifestyle contribute to the risk of AD onset and progression. These risk factors are associated with structural and functional changes in the brain, leading to cognitive decline. Biomarkers of AD reflect or cause specific changes in brain function, especially changes in pathways associated with neurotransmission, neuroinflammation, bioenergetics, apoptosis, and oxidative and nitrosative stress. Even in the initial stages, AD is associated with Aβ neurotoxicity, mitochondrial dysfunction, and tau neurotoxicity. The integrative amyloid-tau-mitochondrial hypothesis assumes that the primary cause of AD is the neurotoxicity of Aβ oligomers and tau oligomers, mitochondrial dysfunction, and their mutual synergy. For the development of new efficient AD drugs, targeting the elimination of neurotoxicity, mutual potentiation of effects, and unwanted protein interactions of risk factors and biomarkers (mainly Aβ oligomers, tau oligomers, and mitochondrial dysfunction) in the early stage of the disease seems promising.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague, Czech Republic
| |
Collapse
|
3
|
Wu T, He J, Shen X. Study of metabolomics in selenium deprived Przewalski's Gazelle ( Procapra przewalskii). Br J Nutr 2022; 128:549-560. [PMID: 34511139 DOI: 10.1017/s000711452100355x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
To understand why Procapra przewalskii does not show the same white myopathy as sheep in Se-deficient regions and to provide reference for feeding nutrition level of artificial population and selection of wild reintroduction areas in the later period, a Se-deficient model was established. The mineral elements content, physiological and biochemical parameters in blood and serum metabonomics were determined. In the Se-deficient group compared with the control group, the Se content was highly significantly lower (P < 0·01), and the Cu content was significantly higher (P < 0·05). The activity of glutathione peroxidase was significantly lower (P < 0·05), but total superoxide dismutase was significantly higher (P < 0·05). By matching the mass spectrum data of compounds with the Kyoto Encyclopedia of Genes and Genomes (KEGG database), eighty-six types of differential metabolites in the serum were identified. The main metabolic pathways included secondary bile acid biosynthesis, biosynthesis of unsaturated fatty acids and pyrimidine metabolism. Further analysis showed that there were seven different metabolites in pyrimidine metabolism pathway between the two groups. And there was no significant difference in erythrocyte, Hb and total antioxidant capacity between the two groups (P > 0·05). The above results showed that the differential metabolism of substances exhibited complementary functions, thus alleviating some adverse effects and resulting normal activities of P. przewalskii can be carried out under the condition of dietary Se content lower than 0·05 mg/kg.
Collapse
Affiliation(s)
- Ting Wu
- College of Life Science, Southwest University of Science and Technology, Mianyang, Sichuan621010, People's Republic of China
- Feng Guang De Laboratory, Tie Qi Li Shi Group, Mianyang, Sichuan, People's Republic of China
| | - Jian He
- College of Life Science, Southwest University of Science and Technology, Mianyang, Sichuan621010, People's Republic of China
- Feng Guang De Laboratory, Tie Qi Li Shi Group, Mianyang, Sichuan, People's Republic of China
| | - Xiaoyun Shen
- College of Life Science, Southwest University of Science and Technology, Mianyang, Sichuan621010, People's Republic of China
- World Bank Poverty Alleviation Project Office in Guizhou, Southwest China, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
4
|
Seminotti B, Grings M, Tucci P, Leipnitz G, Saso L. Nuclear Factor Erythroid-2-Related Factor 2 Signaling in the Neuropathophysiology of Inherited Metabolic Disorders. Front Cell Neurosci 2021; 15:785057. [PMID: 34955754 PMCID: PMC8693715 DOI: 10.3389/fncel.2021.785057] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/05/2021] [Indexed: 01/14/2023] Open
Abstract
Inherited metabolic disorders (IMDs) are rare genetic conditions that affect multiple organs, predominantly the central nervous system. Since treatment for a large number of IMDs is limited, there is an urgent need to find novel therapeutical targets. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a transcription factor that has a key role in controlling the intracellular redox environment by regulating the expression of antioxidant enzymes and several important genes related to redox homeostasis. Considering that oxidative stress along with antioxidant system alterations is a mechanism involved in the neuropathophysiology of many IMDs, this review focuses on the current knowledge about Nrf2 signaling dysregulation observed in this group of disorders characterized by neurological dysfunction. We review here Nrf2 signaling alterations observed in X-linked adrenoleukodystrophy, glutaric acidemia type I, hyperhomocysteinemia, and Friedreich’s ataxia. Additionally, beneficial effects of different Nrf2 activators are shown, identifying a promising target for treatment of patients with these disorders. We expect that this article stimulates research into the investigation of Nrf2 pathway involvement in IMDs and the use of potential pharmacological modulators of this transcription factor to counteract oxidative stress and exert neuroprotection.
Collapse
Affiliation(s)
- Bianca Seminotti
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mateus Grings
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Guilhian Leipnitz
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Postgraduate Program in Biological Sciences: Physiology, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Abstract
Platelet mitochondria can be used in the study of mitochondrial dysfunction in various complex diseases and can help in finding biological markers for diagnosing the disease, monitoring its course and the effects of treatment. The aim of this chapter was to describe in detail the method of measuring mitochondrial respiration in platelets using high-resolution respirometry. The described method was successfully used for the study of mitochondrial dysfunction in neuropsychiatric diseases.
Collapse
|
6
|
Abstract
Mitochondria are targets of newly synthesized drugs and being tested for the treatment of various diseases caused or accompanied by disruption of cellular bioenergetics. In drug development, it is necessary to test for drug-induced changes in mitochondrial enzyme activity that may be related to therapeutic or adverse drug effects. Measurement of drug effect on mitochondrial oxygen consumption kinetics and/or protective effects of drugs against calcium-induced inhibition of the mitochondrial respiration can be used for the study mitochondrial toxicity and neuroprotective effects of drugs. Supposing that the drug-induced inhibition of the mitochondrial respiratory rate and/or individual mitochondrial complexes is associated with adverse drug effects, the effects of drugs on mitochondrial respiration in isolated mitochondria allow selection of novel molecules that are relatively safe for mitochondrial toxicity.
Collapse
Affiliation(s)
- Jana Hroudová
- First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Zdeněk Fišar
- First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| |
Collapse
|
7
|
Błaszczyk JW. Energy Metabolism Decline in the Aging Brain-Pathogenesis of Neurodegenerative Disorders. Metabolites 2020; 10:metabo10110450. [PMID: 33171879 PMCID: PMC7695180 DOI: 10.3390/metabo10110450] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/31/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
There is a growing body of evidencethat indicates that the aging of the brain results from the decline of energy metabolism. In particular, the neuronal metabolism of glucose declines steadily, resulting in a growing deficit of adenosine triphosphate (ATP) production-which, in turn, limits glucose access. This vicious circle of energy metabolism at the cellular level is evoked by a rising deficiency of nicotinamide adenine dinucleotide (NAD) in the mitochondrial salvage pathway and subsequent impairment of the Krebs cycle. A decreasing NAD level also impoverishes the activity of NAD-dependent enzymes that augments genetic errors and initiate processes of neuronal degeneration and death.This sequence of events is characteristic of several brain structures in which neurons have the highest energy metabolism. Neurons of the cerebral cortex and basal ganglia with long unmyelinated axons and these with numerous synaptic junctions are particularly prone to senescence and neurodegeneration. Unfortunately, functional deficits of neurodegeneration are initially well-compensated, therefore, clinical symptoms are recognized too late when the damages to the brain structures are already irreversible. Therefore, future treatment strategies in neurodegenerative disorders should focus on energy metabolism and compensation age-related NAD deficit in neurons. This review summarizes the complex interrelationships between metabolic processes on the systemic and cellular levels and provides directions on how to reduce the risk of neurodegeneration and protect the elderly against neurodegenerative diseases.
Collapse
Affiliation(s)
- Janusz Wiesław Błaszczyk
- Department of Human Motor Behavior, Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
| |
Collapse
|
8
|
Vidoni ED, Choi IY, Lee P, Reed G, Zhang N, Pleen J, Mahnken JD, Clutton J, Becker A, Sherry E, Bothwell R, Anderson H, Harris RA, Brooks W, Wilkins HM, Mosconi L, Burns JM, Swerdlow RH. Safety and target engagement profile of two oxaloacetate doses in Alzheimer's patients. Alzheimers Dement 2020; 17:7-17. [PMID: 32715609 DOI: 10.1002/alz.12156] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/19/2020] [Accepted: 06/28/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Brain bioenergetics are defective in Alzheimer's disease (AD). Preclinical studies find oxaloacetate (OAA) enhances bioenergetics, but human safety and target engagement data are lacking. METHODS We orally administered 500 or 1000 mg OAA, twice daily for 1 month, to AD participants (n = 15 each group) and monitored safety and tolerability. To assess brain metabolism engagement, we performed fluorodeoxyglucose positron emission tomography (FDG PET) and magnetic resonance spectroscopy before and after the intervention. We also assessed pharmacokinetics and cognitive performance. RESULTS Both doses were safe and tolerated. Compared to the lower dose, the higher dose benefited FDG PET glucose uptake across multiple brain regions (P < .05), and the higher dose increased parietal and frontoparietal glutathione (P < .05). We did not demonstrate consistent blood level changes and cognitive scores did not improve. CONCLUSIONS 1000 mg OAA, taken twice daily for 1 month, is safe in AD patients and engages brain energy metabolism.
Collapse
Affiliation(s)
- Eric D Vidoni
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - In-Young Choi
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Hoglund Biomedical Imaging Center, Kansas City, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Phil Lee
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Hoglund Biomedical Imaging Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Radiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Gregory Reed
- Department of Pharmacology and Toxicology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Na Zhang
- Department of Pharmacology and Toxicology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Joseph Pleen
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Jonathan D Mahnken
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Biostatistics, Kansas University Medical Center, Kansas City, Kansas, USA
| | | | - Annette Becker
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
| | - Erica Sherry
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
| | | | - Heidi Anderson
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
| | - Robert A Harris
- Biochemistry and Molecular Biology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - William Brooks
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Hoglund Biomedical Imaging Center, Kansas City, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| | - Jeffrey M Burns
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
- Biochemistry and Molecular Biology, Kansas University Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
9
|
Avram S, Mernea M, Limban C, Borcan F, Chifiriuc C. Potential Therapeutic Approaches to Alzheimer's Disease By Bioinformatics, Cheminformatics And Predicted Adme-Tox Tools. Curr Neuropharmacol 2020; 18:696-719. [PMID: 31885353 PMCID: PMC7536829 DOI: 10.2174/1570159x18666191230120053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/24/2019] [Accepted: 12/28/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is considered a severe, irreversible and progressive neurodegenerative disorder. Currently, the pharmacological management of AD is based on a few clinically approved acethylcholinesterase (AChE) and N-methyl-D-aspartate (NMDA) receptor ligands, with unclear molecular mechanisms and severe side effects. METHODS Here, we reviewed the most recent bioinformatics, cheminformatics (SAR, drug design, molecular docking, friendly databases, ADME-Tox) and experimental data on relevant structurebiological activity relationships and molecular mechanisms of some natural and synthetic compounds with possible anti-AD effects (inhibitors of AChE, NMDA receptors, beta-secretase, amyloid beta (Aβ), redox metals) or acting on multiple AD targets at once. We considered: (i) in silico supported by experimental studies regarding the pharmacological potential of natural compounds as resveratrol, natural alkaloids, flavonoids isolated from various plants and donepezil, galantamine, rivastagmine and memantine derivatives, (ii) the most important pharmacokinetic descriptors of natural compounds in comparison with donepezil, memantine and galantamine. RESULTS In silico and experimental methods applied to synthetic compounds led to the identification of new AChE inhibitors, NMDA antagonists, multipotent hybrids targeting different AD processes and metal-organic compounds acting as Aβ inhibitors. Natural compounds appear as multipotent agents, acting on several AD pathways: cholinesterases, NMDA receptors, secretases or Aβ, but their efficiency in vivo and their correct dosage should be determined. CONCLUSION Bioinformatics, cheminformatics and ADME-Tox methods can be very helpful in the quest for an effective anti-AD treatment, allowing the identification of novel drugs, enhancing the druggability of molecular targets and providing a deeper understanding of AD pathological mechanisms.
Collapse
Affiliation(s)
| | - Maria Mernea
- Address correspondence to this author at the Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95th Spl. Independentei, Bucharest, Romania; Tel/Fax: ++4-021-318-1573; E-mail:
| | | | | | | |
Collapse
|
10
|
Perez Ortiz JM, Swerdlow RH. Mitochondrial dysfunction in Alzheimer's disease: Role in pathogenesis and novel therapeutic opportunities. Br J Pharmacol 2019; 176:3489-3507. [PMID: 30675901 PMCID: PMC6715612 DOI: 10.1111/bph.14585] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022] Open
Abstract
Dysfunction of cell bioenergetics is a common feature of neurodegenerative diseases, the most common of which is Alzheimer's disease (AD). Disrupted energy utilization implicates mitochondria at its nexus. This review summarizes some of the evidence that points to faulty mitochondrial function in AD and highlights past and current therapeutic development efforts. Classical neuropathological hallmarks of disease (β-amyloid and τ) and sporadic AD risk genes (APOE) may trigger mitochondrial disturbance, yet mitochondrial dysfunction may incite pathology. Preclinical and clinical efforts have overwhelmingly centred on the amyloid pathway, but clinical trials have yet to reveal clear-cut benefits. AD therapies aimed at mitochondrial dysfunction are few and concentrate on reversing oxidative stress and cell death pathways. Novel research efforts aimed at boosting mitochondrial and bioenergetic function offer an alternative treatment strategy. Enhancing cell bioenergetics in preclinical models may yield widespread favourable effects that could benefit persons with AD. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Judit M. Perez Ortiz
- University of Kansas Alzheimer's Disease CenterFairwayKSUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Russell H. Swerdlow
- University of Kansas Alzheimer's Disease CenterFairwayKSUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKSUSA
- Department of Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityKSUSA
- Department of Biochemistry and Molecular BiologyUniversity of Kansas Medical CenterKansas CityKSUSA
| |
Collapse
|
11
|
Jiang S, Shao C, Tang F, Wang W, Zhu X. Dynamin-like protein 1 cleavage by calpain in Alzheimer's disease. Aging Cell 2019; 18:e12912. [PMID: 30767411 PMCID: PMC6516178 DOI: 10.1111/acel.12912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/30/2018] [Accepted: 12/22/2018] [Indexed: 01/25/2023] Open
Abstract
Abnormal mitochondrial dynamics contributes to mitochondrial dysfunction in Alzheimer's disease (AD), yet the underlying mechanism remains elusive. In the current study, we reported that DLP1, the key mitochondrial fission GTPase, is a substrate of calpain which produced specific N-terminal DLP1 cleavage fragments. In addition, various AD-related insults such as exposure to glutamate, soluble amyloid-β oligomers, or reagents inducing tau hyperphosphorylation (i.e., okadaic acid) led to calpain-dependent cleavage of DLP1 in primary cortical neurons. DLP1 cleavage fragments were found in cortical neurons of CRND8 APP transgenic mice which can be inhibited by calpeptin, a potent small molecule inhibitor of calpain. Importantly, these N-terminal DLP1 fragments were also present in the human brains, and the levels of both full-length and N-terminal fragments of DLP1 and the full-length and calpain-specific cleavage product of spectrin were significantly reduced in AD brains along with significantly increased calpain. These results suggest that calpain-dependent cleavage is at least one of the posttranscriptional mechanisms that contribute to the dysregulation of mitochondrial dynamics in AD.
Collapse
Affiliation(s)
- Sirui Jiang
- Department of PathologyCase Western Reserve UniversityClevelandOhio
| | - Changjuan Shao
- Department of PathologyCase Western Reserve UniversityClevelandOhio
| | - Fangqiang Tang
- Department of PathologyCase Western Reserve UniversityClevelandOhio
| | - Wenzhang Wang
- Department of PathologyCase Western Reserve UniversityClevelandOhio
| | - Xiongwei Zhu
- Department of PathologyCase Western Reserve UniversityClevelandOhio
| |
Collapse
|
12
|
Abstract
Decades of research indicate mitochondria from Alzheimer's disease (AD) patients differ from those of non-AD individuals. Initial studies revealed structural differences, and subsequent studies showed functional deficits. Observations of structure and function changes prompted investigators to consider the consequences, significance, and causes of AD-related mitochondrial dysfunction. Currently, extensive research argues mitochondria may mediate, drive, or contribute to a variety of AD pathologies. The perceived significance of these mitochondrial changes continues to grow, and many currently believe AD mitochondrial dysfunction represents a reasonable therapeutic target. Debate continues over the origin of AD mitochondrial changes. Some argue amyloid-β (Aβ) induces AD mitochondrial dysfunction, a view that does not challenge the amyloid cascade hypothesis and that may in fact help explain that hypothesis. Alternatively, data indicate mitochondrial dysfunction exists independent of Aβ, potentially lies upstream of Aβ deposition, and suggest a primary mitochondrial cascade hypothesis that assumes mitochondrial pathology hierarchically supersedes Aβ pathology. Mitochondria, therefore, appear at least to mediate or possibly even initiate pathologic molecular cascades in AD. This review considers studies and data that inform this area of AD research.
Collapse
Affiliation(s)
- Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center and Departments of Neurology, Molecular and Integrative Physiology, and Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
13
|
Fišar Z, Jirák R, Zvěřová M, Setnička V, Habartová L, Hroudová J, Vaníčková Z, Raboch J. Plasma amyloid beta levels and platelet mitochondrial respiration in patients with Alzheimer's disease. Clin Biochem 2019; 72:71-80. [PMID: 30954436 DOI: 10.1016/j.clinbiochem.2019.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Altered amyloid metabolism and mitochondrial dysfunction play key roles in the development of Alzheimer's disease (AD). We asked whether an association exists between disturbed platelet mitochondrial respiration and the plasma concentrations of Aβ40 and Aβ42 in patients with AD. DESIGN AND METHODS Plasma Aβ40 and Aβ42 concentrations and mitochondrial respiration in intact and permeabilized platelets were measured in 50 patients with AD, 15 patients with vascular dementia and 25 control subjects. A pilot longitudinal study was performed to monitor the progression of AD in a subgroup 11 patients with AD. RESULTS The mean Aβ40, Aβ42 and Aβ42/Aβ40 levels were not significantly altered in patients with AD compared with controls. The mitochondrial respiratory rate in intact platelets was significantly reduced in patients with AD compared to controls, particularly the basal respiratory rate, maximum respiratory capacity, and respiratory reserve; however, the flux control ratio for basal respiration was increased. A correlation between the plasma Aβ42 concentration and mitochondrial respiration in both intact and permeabilized platelets differs in controls and patients with AD. CONCLUSIONS Based on our data, (1) mitochondrial respiration in intact platelets, but not the Aβ level itself, may be included in a panel of biomarkers for AD; (2) dysfunctional mitochondrial respiration in platelets is not explained by changes in plasma Aβ concentrations; and (3) the association between mitochondrial respiration in platelets and plasma Aβ levels differs in patients with AD and controls. The results supported the hypothesis that mitochondrial dysfunction is the primary factor contributing to the development of AD.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Martina Zvěřová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Vladimír Setnička
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czech Republic.
| | - Lucie Habartová
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czech Republic.
| | - Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Zdislava Vaníčková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| |
Collapse
|
14
|
Desler C, Lillenes MS, Tønjum T, Rasmussen LJ. The Role of Mitochondrial Dysfunction in the Progression of Alzheimer's Disease. Curr Med Chem 2019; 25:5578-5587. [PMID: 28618998 PMCID: PMC6446443 DOI: 10.2174/0929867324666170616110111] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 01/02/2017] [Accepted: 01/02/2017] [Indexed: 11/22/2022]
Abstract
The current molecular understanding of Alzheimer's disease (AD) has still not resulted in successful interventions. Mitochondrial dysfunction of the AD brain is currently emerging as a hallmark of this disease. One mitochondrial function often affected in AD is oxidative phosphorylation responsible for ATP production, but also for production of reactive oxygen species (ROS) and for the de novo synthesis of pyrimidines. This paper reviews the role of mitochondrial produced ROS and pyrimidines in the aetiology of AD and their proposed role in oxidative degeneration of macromolecules, synthesis of essential phospholipids and maintenance of mitochondrial viability in the AD brain.
Collapse
Affiliation(s)
- Claus Desler
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - Meryl S Lillenes
- Healthy Brain Aging Centre (HBAC), Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Tone Tønjum
- Healthy Brain Aging Centre (HBAC), Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| |
Collapse
|
15
|
Niño SA, Morales-Martínez A, Chi-Ahumada E, Carrizales L, Salgado-Delgado R, Pérez-Severiano F, Díaz-Cintra S, Jiménez-Capdeville ME, Zarazúa S. Arsenic Exposure Contributes to the Bioenergetic Damage in an Alzheimer's Disease Model. ACS Chem Neurosci 2019; 10:323-336. [PMID: 30141907 DOI: 10.1021/acschemneuro.8b00278] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Worldwide, every year there is an increase in the number of people exposed to inorganic arsenic (iAs) via drinking water. Human populations present impaired cognitive function as a result of prenatal and childhood iAs exposure, while studies in animal models demonstrate neurobehavioral deficits accompanied by neurotransmitter, protein, and enzyme alterations. Similar impairments have been observed in close association with Alzheimer's disease (AD). In order to determine whether iAs promotes the pathophysiological progress of AD, we used the 3xTgAD mouse model. Mice were exposed to iAs in drinking water from gestation until 6 months (As-3xTgAD group) and compared with control animals without arsenic (3xTgAD group). We investigated the behavior phenotype on a test battery (circadian rhythm, locomotor behavior, Morris water maze, and contextual fear conditioning). Adenosine triphosphate (ATP), reactive oxygen species, lipid peroxidation, and respiration rates of mitochondria were evaluated, antioxidant components were detected by immunoblots, and immunohistochemical studies were performed to reveal AD markers. As-3xTgAD displayed alterations in their circadian rhythm and exhibited longer freezing time and escape latencies compared to the control group. The bioenergetic profile revealed decreased ATP levels accompanied by the decline of complex I, and an oxidant state in the hippocampus. On the other hand, the cortex showed no changes of oxidant stress and complex I; however, the antioxidant response was increased. Higher immunopositivity to amyloid isoforms and to phosphorylated tau was observed in frontal cortex and hippocampus of exposed animals. In conclusion, mitochondrial dysfunction may be one of the triggering factors through which chronic iAs exposure exacerbates brain AD-like pathology.
Collapse
Affiliation(s)
- Sandra Aurora Niño
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, CP 78210 San Luis Potosí, SLP, México
| | - Adriana Morales-Martínez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur 3877, CP 14269, México D.F., México
| | - Erika Chi-Ahumada
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosi, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Leticia Carrizales
- Centro de Investigación Aplicada en Ambiente y Salud, CIACYT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Roberto Salgado-Delgado
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava Martínez S/N, CP 78290 San Luis Potosí, SLP, Mexico
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”, Insurgentes Sur 3877, CP 14269, México D.F., México
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, CP 76230 Juriquilla, Querétaro, México
| | - María E. Jiménez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosi, Av. Venustiano Carranza 2405, CP 78210 San Luis Potosí, SLP, México
| | - Sergio Zarazúa
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, CP 78210 San Luis Potosí, SLP, México
| |
Collapse
|
16
|
Dienel GA. Metabolomic Assays of Postmortem Brain Extracts: Pitfalls in Extrapolation of Concentrations of Glucose and Amino Acids to Metabolic Dysregulation In Vivo in Neurological Diseases. Neurochem Res 2018; 44:2239-2260. [DOI: 10.1007/s11064-018-2611-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 01/03/2023]
|
17
|
Yashin AI, Fang F, Kovtun M, Wu D, Duan M, Arbeev K, Akushevich I, Kulminski A, Culminskaya I, Zhbannikov I, Yashkin A, Stallard E, Ukraintseva S. Hidden heterogeneity in Alzheimer's disease: Insights from genetic association studies and other analyses. Exp Gerontol 2018; 107:148-160. [PMID: 29107063 PMCID: PMC5920782 DOI: 10.1016/j.exger.2017.10.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/20/2017] [Accepted: 10/22/2017] [Indexed: 02/08/2023]
Abstract
Despite evident success in clarifying many important features of Alzheimer's disease (AD) the efficient methods of its prevention and treatment are not yet available. The reasons are likely to be the fact that AD is a multifactorial and heterogeneous health disorder with multiple alternative pathways of disease development and progression. The availability of genetic data on individuals participated in longitudinal studies of aging health and longevity, as well as on participants of cross-sectional case-control studies allow for investigating genetic and non-genetic connections with AD and to link the results of these analyses with research findings obtained in clinical, experimental, and molecular biological studies of this health disorder. The objective of this paper is to perform GWAS of AD in several study populations and investigate possible roles of detected genetic factors in developing AD hallmarks and in other health disorders. The data collected in the Framingham Heart Study (FHS), Cardiovascular Health Study (CHS), Health and Retirement Study (HRS) and Late Onset Alzheimer's Disease Family Study (LOADFS) were used in these analyses. The logistic regression and Cox's regression were used as statistical models in GWAS. The results of analyses confirmed strong associations of genetic variants from well-known genes APOE, TOMM40, PVRL2 (NECTIN2), and APOC1 with AD. Possible roles of these genes in pathological mechanisms resulting in development of hallmarks of AD are described. Many genes whose connection with AD was detected in other studies showed nominally significant associations with this health disorder in our study. The evidence on genetic connections between AD and vulnerability to infection, as well as between AD and other health disorders, such as cancer and type 2 diabetes, were investigated. The progress in uncovering hidden heterogeneity in AD would be substantially facilitated if common mechanisms involved in development of AD, its hallmarks, and AD related chronic conditions were investigated in their mutual connection.
Collapse
Affiliation(s)
- Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA.
| | - Fang Fang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Mikhail Kovtun
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Matt Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Igor Akushevich
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Ilya Zhbannikov
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Arseniy Yashkin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA.
| |
Collapse
|
18
|
Mecocci P, Baroni M, Senin U, Boccardi V. Brain Aging and Late-Onset Alzheimer’s Disease: A Matter of Increased Amyloid or Reduced Energy? J Alzheimers Dis 2018; 64:S397-S404. [DOI: 10.3233/jad-179903] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Patrizia Mecocci
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| | - Marta Baroni
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| | - Umberto Senin
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| | - Virginia Boccardi
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| |
Collapse
|
19
|
Mattson MP, Arumugam TV. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab 2018; 27:1176-1199. [PMID: 29874566 PMCID: PMC6039826 DOI: 10.1016/j.cmet.2018.05.011] [Citation(s) in RCA: 677] [Impact Index Per Article: 96.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023]
Abstract
During aging, the cellular milieu of the brain exhibits tell-tale signs of compromised bioenergetics, impaired adaptive neuroplasticity and resilience, aberrant neuronal network activity, dysregulation of neuronal Ca2+ homeostasis, the accrual of oxidatively modified molecules and organelles, and inflammation. These alterations render the aging brain vulnerable to Alzheimer's and Parkinson's diseases and stroke. Emerging findings are revealing mechanisms by which sedentary overindulgent lifestyles accelerate brain aging, whereas lifestyles that include intermittent bioenergetic challenges (exercise, fasting, and intellectual challenges) foster healthy brain aging. Here we provide an overview of the cellular and molecular biology of brain aging, how those processes interface with disease-specific neurodegenerative pathways, and how metabolic states influence brain health.
Collapse
Affiliation(s)
- Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
20
|
Srivastava R, Faust T, Ramos A, Ishizuka K, Sawa A. Dynamic Changes of the Mitochondria in Psychiatric Illnesses: New Mechanistic Insights From Human Neuronal Models. Biol Psychiatry 2018; 83:751-760. [PMID: 29486891 PMCID: PMC6469392 DOI: 10.1016/j.biopsych.2018.01.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/18/2017] [Accepted: 01/07/2018] [Indexed: 02/06/2023]
Abstract
Mitochondria play a crucial role in neuronal function, especially in energy production, the generation of reactive oxygen species, and calcium signaling. Multiple lines of evidence have suggested the possible involvement of mitochondrial deficits in major psychiatric disorders, such as schizophrenia and bipolar disorder. This review will outline the current understanding of the physiological role of mitochondria and their dysfunction under pathological conditions, particularly in psychiatric disorders. The current knowledge about mitochondrial deficits in these disorders is somewhat limited because of the lack of effective methods to dissect dynamic changes in functional deficits that are directly associated with psychiatric conditions. Human neuronal cell model systems have been dramatically developed in recent years with the use of stem cell technology, and these systems may be key tools for overcoming this dilemma and improving our understanding of the dynamic changes in the mitochondrial deficits in patients with psychiatric disorders. We introduce recent discoveries from new experimental models and conclude the discussion by referring to future perspectives. We emphasize the significance of combining studies of human neuronal cell models with those of other experimental systems, including animal models.
Collapse
Affiliation(s)
- Rupali Srivastava
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Travis Faust
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Adriana Ramos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Koko Ishizuka
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
21
|
Jiang S, Nandy P, Wang W, Ma X, Hsia J, Wang C, Wang Z, Niu M, Siedlak SL, Torres S, Fujioka H, Xu Y, Lee HG, Perry G, Liu J, Zhu X. Mfn2 ablation causes an oxidative stress response and eventual neuronal death in the hippocampus and cortex. Mol Neurodegener 2018; 13:5. [PMID: 29391029 PMCID: PMC5796581 DOI: 10.1186/s13024-018-0238-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/24/2018] [Indexed: 12/20/2022] Open
Abstract
Background Mitochondria are the organelles responsible for energy metabolism and have a direct impact on neuronal function and survival. Mitochondrial abnormalities have been well characterized in Alzheimer Disease (AD). It is believed that mitochondrial fragmentation, due to impaired fission and fusion balance, likely causes mitochondrial dysfunction that underlies many aspects of neurodegenerative changes in AD. Mitochondrial fission and fusion proteins play a major role in maintaining the health and function of these important organelles. Mitofusion 2 (Mfn2) is one such protein that regulates mitochondrial fusion in which mutations lead to the neurological disease. Methods To examine whether and how impaired mitochondrial fission/fusion balance causes neurodegeneration in AD, we developed a transgenic mouse model using the CAMKII promoter to knockout neuronal Mfn2 in the hippocampus and cortex, areas significantly affected in AD. Results Electron micrographs of neurons from these mice show swollen mitochondria with cristae damage and mitochondria membrane abnormalities. Over time the Mfn2 cKO model demonstrates a progression of neurodegeneration via mitochondrial morphological changes, oxidative stress response, inflammatory changes, and loss of MAP2 in dendrites, leading to severe and selective neuronal death. In this model, hippocampal CA1 neurons were affected earlier and resulted in nearly total loss, while in the cortex, progressive neuronal death was associated with decreased cortical size. Conclusions Overall, our findings indicate that impaired mitochondrial fission and fusion balance can cause many of the neurodegenerative changes and eventual neuron loss that characterize AD in the hippocampus and cortex which makes it a potential target for treatment strategies for AD. Electronic supplementary material The online version of this article (10.1186/s13024-018-0238-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sirui Jiang
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Priya Nandy
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Wenzhang Wang
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Xiaopin Ma
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Jeffrey Hsia
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Chunyu Wang
- Department of Neurology, the second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Zhenlian Wang
- School of Pharmaceutical Engineering & Life Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Mengyue Niu
- Department of Neurology & Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sandra L Siedlak
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Sandy Torres
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA
| | - Hisashi Fujioka
- Electron Microscopy Core Facility, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ying Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14222, USA
| | - Hyoung-Gon Lee
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - George Perry
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Jun Liu
- Department of Neurology & Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH, USA.
| |
Collapse
|
22
|
Kim DI, Lee KH, Oh JY, Kim JS, Han HJ. Relationship Between β-Amyloid and Mitochondrial Dynamics. Cell Mol Neurobiol 2017; 37:955-968. [PMID: 27766447 DOI: 10.1007/s10571-016-0434-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/13/2016] [Indexed: 01/29/2023]
Abstract
Mitochondria as dynamic organelles undergo morphological changes through the processes of fission and fusion which are major factors regulating their functions. A disruption in the balance of mitochondrial dynamics induces functional disorders in mitochondria such as failed energy production and the generation of reactive oxygen species, which are closely related to pathophysiological changes associated with Alzheimer's disease (AD). Recent studies have demonstrated a relationship between abnormalities in mitochondrial dynamics and impaired mitochondrial function, clarifying the effects of morphofunctional aberrations which promote neuronal cell death in AD. Several possible signaling pathways have been suggested for a better understanding of the mechanism behind the key molecules regulating mitochondrial morphologies. However, the exact machinery involved in mitochondrial dynamics still has yet to be elucidated. This paper reviews the current knowledge on signaling mechanisms involved in mitochondrial dynamics and the significance of mitochondrial dynamics in controlling associated functions in neurodegenerative diseases, particularly in AD.
Collapse
Affiliation(s)
- Dah Ihm Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Ki Hoon Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Ji Young Oh
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
- BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
23
|
Hohnholt MC, Blumrich EM, Waagepetersen HS, Dringen R. The tricarboxylic acid cycle activity in cultured primary astrocytes is strongly accelerated by the protein tyrosine kinase inhibitor tyrphostin 23. Neurochem Int 2016; 102:13-21. [PMID: 27894844 DOI: 10.1016/j.neuint.2016.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 11/28/2022]
Abstract
Tyrphostin 23 (T23) is a well-known inhibitor of protein tyrosine kinases and has been considered as potential anti-cancer drug. T23 was recently reported to acutely stimulate the glycolytic flux in primary cultured astrocytes. To investigate whether T23 also affects the tricarboxylic acid (TCA) cycle, we incubated primary rat astrocyte cultures with [U-13C]glucose in the absence or the presence of 100 μM T23 for 2 h and analyzed the 13C metabolite pattern. These incubation conditions did not compromise cell viability and confirmed that the presence of T23 doubled glycolytic lactate production. In addition, T23-treatment strongly increased the molecular carbon labeling of the TCA cycle intermediates citrate, succinate, fumarate and malate, and significantly increased the incorporation of 13C-labelling into the amino acids glutamate, glutamine and aspartate. These results clearly demonstrate that, in addition to glycolysis, also the mitochondrial TCA cycle is strongly accelerated after exposure of astrocytes to T23, suggesting that a protein tyrosine kinase may be involved in the regulation of the TCA cycle in astrocytes.
Collapse
Affiliation(s)
- Michaela C Hohnholt
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Eva-Maria Blumrich
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, PO. Box 330440, D-28334 Bremen, Germany; Centre for Environmental Research and Sustainable Technology, Leobener Strasse, D-28359 Bremen, Germany
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, PO. Box 330440, D-28334 Bremen, Germany; Centre for Environmental Research and Sustainable Technology, Leobener Strasse, D-28359 Bremen, Germany
| |
Collapse
|
24
|
Vidali S, Chéret J, Giesen M, Haeger S, Alam M, Watson REB, Langton AK, Klinger M, Knuever J, Funk W, Kofler B, Paus R. Thyroid Hormones Enhance Mitochondrial Function in Human Epidermis. J Invest Dermatol 2016; 136:2003-2012. [PMID: 27349864 DOI: 10.1016/j.jid.2016.05.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/17/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
Since it is unknown whether thyroid hormones (THs) regulate mitochondrial function in human epidermis, we treated organ-cultured human skin, or isolated cultured human epidermal keratinocytes, with triiodothyronine (100 pmol/L) or thyroxine (100 nmol/L). Both THs significantly increased protein expression of the mitochondrially encoded cytochrome C oxidase I (MTCO1), complex I activity, and the number of perinuclear mitochondria. Triiodothyronine also increased mitochondrial transcription factor A (TFAM) protein expression, and thyroxine stimulated complex II/IV activity. Increased mitochondrial function can correlate with increased reactive oxygen species production, DNA damage, and accelerated tissue aging. However, THs neither raised reactive oxygen species production or matrix metalloproteinase-1, -2 and -9 activity nor decreased sirtuin1 (Sirt1) immunoreactivity. Instead, triiodothyronine increased sirtuin-1, fibrillin-1, proliferator-activated receptor-gamma 1-alpha (PGC1α), collagen I and III transcription, and thyroxine decreased cyclin-dependent kinase inhibitor 2A (p16(ink4)) expression in organ-cultured human skin. Moreover, TH treatment increased intracutaneous fibrillin-rich microfibril and collagen III deposition and decreased mammalian target of rapamycin (mTORC1/2) expression ex vivo. This identifies THs as potent endocrine stimulators of mitochondrial function in human epidermis, which down-regulates rather than enhance the expression of skin aging-related biomarkers ex vivo. Therefore, topically applied THs deserve further exploration as candidate agents for treating skin conditions characterized by reduced mitochondrial function.
Collapse
Affiliation(s)
- Silvia Vidali
- Department of Dermatology, University of Luebeck, Luebeck, Germany; Research Program for Receptor Biochemistry and Tumor Metabolism, Laura Bassi Centre of Expertise-THERAPEP, Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Jérémy Chéret
- Department of Dermatology, University of Münster, Münster, Germany
| | - Melanie Giesen
- Henkel Beauty Care, Henkel AG and Co. KgaA, Düsseldorf, Germany
| | - Swantje Haeger
- Department of Dermatology, University of Luebeck, Luebeck, Germany
| | - Majid Alam
- Department of Dermatology, University of Münster, Münster, Germany
| | - Rachel E B Watson
- Center for Dermatology Research, University of Manchester, Manchester, UK
| | - Abigail K Langton
- Center for Dermatology Research, University of Manchester, Manchester, UK
| | | | - Jana Knuever
- Department of Dermatology, University of Luebeck, Luebeck, Germany
| | | | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Laura Bassi Centre of Expertise-THERAPEP, Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Ralf Paus
- Department of Dermatology, University of Münster, Münster, Germany; Center for Dermatology Research, University of Manchester, Manchester, UK.
| |
Collapse
|
25
|
Hullinger R, Puglielli L. Molecular and cellular aspects of age-related cognitive decline and Alzheimer's disease. Behav Brain Res 2016; 322:191-205. [PMID: 27163751 DOI: 10.1016/j.bbr.2016.05.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/19/2016] [Accepted: 05/03/2016] [Indexed: 01/14/2023]
Abstract
As the population of people aged 60 or older continues to rise, it has become increasingly important to understand the molecular basis underlying age-related cognitive decline. In fact, a better understanding of aging biology will help us identify ways to maintain high levels of cognitive functioning throughout the aging process. Many cellular and molecular aspects of brain aging are shared with other organ systems; however, certain age-related changes are unique to the nervous system due to its structural, cellular and molecular complexity. Importantly, the brain appears to show differential changes throughout the aging process, with certain regions (e.g. frontal and temporal regions) being more vulnerable than others (e.g. brain stem). Within the medial temporal lobe, the hippocampus is especially susceptible to age-related changes. The important role of the hippocampus in age-related cognitive decline and in vulnerability to disease processes such as Alzheimer's disease has prompted this review, which will focus on the complexity of changes that characterize aging, and on the molecular connections that exist between normal aging and Alzheimer's disease. Finally, it will discuss behavioral interventions and emerging insights for promoting healthy cognitive aging.
Collapse
Affiliation(s)
- Rikki Hullinger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Geriatric Research Education Clinical Center, VA Medical Center, Madison, WI 53705, USA.
| |
Collapse
|