51
|
Robustness of the Krebs Cycle under Physiological Conditions and in Cancer: New Clues for Evaluating Metabolism-Modifying Drug Therapies. Biomedicines 2022; 10:biomedicines10051199. [PMID: 35625935 PMCID: PMC9138339 DOI: 10.3390/biomedicines10051199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
The Krebs cycle in cells that contain mitochondria is necessary for both energy production and anabolic processes. In given cell/condition, the Krebs cycle is dynamic but remains at a steady state. In this article, we first aimed at comparing the properties of a closed cycle versus the same metabolism in a linear array. The main finding is that, unlike a linear metabolism, the closed cycle can reach a steady state (SS) regardless of the nature and magnitude of the disturbance. When the cycle is modeled with input and output reactions, the “open” cycle is robust and reaches a steady state but with exceptions that lead to sustained accumulation of intermediate metabolites, i.e., conditions at which no SS can be achieved. The modeling of the cycle in cancer, trying to obtain marked reductions in flux, shows that these reductions are limited and therefore the Warburg effect is moderate at most. In general, our results of modeling the cycle in different conditions and looking for the achievement, or not, of SS, suggest that the cycle may have a regulation, not yet discovered, to go from an open cycle to a closed one. Said regulation could allow for reaching the steady state, thus avoiding the unwanted effects derived from the aberrant accumulation of metabolites in the mitochondria. The information in this paper might be useful to evaluate metabolism-modifying medicines.
Collapse
|
52
|
Analysis of Mitochondrial Function in Cell Membranes as Indicator of Tissue Vulnerability to Drugs in Humans. Biomedicines 2022; 10:biomedicines10050980. [PMID: 35625717 PMCID: PMC9138415 DOI: 10.3390/biomedicines10050980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Drug side effects are one of the main reasons for treatment withdrawal during clinical trials. Reactive oxygen species formation is involved in many of the drug side effects, mainly by interacting with the components of the cellular respiration. Thus, the early detection of these effects in the drug discovery process is a key aspect for the optimization of pharmacological research. To this end, the superoxide formation of a series of drugs and compounds with antidepressant, antipsychotic, anticholinergic, narcotic, and analgesic properties was evaluated in isolated bovine heart membranes and on cell membrane microarrays from a collection of human tissues, together with specific inhibitors of the mitochondrial electron transport chain. Fluphenazine and PB28 promoted similar effects to those of rotenone, but with lower potency, indicating a direct action on mitochondrial complex I. Moreover, nefazodone, a drug withdrawn from the market due to its mitochondrial hepatotoxic effects, evoked the highest superoxide formation in human liver cell membranes, suggesting the potential of this technology to anticipate adverse effects in preclinical phases.
Collapse
|
53
|
Dexmedetomidine Can Enhance PINK1/Parkin-Mediated Mitophagy in MPTP-Induced PD Mice Model by Activating AMPK. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7511393. [PMID: 35528513 PMCID: PMC9068320 DOI: 10.1155/2022/7511393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/05/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by the degeneration of dopaminergic (DA) neurons in the substantia nigra (SN). Our previous study has shown that dexmedetomidine (Dex) can protect mitochondrial function and reduce apoptosis in MPP+-induced SH-SY5Y cells. Evidences have shown that mitophagy is related to the development of PD. In this study, we investigated whether Dex can enhance mitophagy in MPTP-induced mice to play a neuroprotective effect. In our experiment, mice were injected with MPTP 30 mg/kg intraperitoneally for 5 consecutive days to establish a PD subacute model. Dex (30, 50, and 100 μg/kg) was injected intraperitoneally 30 minutes before each injection of MPTP, respectively. Our results showed that Dex (50 μg/kg) most significantly attenuated MPTP-induced motor dysfunction and restored TH-positive neurons in the SN, increased the expression of the antiapoptotic protein Bcl-2, and decreased the expression of apoptotic proteins cleaved casepase3, cleaved casepase9, and Bax. Moreover, Dex increased the activity of mitochondrial Complexes I-IV and decreased the level of oxidative stress, manifesting as decreased MDA levels and increased SOD and GSH-PX levels. Besides, under transmission electron microscopy, Dex increased the mitophagosome which is an autophagosome with a mitochondrion-like structure inside under the electron microscope. In addition, Dex could also increase the expression of mitophagy-related proteins p-AMPK, LC3II/I, PINK1, and Parkin and decrease P62. However, after using Compound C (CC, 10 mg/kg, AMPK inhibitor), the effects of Dex on increasing PINK1/Parkin-induced mitophagy and neuroprotection were attenuated. In conclusion, Dex may improve mitochondrial function by activating AMPK to enhance PINK1/Parkin-induced mitophagy, thereby protecting dopaminergic neurons.
Collapse
|
54
|
Rhein Ameliorates Cognitive Impairment in an APP/PS1 Transgenic Mouse Model of Alzheimer's Disease by Relieving Oxidative Stress through Activating the SIRT1/PGC-1 α Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2524832. [PMID: 35360200 PMCID: PMC8964225 DOI: 10.1155/2022/2524832] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/08/2022] [Indexed: 01/05/2023]
Abstract
Mitochondrial oxidative stress plays an important role in the pathogenesis of Alzheimer's disease (AD). Recently, antioxidant therapy has been considered an effective strategy for the treatment of AD. Our previous work discovered that rhein relieved mitochondrial oxidative stress in β-amyloid (Aβ) oligomer-induced primary neurons by improving the sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor gamma coactivator 1-alpha- (PGC-1α-) regulated mitochondrial biogenesis. While encouraging results have been provided, mechanisms underlying the beneficial effect of rhein on AD are yet to be elucidated in vivo. In this study, we evaluated the therapeutic effect of rhein on an APP/PS1 transgenic (APP/PS1) mouse model of AD and explored its antioxidant mechanisms. As a result, rhein significantly reduced Aβ burden and neuroinflammation and eventually ameliorated cognitive impairment in APP/PS1 mice. Moreover, rhein reversed oxidative stress in the brain of APP/PS1 mice and protected neurons from oxidative stress-associated apoptosis. Further study revealed that rhein promoted mitochondrial biogenesis against oxidative stress by upregulating SIRT1 and its downstream PGC-1α as well as nuclear respiratory factor 1. Improved mitochondrial biogenesis not only increased the activity of superoxide dismutase to scavenge excess reactive oxygen species (ROS) but also repaired mitochondria by mitochondrial fusion to inhibit the production of ROS from the electron transport chain. Notably, the exposure of rhein in the brain analyzed by tissue distribution study indicated that rhein could permeate into the brain to exert its therapeutic effects. In conclusion, these findings drive rhein to serve as a promising therapeutic antioxidant for the treatment of AD. Our research highlights the therapeutic efficacy for AD through regulating mitochondrial biogenesis via the SIRT1/PGC-1α pathway.
Collapse
|
55
|
Athanasaki A, Melanis K, Tsantzali I, Stefanou MI, Ntymenou S, Paraskevas SG, Kalamatianos T, Boutati E, Lambadiari V, Voumvourakis KI, Stranjalis G, Giannopoulos S, Tsivgoulis G, Paraskevas GP. Type 2 Diabetes Mellitus as a Risk Factor for Alzheimer’s Disease: Review and Meta-Analysis. Biomedicines 2022; 10:biomedicines10040778. [PMID: 35453527 PMCID: PMC9029855 DOI: 10.3390/biomedicines10040778] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 01/16/2023] Open
Abstract
Alzheimer’s disease is the most common type of dementia, reaching 60–80% of case totals, and is one of the major global causes of the elderly population’s decline in functionality concerning daily life activities. Epidemiological research has already indicated that, in addition to several others metabolic factors, diabetes mellitus type 2 is a risk factor of Alzheimer’s disease. Many molecular pathways have been described, and at the same time, there are clues that suggest the connection between type 2 diabetes mellitus and Alzheimer’s disease, through specific genes, autophagy, and even inflammatory pathways. A systematic review with meta-analysis was conducted, and its main goal was to reveal the multilevel connection between these diseases.
Collapse
Affiliation(s)
- Athanasia Athanasaki
- Department of Neurology, Evangelismos Hospital, 10676 Athens, Greece; (A.A.); (S.N.)
| | - Konstantinos Melanis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Ioanna Tsantzali
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Maria Ioanna Stefanou
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Sofia Ntymenou
- Department of Neurology, Evangelismos Hospital, 10676 Athens, Greece; (A.A.); (S.N.)
| | - Sotirios G. Paraskevas
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Theodosis Kalamatianos
- 1st Department of Neurosurgery, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (T.K.); (G.S.)
| | - Eleni Boutati
- 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (E.B.); (V.L.)
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (E.B.); (V.L.)
| | - Konstantinos I. Voumvourakis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - George Stranjalis
- 1st Department of Neurosurgery, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (T.K.); (G.S.)
| | - Sotirios Giannopoulos
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Georgios Tsivgoulis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - George P. Paraskevas
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
- Correspondence: ; Tel.: +30-2105832466
| |
Collapse
|
56
|
Manickam DS. Delivery of mitochondria via extracellular vesicles – A new horizon in drug delivery. J Control Release 2022; 343:400-407. [DOI: 10.1016/j.jconrel.2022.01.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/16/2022]
|
57
|
Effects of DISC1 on Alzheimer's disease cell models assessed by iTRAQ proteomics analysis. Biosci Rep 2022; 42:230594. [PMID: 34981809 PMCID: PMC8753346 DOI: 10.1042/bsr20211150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a form of neurodegenerative disease in the elderly with no cure at present. In a previous study, we found that the scaffold protein, disrupted in Schizophrenia 1 (DISC1) is down-regulated in the AD brains, and ectopic expression of DISC1 can delay the progression of AD by protecting synaptic plasticity and down-regulating BACE1. However, the underlying mechanisms remain not to be elucidated. In the present study, we compared the proteomes of normal and DISC1high AD cells expressing the amyloid precursor protein (APP) using isobaric tag for relative and absolute quantitation (iTRAQ) and mass spectrometry (MS). The differentially expressed proteins (DEPs) were identified, and the protein–protein interaction (PPI) network was constructed to identify the interacting partners of DISC1. Based on the interaction scores, NDE1, GRM3, PTGER3 and KATNA1 were identified as functionally or physically related to DISC1, and may therefore regulate AD development. The DEPs were functionally annotated by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases with the DAVID software, and the Non-supervised Orthologous Groups (eggNOG) database was used to determine their evolutionary relationships. The DEPs were significantly enriched in microtubules and mitochondria-related pathways. Gene set enrichment analysis (GSEA) was performed to identify genes and pathways that are activated when DISC1 is overexpressed. Our findings provide novel insights into the regulatory mechanisms underlying DISC1 function in AD.
Collapse
|
58
|
Disrupted expression of mitochondrial NCLX sensitizes neuroglial networks to excitotoxic stimuli and renders synaptic activity toxic. J Biol Chem 2021; 298:101508. [PMID: 34942149 PMCID: PMC8808183 DOI: 10.1016/j.jbc.2021.101508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023] Open
Abstract
The mitochondrial sodium/calcium/lithium exchanger (NCLX) is an important mediator of calcium extrusion from mitochondria. In this study, we tested the hypothesis that physiological expression levels of NCLX are essential for maintaining neuronal resilience in the face of excitotoxic challenge. Using a short hairpin RNA (shRNA)-mediated approach, we showed that reduced NCLX expression exacerbates neuronal mitochondrial calcium dysregulation, mitochondrial membrane potential (ΔΨm) breakdown, and reactive oxygen species (ROS) generation during excitotoxic stimulation of primary hippocampal cultures. Moreover, NCLX knockdown-which affected both neurons and glia-resulted not only in enhanced neurodegeneration following an excitotoxic insult, but also in neuronal and astrocytic cell death under basal conditions. Our data also revealed that synaptic activity, which promotes neuroprotective signaling, can become lethal upon NCLX depletion; expression of NCLX-targeted shRNA impaired the clearance of mitochondrial calcium following action potential bursts and was associated both with ΔΨmbreakdown and substantial neurodegeneration in hippocampal cultures undergoing synaptic activity. Finally, we showed that NCLX knockdown within the hippocampal cornu ammonis 1 (CA1) region in vivo causes substantial neuro- and astrodegeneration. In summary, we demonstrated that dysregulated NCLX expression not only sensitizes neuroglial networks to excitotoxic stimuli but notably also renders otherwise neuroprotective synaptic activity toxic. These findings may explain the emergence of neuro- and astrodegeneration in patients with disorders characterized by disrupted NCLX expression or function, and suggest that treatments aimed at enhancing or restoring NCLX function may prevent central nervous system damage in these disease states.
Collapse
|
59
|
Sawant N, Morton H, Kshirsagar S, Reddy AP, Reddy PH. Mitochondrial Abnormalities and Synaptic Damage in Huntington's Disease: a Focus on Defective Mitophagy and Mitochondria-Targeted Therapeutics. Mol Neurobiol 2021; 58:6350-6377. [PMID: 34519969 DOI: 10.1007/s12035-021-02556-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022]
Abstract
Huntington's disease (HD) is a fatal and pure genetic disease with a progressive loss of medium spiny neurons (MSN). HD is caused by expanded polyglutamine repeats in the exon 1 of HD gene. Clinically, HD is characterized by chorea, seizures, involuntary movements, dystonia, cognitive decline, intellectual impairment, and emotional disturbances. Several years of intense research revealed that multiple cellular changes, including defective axonal transport, protein-protein interactions, defective bioenergetics, calcium dyshomeostasis, NMDAR activation, synaptic damage, mitochondrial abnormalities, and selective loss of medium spiny neurons are implicated in HD. Recent research on mutant huntingtin (mHtt) and mitochondria has found that mHtt interacts with the mitochondrial division protein, dynamin-related protein 1 (DRP1), enhances GTPase DRP1 enzymatic activity, and causes excessive mitochondrial fragmentation and abnormal distribution, leading to defective axonal transport of mitochondria and selective synaptic degeneration. Recent research also revealed that failure to remove dead and/or dying mitochondria is an early event in the disease progression. Currently, efforts are being made to reduce abnormal protein interactions and enhance synaptic mitophagy as therapeutic strategies for HD. The purpose of this article is to discuss recent research in HD progression. This article also discusses recent developments of cell and mouse models, cellular changes, mitochondrial abnormalities, DNA damage, bioenergetics, oxidative stress, mitophagy, and therapeutics strategies in HD.
Collapse
Affiliation(s)
- Neha Sawant
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hallie Morton
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neurology, Department of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Internal Medicine, Cell Biology & Biochemistry, Public Health and School of Health Professions, Texas Tech University Health Sciences Center, Neuroscience & Pharmacology3601 4th Street, NeurologyLubbock, TX, 79430, USA.
| |
Collapse
|
60
|
Huang SS, Sheng YC, Jiang YY, Liu N, Lin MM, Wu JC, Liang ZQ, Qin ZH, Wang Y. TIGAR plays neuroprotective roles in KA-induced excitotoxicity through reducing neuroinflammation and improving mitochondrial function. Neurochem Int 2021; 152:105244. [PMID: 34826530 DOI: 10.1016/j.neuint.2021.105244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/30/2021] [Accepted: 11/19/2021] [Indexed: 02/05/2023]
Abstract
Excitotoxicity refers to the ability of excessive extracellular excitatory amino acids to damage neurons via receptor activation. It is a crucial pathogenetic process in neurodegenerative diseases. TP53 is confirmed to be involved in excitotoxicity. It is demonstrated that TP53 induced glycolysis and apoptotic regulator (TIGAR)-regulated metabolic pathway can protect against neuronal injury. However, the role of TIGAR in excitotoxicity and specific mechanisms is still unknown. In this study, an in vivo excitotoxicity model was constructed via stereotypical kainic acid (KA) injection into the striatum of mice. KA reduced TIGAR expression levels, neuroinflammatory responses and mitochondrial dysfunction. TIGAR overexpression could reverse KA-induced neuronal injury by reducing neuroinflammation and improving mitochondrial function, thereby exerting neuroprotective effects. Therefore, this study could provide a potential therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Si-Si Huang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi-Chao Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi-Yue Jiang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Na Liu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Miao-Miao Lin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jun-Chao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhong-Qin Liang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
61
|
Tang S, Davoudi Z, Wang G, Xu Z, Rehman T, Prominski A, Tian B, Bratlie KM, Peng H, Wang Q. Soft materials as biological and artificial membranes. Chem Soc Rev 2021; 50:12679-12701. [PMID: 34636824 DOI: 10.1039/d1cs00029b] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The past few decades have seen emerging growth in the field of soft materials for synthetic biology. This review focuses on soft materials involved in biological and artificial membranes. The biological membranes discussed here are mainly those involved in the structure and function of cells and organelles. As building blocks in medicine, non-native membranes including nanocarriers (NCs), especially liposomes and DQAsomes, and polymeric membranes for scaffolds are constructed from amphiphilic combinations of lipids, proteins, and carbohydrates. Artificial membranes can be prepared using synthetic, soft materials and molecules and then incorporated into structures through self-organization to form micelles or niosomes. The modification of artificial membranes can be realized using traditional chemical methods such as click reactions to target the delivery of NCs and control the release of therapeutics. The biomembrane, a lamellar structure inlaid with ion channels, receptors, lipid rafts, enzymes, and other functional units, separates cells and organelles from the environment. An active domain inserted into the membrane and organelles for energy conversion and cellular communication can target disease by changing the membrane's composition, structure, and fluidity and affecting the on/off status of the membrane gates. The biological membrane targets analyzing pathological mechanisms and curing complex diseases, which inspires us to create NCs with artificial membranes.
Collapse
Affiliation(s)
- Shukun Tang
- Department of Pharmaceutics, Daqing Branch, Harbin Medical University, Research and Development of Natural Products Key Laboratory of Harbin Medical University, 39 Xin Yang Road, Daqing, 163319, China.
| | - Zahra Davoudi
- Department of Chemical and Biological Engineering, Iowa State University, 1014 Sweeney Hall, Ames, IA 50011, USA.
| | - Guangtian Wang
- Department of Pharmaceutics, Daqing Branch, Harbin Medical University, Research and Development of Natural Products Key Laboratory of Harbin Medical University, 39 Xin Yang Road, Daqing, 163319, China.
| | - Zihao Xu
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
| | - Tanzeel Rehman
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
| | - Aleksander Prominski
- The James Franck Institute, Department of Chemistry, The Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, USA
| | - Bozhi Tian
- The James Franck Institute, Department of Chemistry, The Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, USA
| | - Kaitlin M Bratlie
- Department of Chemical and Biological Engineering, Iowa State University, 1014 Sweeney Hall, Ames, IA 50011, USA. .,Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Branch, Harbin Medical University, Research and Development of Natural Products Key Laboratory of Harbin Medical University, 39 Xin Yang Road, Daqing, 163319, China.
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, 1014 Sweeney Hall, Ames, IA 50011, USA.
| |
Collapse
|
62
|
Zimmerman MA, Hall M, Qi Q, Mehta SL, Chen G, Li PA. Ubisol Coenzyme Q10 promotes mitochondrial biogenesis in HT22 cells challenged by glutamate. Exp Ther Med 2021; 22:1295. [PMID: 34630650 PMCID: PMC8461507 DOI: 10.3892/etm.2021.10730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/15/2021] [Indexed: 11/25/2022] Open
Abstract
Glutamate-induced excitotoxicity is a well-recognized cause of neuronal cell death. Nutritional supplementation with Coenzyme Q10 (CoQ10) has been previously demonstrated to serve neuro-protective effects against glutamate-induced excitotoxicity. The aim of the present study was to determine whether the protective effect of CoQ10 against glutamate toxicity could be attributed to stimulating mitochondrial biogenesis. Mouse hippocampal neuronal HT22 cells were incubated with glutamate with or without ubisol Q10. The results revealed that glutamate significantly decreased levels of mitochondrial biogenesis related proteins, including peroxisome proliferator-activated receptor gamma coactivator (PGC)-1α and nuclear respiratory factor (NRF)2. Additionally, glutamate reduced mitochondrial biogenesis, as determined using a mitochondrial biogenesis kit. Pretreatment with CoQ10 prevented decreases in phosphorylated (p)-Akt, p-cAMP response element-binding protein, PGC-1α, NRF2 and mitochondrial transcription factor A, increasing mitochondrial biogenesis. Taken together, the results described a novel mechanism of CoQ10-induced neuroprotection and indicated a central role for mitochondrial biogenesis in protecting against glutamate-induced excitotoxicity.
Collapse
Affiliation(s)
- Mary A Zimmerman
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.,Department of Biology, University of Wisconsin La Crosse, La Crosse, WI 54601, USA
| | - Mia Hall
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Qi Qi
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.,Department of Neurology, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, Ningxia 750004, P.R. China
| | - Suresh L Mehta
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.,Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Guisheng Chen
- Department of Neurology, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, Yinchuan, Ningxia 750004, P.R. China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
63
|
Kashyap P, Shikha D, Thakur M, Aneja A. Functionality of apigenin as a potent antioxidant with emphasis on bioavailability, metabolism, action mechanism and in vitro and in vivo studies: A review. J Food Biochem 2021; 46:e13950. [PMID: 34569073 DOI: 10.1111/jfbc.13950] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/06/2021] [Accepted: 09/12/2021] [Indexed: 01/18/2023]
Abstract
Numerous diseases such as cancer, diabetes, cardiovascular, neurodegenerative diseases, etc. are linked with overproduction of reactive oxygen species (ROS) and oxidative stress. Apigenin (5,7,4'-trihydroxyflavone) is a widely distributed flavonoid, responsible for antioxidant potential and chelating redox active metals. Being present as glycosides or polymers, the apigenin degrades to variable amount in the digestive tract; during processing, its activity is also reduced due to high temperature or Fe/Cu addition. Although its metabolism remains elusive, enteric absorption occurs sufficiently to reduce plasma indices of oxidant status. Delayed clearance in plasma and slow liver decomposition enhance its systematic bioavailability. Antioxidant mechanism of apigenin includes: oxidant enzymes inhibition, modulation of redox signaling pathways (NF-kB, Nrf2, MAPK, and P13/Akt), reinforcing enzymatic and nonenzymatic antioxidant, metal chelation, and free radical scavenging. DPPH, ORAC, ABTS, and FRAP are the major in vitro methods for determining the antioxidant potential of apigenin, whereas its protective effects in whole and living cells of animals are examined using in vivo studies. Due to limited information on antioxidant potential of apigenin, its in vitro and in vivo antioxidant effects are, therefore, discussed with action mechanism and interaction with the signaling pathways. This paper concludes that apigenin is a potent antioxidant compound to overcome the difficulties related to oxidative stress and other chronic diseases.
Collapse
Affiliation(s)
- Piyush Kashyap
- Department of Food Engineering and Technology, Sant Longowal Institute of Engineering and Technology, Longowal, Punjab, India
| | - Deep Shikha
- Department of Food Technology, Bhai Gurdas Institute of Engineering and Technology, Sangrur, Punjab, India
| | - Mamta Thakur
- Department of Food Technology, School of Sciences, ITM University, Gwalior, India
| | - Ashwin Aneja
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
64
|
Xu S, Zhang X, Liu C, Liu Q, Chai H, Luo Y, Li S. Role of Mitochondria in Neurodegenerative Diseases: From an Epigenetic Perspective. Front Cell Dev Biol 2021; 9:688789. [PMID: 34513831 PMCID: PMC8429841 DOI: 10.3389/fcell.2021.688789] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Mitochondria, the centers of energy metabolism, have been shown to participate in epigenetic regulation of neurodegenerative diseases. Epigenetic modification of nuclear genes encoding mitochondrial proteins has an impact on mitochondria homeostasis, including mitochondrial biogenesis, and quality, which plays role in the pathogenesis of neurodegenerative diseases like Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. On the other hand, intermediate metabolites regulated by mitochondria such as acetyl-CoA and NAD+, in turn, may regulate nuclear epigenome as the substrate for acetylation and a cofactor of deacetylation, respectively. Thus, mitochondria are involved in epigenetic regulation through bidirectional communication between mitochondria and nuclear, which may provide a new strategy for neurodegenerative diseases treatment. In addition, emerging evidence has suggested that the abnormal modification of mitochondria DNA contributes to disease development through mitochondria dysfunction. In this review, we provide an overview of how mitochondria are involved in epigenetic regulation and discuss the mechanisms of mitochondria in regulation of neurodegenerative diseases from epigenetic perspective.
Collapse
Affiliation(s)
- Sutong Xu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xi Zhang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chenming Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiulu Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huazhen Chai
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuping Luo
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Siguang Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
65
|
A nanophytosomes formulation based on elderberry anthocyanins and Codium lipids to mitigate mitochondrial dysfunctions. Biomed Pharmacother 2021; 143:112157. [PMID: 34517282 DOI: 10.1016/j.biopha.2021.112157] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 01/12/2023] Open
Abstract
The development of nanomedicines to modulate the mitochondrial function is a great scientific challenge since mitochondrial dysfunction is a pathological hallmark of many chronic diseases, including degenerative brain pathologies like Parkinson's and Alzheimer's diseases. To address this challenge, the mitochondriotropic features of the elderberry anthocyanin-enriched extract (Sambucus nigra) were combined with the self-assembling properties of the membrane polar lipids from Codium tomentosum in an innovative SC-Nanophytosomes formulation. Membrane polar lipids, obtained by a new procedure as chlorophyll-free extract, are characterized by 26% of non-phosphorus polar lipids and 74% of phospholipids (dominated by anionic lipids) containing a high degree of polyunsaturated fatty acids. The anthocyanin-enriched extract is dominated by a mixture of four cyanidin-glycosides, representing about 86% of their phenolic content. SC-Nanophytosomes engineered with 600 µM algae membrane polar lipids and 0.5 mg/L of the anthocyanin-enriched extract are nanosized vesicles (diameter =108.74 ± 24.74 nm) with a negative surface charge (Zeta potential = -46.93 ± 6.63 mV) that exhibit stability during storage at 4 ºC. In vitro assays with SH-SY5Y cells showed that SC-Nanophytosomes have the competence to target mitochondria, improving the mitochondrial respiratory chain complexes I and II and preserving the mitochondrial membrane potential in the presence of rotenone. Additionally, SC-Nanophytosomes protect SH-SY5Y cells against the toxicity induced by rotenone or glutamate. Green-fluorescent labeled SC-Nanophytosomes were used to reveal that they are mainly internalized by cells via caveola-mediated endocytosis, escape from endosome and reach the cytoplasm organelles, including mitochondria. Overall, data indicate that SC-Nanophytosomes have the potential to support a mitochondria-targeted therapy for neurodegenerative diseases.
Collapse
|
66
|
An Overview of the Nrf2/ARE Pathway and Its Role in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22179592. [PMID: 34502501 PMCID: PMC8431732 DOI: 10.3390/ijms22179592] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 12/20/2022] Open
Abstract
Nrf2 is a basic region leucine-zipper transcription factor that plays a pivotal role in the coordinated gene expression of antioxidant and detoxifying enzymes, promoting cell survival in adverse environmental or defective metabolic conditions. After synthesis, Nrf2 is arrested in the cytoplasm by the Kelch-like ECH-associated protein 1 suppressor (Keap1) leading Nrf2 to ubiquitin-dependent degradation. One Nrf2 activation mechanism relies on disconnection from the Keap1 homodimer through the oxidation of cysteine at specific sites of Keap1. Free Nrf2 enters the nucleus, dimerizes with small musculoaponeurotic fibrosarcoma proteins (sMafs), and binds to the antioxidant response element (ARE) sequence of the target genes. Since oxidative stress, next to neuroinflammation and mitochondrial dysfunction, is one of the hallmarks of neurodegenerative pathologies, a molecular intervention into Nrf2/ARE signaling and the enhancement of the transcriptional activity of particular genes are targets for prevention or delaying the onset of age-related and inherited neurogenerative diseases. In this study, we review evidence for the Nrf2/ARE-driven pathway dysfunctions leading to various neurological pathologies, such as Alzheimer’s, Parkinson’s, and Huntington’s diseases, as well as amyotrophic lateral sclerosis, and the beneficial role of natural and synthetic molecules that are able to interact with Nrf2 to enhance its protective efficacy.
Collapse
|
67
|
Shen P, Qi H. Cell Models to Evaluate Antioxidant Properties of the Phlorotannins in Brown Seaweed: A Review. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.1967379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ping Shen
- School of Food Science and Technology, Dalian Polytechnic University, National Engineering Research Center of Seafood, Dalian, P. R. China
| | - Hang Qi
- School of Food Science and Technology, Dalian Polytechnic University, National Engineering Research Center of Seafood, Dalian, P. R. China
| |
Collapse
|
68
|
Lin S, Zhou Z, Zhao H, Xu C, Guo Y, Gao S, Mei X, Tian H. TNF promotes M1 polarization through mitochondrial metabolism in injured spinal cord. Free Radic Biol Med 2021; 172:622-632. [PMID: 34252538 DOI: 10.1016/j.freeradbiomed.2021.07.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
Macrophages and microglia (M/Ms) in the injured spinal cord maintain a predominantly neurotoxic M1 phenotype that is disadvantageous to repair in the development of spinal cord injury (SCI). It has been reported that tumor necrosis factor (TNF) that polarize M/Ms toward M1 state in various disorders. In this study, we found that ablation of TNF endorsed the beneficial conversion from M1 to M2 phenotype and improved the mitochondrial metabolism in vivo and in vitro. In addition, PGC-1α that accumulates in TNF null mice, a major participant of mitochondrial metabolism, downregulated ROS activity and the expressions of M1-specific mRNA. Moreover, the absence of TNF upgraded the morphology and quantity of damaged mitochondria and rapidly switched to M2 phenotype as compare to administration of N-Acetyl-l-cysteine (NAC). Furthermore, systemic application of TPEN showed that increased ratio of M1 M/Ms. These combined results supporting predominant and prolonged TNF expression that is destructive to recovery after SCI. These results indicated that TNF would have great potential immunomodulatory for the treatment of SCI.
Collapse
Affiliation(s)
- Sen Lin
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Zipeng Zhou
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Haosen Zhao
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Chang Xu
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Yue Guo
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Shuang Gao
- Department of Basic Medical, Jinzhou Medical University, Jinzhou, PR China
| | - Xifan Mei
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China.
| | - He Tian
- Department of Basic Medical, Jinzhou Medical University, Jinzhou, PR China
| |
Collapse
|
69
|
Koklesova L, Liskova A, Samec M, Zhai K, AL-Ishaq RK, Bugos O, Šudomová M, Biringer K, Pec M, Adamkov M, Hassan STS, Saso L, Giordano FA, Büsselberg D, Kubatka P, Golubnitschaja O. Protective Effects of Flavonoids Against Mitochondriopathies and Associated Pathologies: Focus on the Predictive Approach and Personalized Prevention. Int J Mol Sci 2021; 22:ijms22168649. [PMID: 34445360 PMCID: PMC8395457 DOI: 10.3390/ijms22168649] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 01/10/2023] Open
Abstract
Multi-factorial mitochondrial damage exhibits a “vicious circle” that leads to a progression of mitochondrial dysfunction and multi-organ adverse effects. Mitochondrial impairments (mitochondriopathies) are associated with severe pathologies including but not restricted to cancers, cardiovascular diseases, and neurodegeneration. However, the type and level of cascading pathologies are highly individual. Consequently, patient stratification, risk assessment, and mitigating measures are instrumental for cost-effective individualized protection. Therefore, the paradigm shift from reactive to predictive, preventive, and personalized medicine (3PM) is unavoidable in advanced healthcare. Flavonoids demonstrate evident antioxidant and scavenging activity are of great therapeutic utility against mitochondrial damage and cascading pathologies. In the context of 3PM, this review focuses on preclinical and clinical research data evaluating the efficacy of flavonoids as a potent protector against mitochondriopathies and associated pathologies.
Collapse
Affiliation(s)
- Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (K.Z.); (R.K.A.-I.)
| | - Raghad Khalid AL-Ishaq
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (K.Z.); (R.K.A.-I.)
| | | | - Miroslava Šudomová
- Museum of Literature in Moravia, Klášter 1, 664 61 Rajhrad, Czech Republic;
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Sherif T. S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Czech Republic;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, 00185 Rome, Italy;
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (K.Z.); (R.K.A.-I.)
- Correspondence: (D.B.); (P.K.); (O.G.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1150 Brussels, Belgium
- Correspondence: (D.B.); (P.K.); (O.G.)
| | - Olga Golubnitschaja
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1150 Brussels, Belgium
- Predictive, Preventive, Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
- Correspondence: (D.B.); (P.K.); (O.G.)
| |
Collapse
|
70
|
Molaei P, Vaseghi S, Entezari M, Hashemi M, Nasehi M. The Effect of NeuroAid (MLC901) on Cholestasis-Induced Spatial Memory Impairment with Respect to the Expression of BAX, BCL-2, BAD, PGC-1α and TFAM Genes in the Hippocampus of Male Wistar Rats. Neurochem Res 2021; 46:2154-2166. [PMID: 34031842 DOI: 10.1007/s11064-021-03353-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/31/2022]
Abstract
Cholestasis is a bile flow reduction that is induced following Bile Duct Ligation (BDL). Cholestasis impairs memory and induces apoptosis. Apoptosis consists of two pathways: intrinsic and extrinsic. The intrinsic pathway is modulated by BCL-2 (B cell lymphoma-2) family proteins. BCL-2 (a pro-survival BCL-2 protein) has anti-apoptotic effect, while BAD (BCL-2-associated death) and BAX (BCL-2-associated X), the other members of BCL-2 family have pro-apoptotic effect. Furthermore, TFAM (mitochondrial transcriptional factor A) is involved in transcription and maintenance of mitochondrial DNA and PGC-1α (peroxisome proliferator-activated receptor γ coactivator-1α) is a master regulator of mitochondrial biogenesis. On the other hand, NeuroAid is a Traditional Chinese Medicine with neuroprotective and anti-apoptosis effects. In this study, we evaluated the effect of cholestasis on spatial memory and expression of BCL-2, BAD, BAX, TFAM, and PGC-1α in the hippocampus of rats. Additionally, we assessed the effect of NeuroAid on cholestasis-induced cognitive and genetic alterations. Cholestasis was induced by BDL surgery and NeuroAid was injected intraperitoneal at the dose of 0.4 mg/kg. Furthermore, spatial memory was evaluated using Morris Water Maze (MWM) apparatus. The results showed cholestasis impaired spatial memory, increased the expression of BAD and BAX, decreased the expression of TFAM and PGC-1α, and did not alter the expression of BCL-2. Also, NeuroAid decreased the expression of BAD and BAX and increased the expression of TFAM, PGC-1α, and BCL-2. In conclusion, cholestasis impaired spatial memory and increased the expression of pro-apoptotic genes. Also, cholestasis decreased the expression of TFAM and PGC-1α. Interestingly, NeuroAid restored the effects of cholestasis.
Collapse
Affiliation(s)
- Pejman Molaei
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, P.O. Box: 13145-784, Tehran, Iran
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, P.O. Box: 13145-784, Tehran, Iran.
| |
Collapse
|
71
|
Taghizadeh E, Gheibihayat SM, Taheri F, Afshani SM, Farahani N, Saberi A. LncRNAs as putative biomarkers and therapeutic targets for Parkinson's disease. Neurol Sci 2021; 42:4007-4015. [PMID: 34254198 DOI: 10.1007/s10072-021-05408-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/13/2021] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is known as one of the most common degenerative disorders related to the damage of the central nervous system (CNS). This brain disorder is also characterized by the formation of Lewy bodies in the cytoplasm of the dopaminergic neurons in the substantia nigra pars compacta (SNc), which consequently leads to motor and non-motor symptoms. With regard to the growing trend in the number of cases with PD and its effects on individuals, families, and communities, immediate treatments together with diagnostic methods are required. In this respect, long non-coding ribonucleic acids (lncRNAs) represent a large class of ncRNAs with more than 200 nucleotides in length, playing key roles in some important processes including gene expression, cell differentiation, genomic imprinting, apoptosis, and cell cycle. They are highly expressed in the CNS and previous studies have further reported that the expression profile of lncRNAs is disrupted in human diseases such as neurodegenerative disorders. Since the levels of some lncRNAs change over time in the brains of patients with PD, a number of previous studies have examined their potentials as biomarkers for this brain disorder. Therefore, the main purpose of this study was to review the advances in the related literature on lncRNAs as diagnostic, therapeutic, and prognostic biomarkers for PD.
Collapse
Affiliation(s)
- Eskandar Taghizadeh
- Department of Medical Genetics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. .,Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Seyed Mohammad Gheibihayat
- Department of Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Forough Taheri
- Islamic Azad University (Shahrekord Branch), Shahrekord, Iran
| | - Seyed Mohammadreza Afshani
- Department of Cardiology, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmeh Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alihossein Saberi
- Department of Medical Genetics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
72
|
Jadiya P, Garbincius JF, Elrod JW. Reappraisal of metabolic dysfunction in neurodegeneration: Focus on mitochondrial function and calcium signaling. Acta Neuropathol Commun 2021; 9:124. [PMID: 34233766 PMCID: PMC8262011 DOI: 10.1186/s40478-021-01224-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The cellular and molecular mechanisms that drive neurodegeneration remain poorly defined. Recent clinical trial failures, difficult diagnosis, uncertain etiology, and lack of curative therapies prompted us to re-examine other hypotheses of neurodegenerative pathogenesis. Recent reports establish that mitochondrial and calcium dysregulation occur early in many neurodegenerative diseases (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, and others. However, causal molecular evidence of mitochondrial and metabolic contributions to pathogenesis remains insufficient. Here we summarize the data supporting the hypothesis that mitochondrial and metabolic dysfunction result from diverse etiologies of neuropathology. We provide a current and comprehensive review of the literature and interpret that defective mitochondrial metabolism is upstream and primary to protein aggregation and other dogmatic hypotheses of NDDs. Finally, we identify gaps in knowledge and propose therapeutic modulation of mCa2+ exchange and mitochondrial function to alleviate metabolic impairments and treat NDDs.
Collapse
Affiliation(s)
- Pooja Jadiya
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA
| | - Joanne F Garbincius
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA.
| |
Collapse
|
73
|
Tandon A, Singh SJ, Chaturvedi RK. Nanomedicine against Alzheimer's and Parkinson's Disease. Curr Pharm Des 2021; 27:1507-1545. [PMID: 33087025 DOI: 10.2174/1381612826666201021140904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's and Parkinson's are the two most rampant neurodegenerative disorders worldwide. Existing treatments have a limited effect on the pathophysiology but are unable to fully arrest the progression of the disease. This is due to the inability of these therapeutic molecules to efficiently cross the blood-brain barrier. We discuss how nanotechnology has enabled researchers to develop novel and efficient nano-therapeutics against these diseases. The development of nanotized drug delivery systems has permitted an efficient, site-targeted, and controlled release of drugs in the brain, thereby presenting a revolutionary therapeutic approach. Nanoparticles are also being thoroughly studied and exploited for their role in the efficient and precise diagnosis of neurodegenerative conditions. We summarize the role of different nano-carriers and RNAi-conjugated nanoparticle-based therapeutics for their efficacy in pre-clinical studies. We also discuss the challenges underlying the use of nanomedicine with a focus on their route of administration, concentration, metabolism, and any toxic effects for successful therapeutics in these diseases.
Collapse
Affiliation(s)
- Ankit Tandon
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Sangh J Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Rajnish K Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| |
Collapse
|
74
|
Boley N, Patil S, Garnett EO, Li H, Chugani DC, Chang SE, Chow HM. Association Between Gray Matter Volume Variations and Energy Utilization in the Brain: Implications for Developmental Stuttering. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2021; 64:2317-2324. [PMID: 33719533 PMCID: PMC8740693 DOI: 10.1044/2020_jslhr-20-00325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/23/2020] [Accepted: 10/02/2020] [Indexed: 06/12/2023]
Abstract
Purpose The biological mechanisms underlying developmental stuttering remain unclear. In a previous investigation, we showed that there is significant spatial correspondence between regional gray matter structural anomalies and the expression of genes linked to energy metabolism. In the current study, we sought to further examine the relationship between structural anomalies in the brain in children with persistent stuttering and brain regional energy metabolism. Method High-resolution structural MRI scans were acquired from 26 persistent stuttering and 44 typically developing children. Voxel-based morphometry was used to quantify the between-group gray matter volume (GMV) differences across the whole brain. Group differences in GMV were then compared with published values for the pattern of glucose metabolism measured via F18 fluorodeoxyglucose uptake in the brains of 29 healthy volunteers using positron emission tomography. Results A significant positive correlation between GMV differences and F18 fluorodeoxyglucose uptake was found in the left hemisphere (ρ = .36, p < .01), where speech-motor and language processing are typically localized. No such correlation was observed in the right hemisphere (ρ = .05, p = .70). Conclusions Corroborating our previous gene expression studies, the results of the current study suggest a potential connection between energy metabolism and stuttering. Brain regions with high energy utilization may be particularly vulnerable to anatomical changes associated with stuttering. Such changes may be further exacerbated when there are sharp increases in brain energy utilization, which coincides with the developmental period of rapid speech/language acquisition and the onset of stuttering during childhood. Supplemental Material https://doi.org/10.23641/asha.14110454.
Collapse
Affiliation(s)
- Nathaniel Boley
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Sanath Patil
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
- Premedical-Medical Program, Eberly College of Science, The Pennsylvania State University, University Park
| | - Emily O. Garnett
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor
| | - Hua Li
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Diane C. Chugani
- Department of Communication Sciences and Disorders, College of Health Sciences, University of Delaware, Newark
| | - Soo-Eun Chang
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor
- Cognitive Imaging Research Center, Department of Radiology, College of Osteopathic Medicine, Michigan State University, East Lansing
- Department of Communicative Sciences and Disorders, College of Communication Arts and Sciences, Michigan State University, East Lansing
| | - Ho Ming Chow
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
- Department of Communication Sciences and Disorders, College of Health Sciences, University of Delaware, Newark
| |
Collapse
|
75
|
Budni J, Braga Brandão A, da Silva S, Lima Garcez M, Mina F, Bellettini-Santos T, Casagrande Zabot G, Behenck Medeiros E, Scaini G, de Oliveira J, Streck EL, Quevedo J. Oral administration of D-galactose increases brain tricarboxylic acid cycle enzymes activities in Wistar rats. Metab Brain Dis 2021; 36:1057-1067. [PMID: 33616841 DOI: 10.1007/s11011-021-00682-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 01/31/2021] [Indexed: 12/19/2022]
Abstract
D-galactose (D-gal) is a carbohydrate widely distributed in regular diets. However, D-gal administration in rodents is associated with behavioral and neurochemical alterations similar to features observed in aging. In this regard, this study aimed to investigate the effects of D-gal exposure, in different periods, in rats' brain regions' activities of creatine kinase (CK) and tricarboxylic acid (TCA) cycle enzymes. Male adult Wistar rats received D-gal (100 mg/kg, gavage) for 1, 2, 4, 6 or 8 weeks. CK and TCA enzymes' activities were evaluated in rats' prefrontal cortex and hippocampus. In general, the results showed an increase in citrate synthase (CS) and succinate dehydrogenase (SDH) activities in animals treated with D-gal compared to the control group in the prefrontal cortex and hippocampus. Also, in the fourth week, the malate dehydrogenase (MD) activity increased in the hippocampus of rats that received D-gal compared to control rats. In addition, we observed an increase in the CK activity in the prefrontal cortex and hippocampus in the first and eighth weeks of treatment in the D-gal group compared to the control group. D-gal administration orally administered modulated TCA cycle enzymes and CK activities in the prefrontal cortex and hippocampus, which were also observed in aging and neurodegenerative diseases. However, more studies using experimental models are necessary to understand better the impact and contribution of these brain metabolic abnormalities associated with D-gal consumption for aging.
Collapse
Affiliation(s)
- Josiane Budni
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil.
| | - Arleide Braga Brandão
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Sabrina da Silva
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Michelle Lima Garcez
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Francielle Mina
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Tatiani Bellettini-Santos
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Gabriel Casagrande Zabot
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Eduarda Behenck Medeiros
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Giselli Scaini
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Emílio Luiz Streck
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, 88806-000, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence On Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
76
|
Zeng H, Qi Y, Zhang Z, Liu C, Peng W, Zhang Y. Nanomaterials toward the treatment of Alzheimer’s disease: Recent advances and future trends. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
77
|
Varshney V, Garabadu D. Ang(1-7) exerts Nrf2-mediated neuroprotection against amyloid beta-induced cognitive deficits in rodents. Mol Biol Rep 2021; 48:4319-4331. [PMID: 34075536 DOI: 10.1007/s11033-021-06447-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with cognitive deficits in an individual. Ang(1-7) exhibits neuroprotection against amyloid beta (Aβ)-induced mitochondrial dysfunction and neurotoxicity in experimental conditions. Further, Ang(1-7) also exhibits nrf2-mediated antioxidant activity in experimental conditions. However, its therapeutic role on nrf2-mediated mitochondrial function is yet to be established in the Aβ-induced neurotoxicity. The experimental dementia was induced in the male rats by intracerebroventricular administration of Aβ(1-42) on day-1 (D-1) of the experimental schedule of 14 days. Ang(1-7) was administered once daily from D-1 toD-14 to the Aβ-challenged rodents. Ang(1-7) attenuated Aβ-induced increase in escape latency and decrease in the time spent in the target quadrant during Morris water maze and percentage of spontaneous alteration behavior during Y-maze tests in the rats. Further, Ang(1-7) attenuated Aβ-induced cholinergic dysfunction in terms of decrease in the level of acetylcholine and activity of choline acetyltransferase, and increase in the activity of acetylcholinesterase, and increase in the level of Aβ in rat hippocampus, pre-frontal cortex and amygdala. Furthermore, Ang(1-7) reversed Aβ-induced decrease in the mitochondrial function, integrity and bioenergetics in all brain regions. Additionally, Ang(1-7) attenuated Aβ-induced increase in the extent of apoptosis and decrease in the level of heme oxygenase-1 in all selected brain regions. Trigonelline significantly abolished the therapeutic effectiveness of Ang(1-7) on Aβ-induced alterations in the behavioral, neurochemicals and molecular observations in the animals. Ang(1-7) may exhibit nrf2-mediated neuroprotection in these rodents. Hence, Ang(1-7) could be a potential therapeutic option in the pharmacotherapy of AD.
Collapse
Affiliation(s)
- Vibhav Varshney
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Debapriya Garabadu
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India.
| |
Collapse
|
78
|
Navazani P, Vaseghi S, Hashemi M, Shafaati MR, Nasehi M. Effects of Treadmill Exercise on the Expression Level of BAX, BAD, BCL-2, BCL-XL, TFAM, and PGC-1α in the Hippocampus of Thimerosal-Treated Rats. Neurotox Res 2021; 39:1274-1284. [PMID: 33939098 DOI: 10.1007/s12640-021-00370-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/10/2021] [Accepted: 04/26/2021] [Indexed: 01/11/2023]
Abstract
Thimerosal (THIM) induces neurotoxic changes including neuronal death and releases apoptosis inducing factors from mitochondria to cytosol. THIM alters the expression level of factors involved in apoptosis. On the other hand, the anti-apoptotic effects of exercise have been reported. In this study, we aimed to discover the effect of three protocols of treadmill exercise on the expression level of mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), BCL-2-associated death (BAD), BCL-2-associated X (BAX), BCL-XL, and BCL-2 (a pro-survival BCL-2 protein) in the hippocampus of control and THIM-exposed rats. Male Wistar rats were used in this research. Real-time PCR was applied to assess genes expression. The results showed that THIM increased the expression of pro-apoptotic factors (BAD and BAX), decreased the expression of anti-apoptotic factors (BCL-2 and BCL-XL), and decreased the expression of factors involved in mitochondrial biogenesis (TFAM and PGC-1α). Treadmill exercise protocols reversed the effect of THIM on all genes. In addition, treadmill exercise protocols decreased the expression of BAD and BAX, increased the expression of BCL-2, and increased the expression of TFAM and PGC-1α in control rats. In conclusion, THIM induced a pro-apoptotic effect and disturbed mitochondrial biogenesis and stability, whereas treadmill exercise reversed these effects.
Collapse
Affiliation(s)
- Pouria Navazani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Shafaati
- Department of Cellular and Molecular Biology, Faculty of Basic Sciences, Hamadan Branch, Islamic Azad University, Hamadan, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
79
|
Role of Thioredoxin-Interacting Protein in Diseases and Its Therapeutic Outlook. Int J Mol Sci 2021; 22:ijms22052754. [PMID: 33803178 PMCID: PMC7963165 DOI: 10.3390/ijms22052754] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Thioredoxin-interacting protein (TXNIP), widely known as thioredoxin-binding protein 2 (TBP2), is a major binding mediator in the thioredoxin (TXN) antioxidant system, which involves a reduction-oxidation (redox) signaling complex and is pivotal for the pathophysiology of some diseases. TXNIP increases reactive oxygen species production and oxidative stress and thereby contributes to apoptosis. Recent studies indicate an evolving role of TXNIP in the pathogenesis of complex diseases such as metabolic disorders, neurological disorders, and inflammatory illnesses. In addition, TXNIP has gained significant attention due to its wide range of functions in energy metabolism, insulin sensitivity, improved insulin secretion, and also in the regulation of glucose and tumor suppressor activities in various cancers. This review aims to highlight the roles of TXNIP in the field of diabetology, neurodegenerative diseases, and inflammation. TXNIP is found to be a promising novel therapeutic target in the current review, not only in the aforementioned diseases but also in prolonged microvascular and macrovascular diseases. Therefore, TXNIP inhibitors hold promise for preventing the growing incidence of complications in relevant diseases.
Collapse
|
80
|
Seydi E, Mehrpouya L, Sadeghi H, Rahimi S, Pourahmad J. Luteolin attenuates Fipronil-induced neurotoxicity through reduction of the ROS-mediated oxidative stress in rat brain mitochondria. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 173:104785. [PMID: 33771263 DOI: 10.1016/j.pestbp.2021.104785] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/03/2021] [Accepted: 01/13/2021] [Indexed: 06/12/2023]
Abstract
Luteolin (LUT) as a natural compound found in vegetables and fruits has various pharmacological effects. Fipronil (FPN), as a pesticide, has been considered for its effect on the antioxidant system and induction of oxidative stress. This study was designed to investigate the protective effects of LUT against the oxidative stress and mitochondrial toxicity induced by FPN on the rat brain. Several parameters such as mitochondrial reactive oxygen species (ROS) level, mitochondrial membrane potential (MMP) collapse, mitochondrial swelling, cytochrome c release, mitochondrial glutathione (GSH), lipid peroxidation (LPO) and Adenosine triphosphate (ATP) levels were assessed. Results indicated that the administration of LUT (25 μM) significantly improved oxidative stress and mitochondrial damages induced via FPN (6, 12 and 24 μM) in isolated mitochondria from the brain. These results show that LUT exerted protective effects against FPN-induced neurotoxicity in vitro through improving oxidative stress and mitochondrial damages.
Collapse
Affiliation(s)
- Enayatollah Seydi
- Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran; Research Center for Health, Safety and Environment, Alborz University of Medical Sciences, Karaj, Iran
| | - Leila Mehrpouya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadiseh Sadeghi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Rahimi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
81
|
Fišar Z, Musílek K, Benek O, Hroch L, Vinklářová L, Schmidt M, Hroudová J, Raboch J. Effects of novel 17β-hydroxysteroid dehydrogenase type 10 inhibitors on mitochondrial respiration. Toxicol Lett 2020; 339:12-19. [PMID: 33359020 DOI: 10.1016/j.toxlet.2020.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/02/2020] [Accepted: 12/19/2020] [Indexed: 12/29/2022]
Abstract
Mitochondrial enzymes are targets of newly synthesized drugs being tested for the treatment of neurodegenerative disorders, such as Alzheimer's disease (AD). The enzyme 17β-hydroxysteroid dehydrogenase type 10 (HSD10) is a multifunctional mitochondrial protein that is thought to play a role in the pathophysiology of AD and is one of the targets of new potential AD drugs. The in vitro effects of frentizole, riluzole, AG18051, and 42 novel modulators of HSD10 (potential AD drugs) on citrate synthase (CS) activity, monoamine oxidase (MAO) activity, complex I- or complex II-linked mitochondrial respiratory rate, and complex I activity were measured in isolated pig brain mitochondria. Based on their minimal inhibitory effects on the respiratory rate of mitochondria and CS and complex I activity, six novel compounds were selected for further testing. Assuming that inhibition of MAO-B could be a desirable effect of AD drugs, only AG18051 and one new compound met the criteria for MAO-B inhibition with minimal drug-induced effects on mitochondrial respiration. In conclusion, our in vitro screening of mitochondrial effect of novel potential AD drugs has enabled the selection of the most promising molecules for further testing that are relatively safe in terms of drug-induced mitochondrial toxicity.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 120 00, Prague 2, Czech Republic.
| | - Kamil Musílek
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic.
| | - Ondřej Benek
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Lukáš Hroch
- University Hospital in Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Lucie Vinklářová
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Monika Schmidt
- University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Jana Hroudová
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - Jiří Raboch
- Charles University and General University Hospital in Prague, First Faculty of Medicine, Department of Psychiatry, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| |
Collapse
|
82
|
Potential role of TrkB agonist in neuronal survival by promoting CREB/BDNF and PI3K/Akt signaling in vitro and in vivo model of 3-nitropropionic acid (3-NP)-induced neuronal death. Apoptosis 2020; 26:52-70. [PMID: 33226552 DOI: 10.1007/s10495-020-01645-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Striatal neurons depends on an afferent supply of brain-derived neurotrophic factor-(BDNF) that explicitly interacts with tropomyosin receptor kinase B (TrkB) receptor and performs sundry functions including synaptic plasticity, neuronal differentiation and growth. Therefore, we aimed to scrutinize an active molecule that functions identical to BDNF in activating TrkB receptor and it's downstream targets for restoring neuronal survival in Huntington disease (HD). Data from in vitro Neuro-2a cell line showed that treatment with 7,8-dihydroxyflavone (7,8-DHF), improved 3-nitropropionic acid (3-NP) induced neuronal death by stabilizing the loss of mitochondrial membrane potential and transiently increased the activity of cAMP-response element-binding protein (CREB) and BDNF via TrkB receptor activation. Consistent with in vitro findings, our in vivo results stated that treatment with 7,8-DHF at a dose of 10 mg/kg body weight ameliorated various behavior alterations caused by 3-NP intoxication. Further histopathological and electron microscopy evidences from striatal region of 3-NP mice brain treated with 7,8-DHF showed more improved neurons with intact mitochondria and less autophagic vacuoles. Protein expression analysis of both in vitro and in vivo study showed that 7,8-DHF promotes neuronal survival through upregulation and phosphorylation of phosphatidylinositol 3-kinase (PI3K) and Akt at serine-473/threonine-308). Akt phosphorylation additionally phosphorylates Bad at serine-136 and inhibits its translocation to mitochondria thereby promoting mitochondrial biogenesis, enhanced ATP production and inhibit apoptosis mediated neuronal death. These aforementioned findings help in strengthening our hypothesis and has come up with a novel neuroprotective mechanism of 7,8-DHF against 3-NP induced neuronal death.
Collapse
|
83
|
Galpayage Dona KNU, Du E, Wei J. An impedimetric assay for the identification of abnormal mitochondrial dynamics in living cells. Electrophoresis 2020; 42:163-170. [PMID: 33169407 DOI: 10.1002/elps.202000125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 01/16/2023]
Abstract
Mitochondrial dynamics (fission and fusion) plays an important role in cell functions. Disruption in mitochondrial dynamics has been associated with diseases such as neurobiological disorders and cardiovascular diseases. Analysis of mitochondrial fission/fusion has been mostly achieved through direct visualization of the fission/fusion events in live-cell imaging of fluorescently labeled mitochondria. In this study, we demonstrated a label-free, non-invasive Electrical Impedance Spectroscopy (EIS) approach to analyze mitochondrial dynamics in a genetically modified human neuroblastoma SH-SY5Y cell line with no huntingtin protein expression. Huntingtin protein has been shown to regulate mitochondria dynamics. We performed EIS studies on normal SH-SY5Y cells and two independent clones of huntingtin-null cells. The impedance data was used to determine the suspension conductivity and further cytoplasmic conductivity and relate to the abnormal mitochondrial dynamics. For instance, the cytoplasm conductivity value was increased by 11% from huntingtin-null cells to normal cells. Results of this study demonstrated that EIS is sensitive to characterize the abnormal mitochondrial dynamics that can be difficult to quantify by the conventional microscopic method.
Collapse
Affiliation(s)
| | - E Du
- Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, Florida, USA
| | - Jianning Wei
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| |
Collapse
|
84
|
Binvignat O, Olloquequi J. Excitotoxicity as a Target Against Neurodegenerative Processes. Curr Pharm Des 2020; 26:1251-1262. [PMID: 31931694 DOI: 10.2174/1381612826666200113162641] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022]
Abstract
The global burden of neurodegenerative diseases is alarmingly increasing in parallel to the aging of population. Although the molecular mechanisms leading to neurodegeneration are not completely understood, excitotoxicity, defined as the injury and death of neurons due to excessive or prolonged exposure to excitatory amino acids, has been shown to play a pivotal role. The increased release and/or decreased uptake of glutamate results in dysregulation of neuronal calcium homeostasis, leading to oxidative stress, mitochondrial dysfunctions, disturbances in protein turn-over and neuroinflammation. Despite the anti-excitotoxic drug memantine has shown modest beneficial effects in some patients with dementia, to date, there is no effective treatment capable of halting or curing neurodegenerative diseases such as Alzheimer's disease, Parkinson disease, Huntington's disease or amyotrophic lateral sclerosis. This has led to a growing body of research focusing on understanding the mechanisms associated with the excitotoxic insult and on uncovering potential therapeutic strategies targeting these mechanisms. In the present review, we examine the molecular mechanisms related to excitotoxic cell death. Moreover, we provide a comprehensive and updated state of the art of preclinical and clinical investigations targeting excitotoxic- related mechanisms in order to provide an effective treatment against neurodegeneration.
Collapse
Affiliation(s)
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Instituto de Ciencias Biomedicas, Facultad de Ciencias de la Salud, Universidad Autonoma de Chile, Talca, Chile
| |
Collapse
|
85
|
Goyal S, Chaturvedi RK. Mitochondrial Protein Import Dysfunction in Pathogenesis of Neurodegenerative Diseases. Mol Neurobiol 2020; 58:1418-1437. [PMID: 33180216 DOI: 10.1007/s12035-020-02200-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Mitochondria play an essential role in maintaining energy homeostasis and cellular survival. In the brain, higher ATP production is required by mature neurons for communication. Most of the mitochondrial proteins transcribe in the nucleus and import in mitochondria through different pathways of the mitochondrial protein import machinery. This machinery plays a crucial role in determining mitochondrial morphology and functions through mitochondrial biogenesis. Failure of this machinery and any alterations during mitochondrial biogenesis underlies neurodegeneration resulting in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD) etc. Current knowledge has revealed the different pathways of mitochondrial protein import machinery such as translocase of the outer mitochondrial membrane complex, the presequence pathway, carrier pathway, β-barrel pathway, and mitochondrial import and assembly machinery etc. In this review, we have discussed the recent studies regarding protein import machinery, beyond the well-known effects of increased oxidative stress and bioenergetics dysfunctions. We have elucidated in detail how these types of machinery help to import and locate the precursor proteins to their specific location inside the mitochondria and play a major role in mitochondrial biogenesis. We further discuss their involvement in mitochondrial dysfunctioning and the induction of toxic aggregates in neurodegenerative diseases like AD and PD. The review supports the importance of import machinery in neuronal functions and its association with toxic aggregated proteins in mitochondrial impairment, suggesting a critical role in fostering and maintaining neurodegeneration and therapeutic response.
Collapse
Affiliation(s)
- Shweta Goyal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
86
|
Scholefield M, Unwin RD, Cooper GJ. Shared perturbations in the metallome and metabolome of Alzheimer's, Parkinson's, Huntington's, and dementia with Lewy bodies: A systematic review. Ageing Res Rev 2020; 63:101152. [PMID: 32846222 DOI: 10.1016/j.arr.2020.101152] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
Despite differences in presentation, age-related dementing diseases such as Alzheimer's (AD), Parkinson's (PD), and Huntington's diseases (HD), and dementia with Lewy bodies (DLB) may share pathogenic processes. This review aims to systematically assemble and compare findings in various biochemical pathways across these four dementias. PubMed and Google Scholar were screened for articles reporting on brain and biofluid measurements of metals and/or metabolites in AD, PD, HD, or DLB. Articles were assessed using specific a priori-defined inclusion and exclusion criteria. Of 284 papers identified, 198 met criteria for inclusion. Although varying coverage levels of metals and metabolites across diseases and tissues made comparison of many analytes impossible, several common findings were identified: elevated glucose in both brain tissue and biofluids of AD, PD, and HD cases; increased iron and decreased copper in AD, PD and HD brain tissue; and decreased uric acid in biofluids of AD and PD cases. Other analytes were found to differ between diseases or were otherwise not covered across all conditions. These findings indicate that disturbances in glucose and purine pathways may be common to AD, PD, and HD. However, standardisation of methodologies and better coverage in some areas - notably of DLB - are necessary to validate and extend these findings.
Collapse
|
87
|
Effects of Novel Tacrine Derivatives on Mitochondrial Energy Metabolism and Monoamine Oxidase Activity-In Vitro Study. Mol Neurobiol 2020; 58:1102-1113. [PMID: 33089424 DOI: 10.1007/s12035-020-02172-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/14/2020] [Indexed: 11/27/2022]
Abstract
The trends of novel AD therapeutics are focused on multitarget-directed ligands (MTDLs), which combine cholinesterase inhibition with additional biological properties such as antioxidant properties to positively affect neuronal energy metabolism as well as mitochondrial function. We examined the in vitro effects of 10 novel MTDLs on the activities of mitochondrial enzymes (electron transport chain complexes and citrate synthase), mitochondrial respiration, and monoamine oxidase isoform (MAO-A and MAO-B) activity. The drug-induced effects of 7-MEOTA-adamantylamine heterodimers (K1011, K1013, K1018, K1020, and K1022) and tacrine/7-MEOTA/6-chlorotacrine-trolox heterodimers (K1046, K1053, K1056, K1060, and K1065) were measured in pig brain mitochondria. Most of the substances inhibited complex I- and complex II-linked respiration at high concentrations; K1046, K1053, K1056, and K1060 resulted in the least inhibition of mitochondrial respiration. Citrate synthase activity was not significantly inhibited by the tested substances; the least inhibition of complex I was observed for compounds K1060 and K1053, while both complex II/III and complex IV activity were markedly inhibited by K1011 and K1018. MAO-A was fully inhibited by K1018 and K1065, and MAO-B was fully inhibited by K1053 and K1065; the other tested drugs were partial inhibitors of both MAO-A and MAO-B. The tacrine/7-MEOTA/6-chlorotacrine-trolox heterodimers K1046, K1053, and K1060 seem to be the most suitable molecules for subsequent in vivo studies. These compounds had balanced inhibitory effects on mitochondrial respiration, with low complex I and complex II/III inhibition and full or partial inhibition of MAO-B activity.
Collapse
|
88
|
Indrieri A, Pizzarelli R, Franco B, De Leonibus E. Dopamine, Alpha-Synuclein, and Mitochondrial Dysfunctions in Parkinsonian Eyes. Front Neurosci 2020; 14:567129. [PMID: 33192254 PMCID: PMC7604532 DOI: 10.3389/fnins.2020.567129] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by motor dysfunctions including bradykinesia, tremor at rest and motor instability. These symptoms are associated with the progressive degeneration of dopaminergic neurons originating in the substantia nigra pars compacta and projecting to the corpus striatum, and by accumulation of cytoplasmic inclusions mainly consisting of aggregated alpha-synuclein, called Lewy bodies. PD is a complex, multifactorial disorder and its pathogenesis involves multiple pathways and mechanisms such as α-synuclein proteostasis, mitochondrial function, oxidative stress, calcium homeostasis, axonal transport, and neuroinflammation. Motor symptoms manifest when there is already an extensive dopamine denervation. There is therefore an urgent need for early biomarkers to apply disease-modifying therapeutic strategies. Visual defects and retinal abnormalities, including decreased visual acuity, abnormal spatial contrast sensitivity, color vision defects, or deficits in more complex visual tasks are present in the majority of PD patients. They are being considered for early diagnosis together with retinal imaging techniques are being considered as non-invasive biomarkers for PD. Dopaminergic cells can be found in the retina in a subpopulation of amacrine cells; however, the molecular mechanisms leading to visual deficits observed in PD patients are still largely unknown. This review provides a comprehensive analysis of the retinal abnormalities observed in PD patients and animal models and of the molecular mechanisms underlying neurodegeneration in parkinsonian eyes. We will review the role of α-synuclein aggregates in the retina pathology and/or in the onset of visual symptoms in PD suggesting that α-synuclein aggregates are harmful for the retina as well as for the brain. Moreover, we will summarize experimental evidence suggesting that the optic nerve pathology observed in PD resembles that seen in mitochondrial optic neuropathies highlighting the possible involvement of mitochondrial abnormalities in the development of PD visual defects. We finally propose that the eye may be considered as a complementary experimental model to identify possible novel disease’ pathways or to test novel therapeutic approaches for PD.
Collapse
Affiliation(s)
- Alessia Indrieri
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Institute for Genetic and Biomedical Research, National Research Council, Milan, Italy
| | - Rocco Pizzarelli
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Medical Genetics, Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Institute of Biochemistry and Cellular Biology, National Research Council, Rome, Italy
| |
Collapse
|
89
|
Qi Y, Chen L, Guo L, Shao C, Liu Y, Yang Y, He Z, Zhu H. An Activatable and Switchable Nanoaggregate Probe for Detecting H
2
S and Its Application in Mice Brains. Chem Asian J 2020; 15:3551-3557. [DOI: 10.1002/asia.202000964] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/17/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Ya‐Lin Qi
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Li‐Li Chen
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Long Guo
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Chen‐Wen Shao
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Ya‐Ni Liu
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Yu‐Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Zhen‐Xiang He
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Hai‐Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| |
Collapse
|
90
|
Mitochondrial biogenesis in organismal senescence and neurodegeneration. Mech Ageing Dev 2020; 191:111345. [DOI: 10.1016/j.mad.2020.111345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 12/19/2022]
|
91
|
Carrier M, Robert MÈ, González Ibáñez F, Desjardins M, Tremblay MÈ. Imaging the Neuroimmune Dynamics Across Space and Time. Front Neurosci 2020; 14:903. [PMID: 33071723 PMCID: PMC7539119 DOI: 10.3389/fnins.2020.00903] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
The immune system is essential for maintaining homeostasis, as well as promoting growth and healing throughout the brain and body. Considering that immune cells respond rapidly to changes in their microenvironment, they are very difficult to study without affecting their structure and function. The advancement of non-invasive imaging methods greatly contributed to elucidating the physiological roles performed by immune cells in the brain across stages of the lifespan and contexts of health and disease. For instance, techniques like two-photon in vivo microscopy were pivotal for studying microglial functional dynamics in the healthy brain. Through these observations, their interactions with neurons, astrocytes, blood vessels and synapses were uncovered. High-resolution electron microscopy with immunostaining and 3D-reconstruction, as well as super-resolution fluorescence microscopy, provided complementary insights by revealing microglial interventions at synapses (phagocytosis, trogocytosis, synaptic stripping, etc.). In addition, serial block-face scanning electron microscopy has provided the first 3D reconstruction of a microglial cell at nanoscale resolution. This review will discuss the technical toolbox that currently allows to study microglia and other immune cells in the brain, as well as introduce emerging methods that were developed and could be used to increase the spatial and temporal resolution of neuroimmune imaging. A special attention will also be placed on positron emission tomography and the development of selective functional radiotracers for microglia and peripheral macrophages, considering their strong potential for research translation between animals and humans, notably when paired with other imaging modalities such as magnetic resonance imaging.
Collapse
Affiliation(s)
- Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Robert
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Fernando González Ibáñez
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Michèle Desjardins
- Axe Oncologie, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Physics, Physical Engineering and Optics, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
92
|
Mignone P, Pio G, D'Elia D, Ceci M. Exploiting transfer learning for the reconstruction of the human gene regulatory network. Bioinformatics 2020; 36:1553-1561. [PMID: 31608946 DOI: 10.1093/bioinformatics/btz781] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/13/2019] [Accepted: 10/09/2019] [Indexed: 01/26/2023] Open
Abstract
MOTIVATION The reconstruction of gene regulatory networks (GRNs) from gene expression data has received increasing attention in recent years, due to its usefulness in the understanding of regulatory mechanisms involved in human diseases. Most of the existing methods reconstruct the network through machine learning approaches, by analyzing known examples of interactions. However, (i) they often produce poor results when the amount of labeled examples is limited, or when no negative example is available and (ii) they are not able to exploit information extracted from GRNs of other (better studied) related organisms, when this information is available. RESULTS In this paper, we propose a novel machine learning method that overcomes these limitations, by exploiting the knowledge about the GRN of a source organism for the reconstruction of the GRN of the target organism, by means of a novel transfer learning technique. Moreover, the proposed method is natively able to work in the positive-unlabeled setting, where no negative example is available, by fruitfully exploiting a (possibly large) set of unlabeled examples. In our experiments, we reconstructed the human GRN, by exploiting the knowledge of the GRN of Mus musculus. Results showed that the proposed method outperforms state-of-the-art approaches and identifies previously unknown functional relationships among the analyzed genes. AVAILABILITY AND IMPLEMENTATION http://www.di.uniba.it/∼mignone/systems/biosfer/index.html. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Paolo Mignone
- Department of Computer Science, University of Bari Aldo Moro, Bari 70125, Italy.,National Interuniversity Consortium for Informatics (CINI), Roma 00185, Italy
| | - Gianvito Pio
- Department of Computer Science, University of Bari Aldo Moro, Bari 70125, Italy.,National Interuniversity Consortium for Informatics (CINI), Roma 00185, Italy
| | - Domenica D'Elia
- Institute for Biomedical Technologies, CNR, Institute for Biomedical Technologies, Bari 70126, Italy
| | - Michelangelo Ceci
- Department of Computer Science, University of Bari Aldo Moro, Bari 70125, Italy.,National Interuniversity Consortium for Informatics (CINI), Roma 00185, Italy.,Department of Knowledge Technologies, Jožef Stefan Institute, Ljubljana 1000, Slovenia
| |
Collapse
|
93
|
Zhang J, Dong Y, Lining Huang, Xu X, Liang F, Soriano SG, Zhang Y, Xie Z. Interaction of Tau, IL-6 and mitochondria on synapse and cognition following sevoflurane anesthesia in young mice. Brain Behav Immun Health 2020; 8:100133. [PMID: 34589883 PMCID: PMC8474534 DOI: 10.1016/j.bbih.2020.100133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023] Open
Abstract
Tau phosphorylation is associated with cognitive impairment in young mice. However, the underlying mechanism and targeted interventions remain mostly unknown. We set out to determine the potential interactions of Tau, interleukin 6 (IL-6) and mitochondria following treatment of anesthetic sevoflurane and to assess their influences on synapse number and cognition in young mice. Sevoflurane (3% for 2 h) was given to wild-type, Tau knockout, IL-6 knockout, and cyclophilin D (CypD) knockout mice on postnatal (P) day 6, 7 and 8. We measured amounts of phosphorylated Tau, IL-6, reactive oxygen species (ROS), mitochondrial membrane potential (MMP), ATP, postsynaptic density 95 (PSD-95), synaptophysin, N-cadherin, synapse number, and cognitive function in the mice, employing Western blot, electron microscope and Morris water maze among others. Here we showed that sevoflurane increased Tau phosphorylation and caused IL-6 elevation, mitochondrial dysfunction, synaptic loss and cognitive impairment in young wild-type, but not Tau knockout, mice. In young IL-6 knockout mice, sevoflurane increased Tau phosphorylation but did not cause mitochondrial dysfunction, synaptic loss or cognitive impairment. Finally, sevoflurane increased Tau phosphorylation and IL-6 amount, but did not induce synaptic loss and cognitive impairment, in young CypD knockout mice or WT mice pretreated with idebenone, an analog of co-enzyme Q10. In conclusion, sevoflurane increased Tau phosphorylation, which caused IL-6 elevation, leading to mitochondrial dysfunction in young mice. Such interactions caused synaptic loss and cognitive impairment in the mice. Idebenone mitigated sevoflurane-induced cognitive impairment in young mice. These studies would promote more research to study Tau in young mice. Research in context.Evidence before this studyTau, a microtubule-associated protein that is predominantly expressed inside neurons, is associated with microtubule assembly and function. Tau phosphorylation, aggregation and spread all serve as the pathogenesis of age-dependent neurodegeneration in the old brain, as well as the neuropathogenesis of Alzheimer’s disease. However, the effects of Tau on the cellular changes and the function of the young brain are undetermined. Our previous studies showed that anesthetic sevoflurane induced Tau phosphorylation, IL-6 elevation, mitochondrial dysfunction and synaptic loss in brain tissues of neonatal mice, as well as cognitive impairment in the mice. However, the potential interactions of the Tau phosphorylation, IL-6 elevation and mitochondrial dysfunction and the influences of these interactions on synapse number and cognitive function in neonatal mice remains largely unknown.
Added value of studyEmploying sevoflurane as a clinically relevant tool, and using the approaches including wild-type, Tau, IL-6, and CypD knockout neonatal mice, the present studies showed that Tau phosphorylation caused IL-6 elevation, which induced mitochondrial dysfunction, leading to synaptic loss and cognitive impairment in the neonatal mice. Idebenone, a synthetic analog of coenzyme Q10, mitigated the sevoflurane-induced cognitive impairment in the neonatal mice.
Implications of all the available evidenceThese findings demonstrated the role of Tau phosphorylation in cognitive impairment in neonatal mice, revealed the effects of the interactions of Tau phosphorylation, IL-6 elevation and mitochondrial dysfunction on the synapse number and cognitive function in the mice, and identified potential targeted intervention of the cognitive impairment in the neonatal mice.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA
| | - Lining Huang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA.,Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Xiaoming Xu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA.,Department of Forensic Clinical Medicine, School of Forensic Medicine, China Medical University, Shenyang, PR China
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA
| | - Sulpicio G Soriano
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA
| |
Collapse
|
94
|
Hosford PS, Ninkina N, Buchman VL, Smith JC, Marina N, SheikhBahaei S. Synuclein Deficiency Results in Age-Related Respiratory and Cardiovascular Dysfunctions in Mice. Brain Sci 2020; 10:brainsci10090583. [PMID: 32846874 PMCID: PMC7563345 DOI: 10.3390/brainsci10090583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 01/16/2023] Open
Abstract
Synuclein (α, β, and γ) proteins are highly expressed in presynaptic terminals, and significant data exist supporting their role in regulating neurotransmitter release. Targeting the gene encoding α-synuclein is the basis of many animal models of Parkinson's disease (PD). However, the physiological role of this family of proteins in not well understood and could be especially relevant as interfering with accumulation of α-synuclein level has therapeutic potential in limiting PD progression. The long-term effects of their removal are unknown and given the complex pathophysiology of PD, could exacerbate other clinical features of the disease, for example dysautonomia. In the present study, we sought to characterize the autonomic phenotypes of mice lacking all synucleins (α, β, and γ; αβγ-/-) in order to better understand the role of synuclein-family proteins in autonomic function. We probed respiratory and cardiovascular reflexes in conscious and anesthetized, young (4 months) and aged (18-20 months) αβγ-/- male mice. Aged mice displayed impaired respiratory responses to both hypoxia and hypercapnia when breathing activities were recorded in conscious animals using whole-body plethysmography. These animals were also found to be hypertensive from conscious blood pressure recordings, to have reduced pressor baroreflex gain under anesthesia, and showed reduced termination of both pressor and depressor reflexes. The present data demonstrate the importance of synuclein in the normal function of respiratory and cardiovascular reflexes during aging.
Collapse
Affiliation(s)
- Patrick S. Hosford
- Department of Neuroscience Physiology and Pharmacology, Center for Cardiovascular and Metabolic Neuroscience, University College London (UCL), London WC1E 6BT, UK; (P.S.H.); (N.M.)
| | - Natalia Ninkina
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK; (N.N.); (V.L.B.)
- Institute of Physiologically Active Compounds, Russian Academy of Sciences (IPAC RAS), 1 Severniy proezd, 142432 Chernogolovka, Moscow Region, Russia
| | - Vladimir L. Buchman
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK; (N.N.); (V.L.B.)
- Institute of Physiologically Active Compounds, Russian Academy of Sciences (IPAC RAS), 1 Severniy proezd, 142432 Chernogolovka, Moscow Region, Russia
| | - Jeffrey C. Smith
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD 20892, USA;
| | - Nephtali Marina
- Department of Neuroscience Physiology and Pharmacology, Center for Cardiovascular and Metabolic Neuroscience, University College London (UCL), London WC1E 6BT, UK; (P.S.H.); (N.M.)
| | - Shahriar SheikhBahaei
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD 20892, USA;
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD 20892, USA
- Correspondence: ; Tel.: +1-301-496-4960; Fax: +1-301-496-1339
| |
Collapse
|
95
|
Yang M, Tan H, Zhang K, Lian N, Yu Y, Yu Y. Protective effects of Coenzyme Q10 against sevoflurane-induced cognitive impairment through regulating apolipoprotein E and phosphorylated Tau expression in young mice. Int J Dev Neurosci 2020; 80:418-428. [PMID: 32473608 DOI: 10.1002/jdn.10041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 01/24/2023] Open
Abstract
Children with multiple exposures to anesthesia and surgery may be more likely to develop the learning disability. Coenzyme Q10 (CoQ10) was reported to reduce the multiple sevoflurane treatment-induced cognitive deficiency in 6-day-old young mice. However, its specific mechanisms have not yet been found. This research aimed to reveal the role of ApoE in the pathogenesis of cognitive deficiency caused by sevoflurane anesthesia and the protective mechanism of CoQ10 in a multiple sevoflurane treatment model of young mice. The mice were randomly divided into four groups: Control + corn oil, Sevoflurane + corn oil, Control + CoQ10, and Sevoflurane + CoQ10. Sevoflurane group mice were anesthetized with 3% sevoflurane and 60% oxygen 2 hr a day for 3 days, while control group mice received only 60% oxygen. Mice received an intraperitoneal injection of 50 mg/kg CoQ10 or the same volume of corn oil 30 min before the inhalation of oxygen or sevoflurane for 3 days. Mice received sevoflurane anesthesia or control treatment from the 6th to 8th day after birth. The cortex and hippocampus were harvested on the 8th day. The ATP, MMP, ApoE mRNA, total ApoE, ApoE fragments, Aβ1-40, Aβ1-42, Tau5, AT8, and PHF levels were detected. The Morris water maze (MWM) tests were performed from P30 to p36 after anesthesia or control treatment. The results indicated that the injection of CoQ10 ahead of sevoflurane treatment could reverse the anesthesia-induced energy deficiency, mitochondrial dysfunction, ApoE, and its fragments expression, Aβ1-42 generation, Tau phosphorylation, and cognitive impairment in young mice. These data reveal that the ApoE and its fragments enhancement may play an important role in the pathogenesis of cognitive deficiency caused by sevoflurane anesthesia. CoQ10 could reduce ApoE expression by improving energy replenishment and mitochondrial functions, thereby alleviating sevoflurane-induced brain damage and cognitive impairment.
Collapse
Affiliation(s)
- Man Yang
- Department of Anesthesia, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Hong Tan
- Department of Anesthesia, Huashan Hospital, Fudan University, Shanghai, China
| | - Kai Zhang
- Department of Anesthesia, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Naqi Lian
- Department of Anesthesia, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yang Yu
- Department of Anesthesia, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yonghao Yu
- Department of Anesthesia, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| |
Collapse
|
96
|
Kang TC. Nuclear Factor-Erythroid 2-Related Factor 2 (Nrf2) and Mitochondrial Dynamics/Mitophagy in Neurological Diseases. Antioxidants (Basel) 2020; 9:antiox9070617. [PMID: 32679689 PMCID: PMC7402121 DOI: 10.3390/antiox9070617] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/01/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria play an essential role in bioenergetics and respiratory functions for cell viability through numerous biochemical processes. To maintain mitochondria quality control and homeostasis, mitochondrial morphologies change rapidly in response to external insults and changes in metabolic status through fusion and fission (so called mitochondrial dynamics). Furthermore, damaged mitochondria are removed via a selective autophagosomal process, referred to as mitophagy. Although mitochondria are one of the sources of reactive oxygen species (ROS), they are themselves vulnerable to oxidative stress. Thus, endogenous antioxidant defense systems play an important role in cell survival under physiological and pathological conditions. Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a redox-sensitive transcription factor that maintains redox homeostasis by regulating antioxidant-response element (ARE)-dependent transcription and the expression of antioxidant defense enzymes. Although the Nrf2 system is positively associated with mitochondrial biogenesis and mitochondrial quality control, the relationship between Nrf2 signaling and mitochondrial dynamics/mitophagy has not been sufficiently addressed in the literature. This review article describes recent clinical and experimental observations on the relationship between Nrf2 and mitochondrial dynamics/mitophagy in various neurological diseases.
Collapse
Affiliation(s)
- Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; ; Tel.: +82-33-248-2524; Fax: +82-33-248-2525
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
97
|
Di Liberto D, D’Anneo A, Carlisi D, Emanuele S, De Blasio A, Calvaruso G, Giuliano M, Lauricella M. Brain Opioid Activity and Oxidative Injury: Different Molecular Scenarios Connecting Celiac Disease and Autistic Spectrum Disorder. Brain Sci 2020; 10:E437. [PMID: 32659996 PMCID: PMC7407635 DOI: 10.3390/brainsci10070437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
Celiac Disease (CD) is an immune-mediated disease triggered by the ingestion of wheat gliadin and related prolamins from other cereals, such as barley and rye. Immunity against these cereal-derived proteins is mediated by pro-inflammatory cytokines produced by both innate and adaptive system response in individuals unable to adequately digest them. Peptides generated in this condition are absorbed across the gut barrier, which in these patients is characterized by the deregulation of its permeability. Here, we discuss a possible correlation between CD and Autistic Spectrum Disorder (ASD) pathogenesis. ASD can be induced by an excessive and inappropriate brain opioid activity during the neonatal period. Cereal-derived peptides produced in celiac patients cross the blood-brain barrier and bind to endogenous opioid receptors interfering with neurotransmission and generating deleterious effects on brain maturation, learning and social relations. Moreover, an increase in oxidative stress and a decrease in the antioxidant capacity, as well as an extended mitochondrial impairment in the brain, could represent a possible connection between ASD and CD. Therefore, we critically discuss the proposed relationship between ASD and CD and the possible usefulness of a gluten-free diet in ASD patients.
Collapse
Affiliation(s)
- Diana Di Liberto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy;
| | - Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy; (A.D.B.); (G.C.); (M.G.)
| | - Daniela Carlisi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy; (D.C.); (S.E.)
| | - Sonia Emanuele
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy; (D.C.); (S.E.)
| | - Anna De Blasio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy; (A.D.B.); (G.C.); (M.G.)
| | - Giuseppe Calvaruso
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy; (A.D.B.); (G.C.); (M.G.)
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy; (A.D.B.); (G.C.); (M.G.)
| | - Marianna Lauricella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Biochemistry, University of Palermo, 90127 Palermo, Italy; (D.C.); (S.E.)
| |
Collapse
|
98
|
Yang X, Zhang D, Li J, Ji W, Yang N, Gu S, Wu Q, Jiang Q, Shi P, Li L. A mitochondrion-targeting Mn(ii)-terpyridine complex for two-photon photodynamic therapy. Chem Commun (Camb) 2020; 56:9032-9035. [PMID: 32643722 DOI: 10.1039/d0cc02051f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We developed a mitochondrion-targeting MnII-terpyridine complex (MTP) for two-photon photodynamic therapy. MTP was subjected to two-photon excitation in the NIR region to generate 1O2 and hence produce a PDT effect. This use of MTP overcame the drawbacks of traditional PDT agents. The MnII center of MTP apparently catalyzed the H2O2-mediated production of ROS and oxygen, which subsequently promoted the 1O2 generation, furthering the PDT effect. Additionally, the mitochondrion targeting of MTP provided a good spatial condition for the generation of 1O2, which further promoted the PDT effect.
Collapse
Affiliation(s)
- Xinda Yang
- School of Chemical Engineering, Jiangsu Key Laboratory of Function Control Technology for Advanced Materials, Jiangsu Ocean University, Lianyungang 222005, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Yang L, Youngblood H, Wu C, Zhang Q. Mitochondria as a target for neuroprotection: role of methylene blue and photobiomodulation. Transl Neurodegener 2020; 9:19. [PMID: 32475349 PMCID: PMC7262767 DOI: 10.1186/s40035-020-00197-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction plays a central role in the formation of neuroinflammation and oxidative stress, which are important factors contributing to the development of brain disease. Ample evidence suggests mitochondria are a promising target for neuroprotection. Recently, methods targeting mitochondria have been considered as potential approaches for treatment of brain disease through the inhibition of inflammation and oxidative injury. This review will discuss two widely studied approaches for the improvement of brain mitochondrial respiration, methylene blue (MB) and photobiomodulation (PBM). MB is a widely studied drug with potential beneficial effects in animal models of brain disease, as well as limited human studies. Similarly, PBM is a non-invasive treatment that promotes energy production and reduces both oxidative stress and inflammation, and has garnered increasing attention in recent years. MB and PBM have similar beneficial effects on mitochondrial function, oxidative damage, inflammation, and subsequent behavioral symptoms. However, the mechanisms underlying the energy enhancing, antioxidant, and anti-inflammatory effects of MB and PBM differ. This review will focus on mitochondrial dysfunction in several different brain diseases and the pathological improvements following MB and PBM treatment.
Collapse
Affiliation(s)
- Luodan Yang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Hannah Youngblood
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Chongyun Wu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
100
|
Testosterone enhances mitochondrial complex V function in the substantia nigra of aged male rats. Aging (Albany NY) 2020; 12:10398-10414. [PMID: 32445551 PMCID: PMC7346067 DOI: 10.18632/aging.103265] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/20/2020] [Indexed: 01/21/2023]
Abstract
Deficits in coordinated motor behavior and mitochondrial complex V activity have been observed in aged males. Testosterone supplementation can improve coordinated motor behavior in aged males. We investigated the effects of testosterone supplementation on mitochondrial complex V function in the substantia nigra (a brain region that regulates motor activity) in aged male rats. These rats exhibited diminished ATP levels, attenuated mitochondrial complex V activity, and reduced expression of 3 of the 17 mitochondrial complex V subunits (ATP6, ATP8 and ATP5C1) in the substantia nigra. Testosterone supplementation increased ATP levels, mitochondrial complex V activity, and ATP6, ATP8 and ATP5C1 expression in the substantia nigra of the rats. Conversely, orchiectomy reduced mitochondrial complex V activity, downregulated ATP6 and ATP8 expression, and upregulated ATP5C1, ATP5I and ATP5L expression in the substantia nigra. Testosterone replacement reversed those effects. Thus, testosterone enhanced mitochondrial complex V function in the substantia nigra of aged male rats by upregulating ATP6 and ATP8. As potential testosterone targets, these two subunits may to some degree maintain nigrostriatal dopaminergic function in aged males.
Collapse
|