51
|
Soares P, Silva C, Chavarria D, Silva FSG, Oliveira PJ, Borges F. Drug discovery and amyotrophic lateral sclerosis: Emerging challenges and therapeutic opportunities. Ageing Res Rev 2023; 83:101790. [PMID: 36402404 DOI: 10.1016/j.arr.2022.101790] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the degeneration of upper and lower motor neurons (MNs) leading to paralysis and, ultimately, death by respiratory failure 3-5 years after diagnosis. Edaravone and Riluzole, the only drugs currently approved for ALS treatment, only provide mild symptomatic relief to patients. Extraordinary progress in understanding the biology of ALS provided new grounds for drug discovery. Over the last two decades, mitochondria and oxidative stress (OS), iron metabolism and ferroptosis, and the major regulators of hypoxia and inflammation - HIF and NF-κB - emerged as promising targets for ALS therapeutic intervention. In this review, we focused our attention on these targets to outline and discuss current advances in ALS drug development. Based on the challenges and the roadblocks, we believe that the rational design of multi-target ligands able to modulate the complex network of events behind the disease can provide effective therapies in a foreseeable future.
Collapse
Affiliation(s)
- Pedro Soares
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| | - Catia Silva
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Filomena S G Silva
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paulo J Oliveira
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; IIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
52
|
Mikhailova MM, Surin AM, Sobolevsky A, Yelshanskaya M, Bolshakov AP. Boris Izrailevich Khodorov: Scientist and Teacher. NEUROCHEM J+ 2022. [DOI: 10.1134/s1819712422040171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
53
|
Saavedra J, Nascimento M, Liz MA, Cardoso I. Key brain cell interactions and contributions to the pathogenesis of Alzheimer's disease. Front Cell Dev Biol 2022; 10:1036123. [PMID: 36523504 PMCID: PMC9745159 DOI: 10.3389/fcell.2022.1036123] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/14/2022] [Indexed: 06/22/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide, with the two major hallmarks being the deposition of extracellular β-amyloid (Aβ) plaques and of intracellular neurofibrillary tangles (NFTs). Additionally, early pathological events such as cerebrovascular alterations, a compromised blood-brain barrier (BBB) integrity, neuroinflammation and synaptic dysfunction, culminate in neuron loss and cognitive deficits. AD symptoms reflect a loss of neuronal circuit integrity in the brain; however, neurons do not operate in isolation. An exclusively neurocentric approach is insufficient to understand this disease, and the contribution of other brain cells including astrocytes, microglia, and vascular cells must be integrated in the context. The delicate balance of interactions between these cells, required for healthy brain function, is disrupted during disease. To design successful therapies, it is critical to understand the complex brain cellular connections in AD and the temporal sequence of their disturbance. In this review, we discuss the interactions between different brain cells, from physiological conditions to their pathological reactions in AD, and how this basic knowledge can be crucial for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Joana Saavedra
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Mariana Nascimento
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Márcia A. Liz
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Isabel Cardoso
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
54
|
Germanova E, Khmil N, Pavlik L, Mikheeva I, Mironova G, Lukyanova L. The Role of Mitochondrial Enzymes, Succinate-Coupled Signaling Pathways and Mitochondrial Ultrastructure in the Formation of Urgent Adaptation to Acute Hypoxia in the Myocardium. Int J Mol Sci 2022; 23:ijms232214248. [PMID: 36430733 PMCID: PMC9696391 DOI: 10.3390/ijms232214248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
The effect of a single one-hour exposure to three modes of hypobaric hypoxia (HBH) differed in the content of O2 in inhaled air (FiO2-14%, 10%, 8%) in the development of mitochondrial-dependent adaptive processes in the myocardium was studied in vivo. The following parameters have been examined: (a) an urgent reaction of catalytic subunits of mitochondrial enzymes (NDUFV2, SDHA, Cyt b, COX2, ATP5A) in the myocardium as an indicator of the state of the respiratory chain electron transport function; (b) an urgent activation of signaling pathways dependent on GPR91, HIF-1α and VEGF, allowing us to assess their role in the formation of urgent mechanisms of adaptation to hypoxia in the myocardium; (c) changes in the ultrastructure of three subpopulations of myocardial mitochondria under these conditions. The studies were conducted on two rat phenotypes: rats with low resistance (LR) and high resistance (HR) to hypoxia. The adaptive and compensatory role of the mitochondrial complex II (MC II) in maintaining the electron transport and energy function of the myocardium in a wide range of reduced O2 concentrations in the initial period of hypoxic exposure has been established. The features of urgent reciprocal regulatory interaction of NAD- and FAD-dependent oxidation pathways in myocardial mitochondria under these conditions have been revealed. The data indicating the participation of GPR91, HIF-1a and VEGF in this process have been obtained. The ultrastructure of the mitochondrial subpopulations in the myocardium of LR and HR rats differed in normoxic conditions and reacted differently to hypoxia of varying severity. The parameters studied together are highly informative indicators of the quality of cardiac activity and metabolic biomarkers of urgent adaptation in various hypoxic conditions.
Collapse
Affiliation(s)
- Elita Germanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
| | - Natalya Khmil
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Lyubov Pavlik
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Irina Mikheeva
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
| | - Galina Mironova
- Institute of Theoretical and Experimental Biophysics RAS, 3 Institutskaya Str., Pushchino 142290, Moscow Region, Russia
- Correspondence: (G.M.); (L.L.)
| | - Ludmila Lukyanova
- Institute of General Pathology and Pathophysiology, 8 Baltijskaya Str., Moscow 125315, Russia
- Correspondence: (G.M.); (L.L.)
| |
Collapse
|
55
|
Zgodova A, Pavlova S, Nekrasova A, Boyarkin D, Pinelis V, Surin A, Bakaeva Z. Isoliquiritigenin Protects Neuronal Cells against Glutamate Excitotoxicity. MEMBRANES 2022; 12:1052. [PMID: 36363608 PMCID: PMC9693036 DOI: 10.3390/membranes12111052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
It is considered that glutamate excitotoxicity may be a major factor in the pathological death of neurons and mediate the development of neurodegenerative diseases in humans. Here, we show that isoliquiritigenin (ILG) at a concentration of 0.5-5 µM protects primary neuroglial cell culture from glutamate-induced death (glutamate 100 µM). ILG (1 µM) prevented a sharp increase in [Ca2+]i and a decrease in mitochondrial potential (ΔΨm). With the background action of ILG (1-5 µM), there was an increase in oxygen consumption rate (OCR) in response to glutamate, as well as in reserve respiration. The neuroprotective effect of ILG (5 µM) was accompanied by an increase in non-mitochondrial respiration. The results show that ILG can protect cortical neurons from death by preventing the development of calcium deregulation and limiting mitochondrial dysfunction caused by a high dose of glutamate. We hypothesize that ILG will be useful in drug development for the prevention or treatment of neurodegenerative diseases accompanied by glutamate excitotoxicity.
Collapse
Affiliation(s)
- Arina Zgodova
- Laboratory of Neurobiology and Fundamentals of Brain Development, National Medical Research Center of Children’s Health, 119991 Moscow, Russia
- Department of Psychiatry and Psychosomatics, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Svetlana Pavlova
- Department of Pharmacology, Clinical Pharmacology and Biochemistry, Chuvash State University named after I.N. Ulyanov, 428015 Cheboksary, Russia
| | - Anastasia Nekrasova
- Laboratory of Neurobiology and Fundamentals of Brain Development, National Medical Research Center of Children’s Health, 119991 Moscow, Russia
| | - Dmitriy Boyarkin
- Laboratory of Neurobiology and Fundamentals of Brain Development, National Medical Research Center of Children’s Health, 119991 Moscow, Russia
| | - Vsevolod Pinelis
- Laboratory of Neurobiology and Fundamentals of Brain Development, National Medical Research Center of Children’s Health, 119991 Moscow, Russia
| | - Alexander Surin
- Laboratory of Neurobiology and Fundamentals of Brain Development, National Medical Research Center of Children’s Health, 119991 Moscow, Russia
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Zanda Bakaeva
- Laboratory of Neurobiology and Fundamentals of Brain Development, National Medical Research Center of Children’s Health, 119991 Moscow, Russia
- Department of Medicine, General Biology and Physiology, Kalmyk State University named after B.B. Gorodovikov, 358000 Elista, Russia
| |
Collapse
|
56
|
Pinelis V, Krasilnikova I, Bakaeva Z, Surin A, Boyarkin D, Fisenko A, Krasilnikova O, Pomytkin I. Insulin Diminishes Superoxide Increase in Cytosol and Mitochondria of Cultured Cortical Neurons Treated with Toxic Glutamate. Int J Mol Sci 2022; 23:ijms232012593. [PMID: 36293449 PMCID: PMC9604026 DOI: 10.3390/ijms232012593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Glutamate excitotoxicity is involved in the pathogenesis of many disorders, including stroke, traumatic brain injury, and Alzheimer’s disease, for which central insulin resistance is a comorbid condition. Neurotoxicity of glutamate (Glu) is primarily associated with hyperactivation of the ionotropic N-methyl-D-aspartate receptors (NMDARs), causing a sustained increase in intracellular free calcium concentration ([Ca2+]i) and synchronous mitochondrial depolarization and an increase in intracellular superoxide anion radical (O2–•) production. Recently, we found that insulin protects neurons against excitotoxicity by decreasing the delayed calcium deregulation (DCD). However, the role of insulin in O2–• production in excitotoxicity still needs to be clarified. The present study aims to investigate insulin’s effects on glutamate-evoked O2–• generation and DCD using the fluorescent indicators dihydroethidium, MitoSOX Red, and Fura-FF in cortical neurons. We found a linear correlation between [Ca2+]i and [O2–•] in primary cultures of the rat neuron exposed to Glu, with insulin significantly reducing the production of intracellular and mitochondrial O2–• in the primary cultures of the rat neuron. MK 801, an inhibitor of NMDAR-gated Ca2+ influx, completely abrogated the glutamate effects in both the presence and absence of insulin. In experiments in sister cultures, insulin diminished neuronal death and O2 consumption rate (OCR).
Collapse
Affiliation(s)
- Vsevolod Pinelis
- Laboratory of Neurobiology, National Medical Research Center of Children’s Health, Russian Ministry of Health, Lomonosov Avenue 2, Bldg 1, 119991 Moscow, Russia
- Correspondence: (V.P.); (I.P.)
| | - Irina Krasilnikova
- Laboratory of Neurobiology, National Medical Research Center of Children’s Health, Russian Ministry of Health, Lomonosov Avenue 2, Bldg 1, 119991 Moscow, Russia
| | - Zanda Bakaeva
- Laboratory of Neurobiology, National Medical Research Center of Children’s Health, Russian Ministry of Health, Lomonosov Avenue 2, Bldg 1, 119991 Moscow, Russia
- Department of General Biology and Physiology, Kalmyk State University Named after B.B. Gorodovikov, St. Pushkin, 11, 358000 Elista, Russia
| | - Alexander Surin
- Laboratory of Neurobiology, National Medical Research Center of Children’s Health, Russian Ministry of Health, Lomonosov Avenue 2, Bldg 1, 119991 Moscow, Russia
- Laboratory of Pathology of Ion Transport and Intracellular Signaling, Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia
| | - Dmitrii Boyarkin
- Laboratory of Neurobiology, National Medical Research Center of Children’s Health, Russian Ministry of Health, Lomonosov Avenue 2, Bldg 1, 119991 Moscow, Russia
| | - Andrei Fisenko
- Laboratory of Neurobiology, National Medical Research Center of Children’s Health, Russian Ministry of Health, Lomonosov Avenue 2, Bldg 1, 119991 Moscow, Russia
| | - Olga Krasilnikova
- Department of Regenerative Medicine, National Medical Research Radiological Center, 4 Koroleva St., 249036 Obninsk, Russia
| | - Igor Pomytkin
- Institute of Pharmacy, The First Sechenov Moscow State Medical University under Ministry of Health of the Russian Federation, St. Trubetskaya, 8, Bldg 2, 119991 Moscow, Russia
- Correspondence: (V.P.); (I.P.)
| |
Collapse
|
57
|
Astaxanthin Protection against Neuronal Excitotoxicity via Glutamate Receptor Inhibition and Improvement of Mitochondrial Function. Mar Drugs 2022; 20:md20100645. [PMID: 36286468 PMCID: PMC9605357 DOI: 10.3390/md20100645] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Excitotoxicity is known to associate with neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, Amyotrophic lateral sclerosis and Huntington’s disease, as well as aging, stroke, trauma, ischemia and epilepsy. Excessive release of glutamate, overactivation of glutamate receptors, calcium overload, mitochondrial dysfunction and excessive reactive oxygen species (ROS) formation are a few of the suggested key mechanisms. Astaxanthin (AST), a carotenoid, is known to act as an antioxidant and protect neurons from excitotoxic injuries. However, the exact molecular mechanism of AST neuroprotection is not clear. Thus, in this study, we investigated the role of AST in neuroprotection in excitotoxicity. We utilized primary cortical neuronal culture and live cell fluorescence imaging for the study. Our results suggest that AST prevents neuronal death, reduces ROS formation and decreases the abnormal mitochondrial membrane depolarization induced by excitotoxic glutamate insult. Additionally, AST modulates intracellular calcium levels by inhibiting peak and irreversible secondary sustained calcium levels in neurons. Furthermore, AST regulates the ionotropic glutamate subtype receptors NMDA, AMPA, KA and mitochondrial calcium. Moreover, AST decreases NMDA and AMPA receptor protein expression levels, while KA remains unaffected. Overall, our results indicate that AST protects neurons from excitotoxic neuronal injury by regulating ionotropic glutamate receptors, cytosolic secondary calcium rise and mitochondrial calcium buffering. Hence, AST could be a promising therapeutic agent against excitotoxic insults in neurodegenerative diseases.
Collapse
|
58
|
Aoiadni N, Jdidi H, Feki AE, Fetoui H, Koubaa FG. Mitochondrial bioenergetics and redox dysfunction in nephrotoxicity induced by pyrethroid permethrin are ameliorated by flavonoid-rich fraction. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:63973-63987. [PMID: 35469380 DOI: 10.1007/s11356-022-20350-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/15/2022] [Indexed: 06/14/2023]
Abstract
The present study was designed to evaluate in vitro and in vivo the potential anti-inflammatory and nephroprotective potential of ethyl acetate fraction extracted from Fumaria officinalis (EAF) against permethrin (PER). Male wistar rats were treated daily by gavage during 7 days as follows: group C: negative control rats received 2 mL/kg bw of corn oil, group EAF: positive control rats received EAF at a dose of 200 mg/kg bw dissolved in water, group PER: rats received PER at a dose of 34.05 mg/kg bw and group (PER + EAF): rats received PER (34.05 mg/kg bw) and EAF (200 mg/kg bw). In vitro study showed the ability of EAF to inhibit protein denaturation and heat-induced hemolysis confirming its anti-inflammatory activity. In vivo, PER treatment decreased calcium (Ca) and phosphorus (P) levels and increased lactate dehydrogenase (LDH) activity in plasma. It induced oxidative stress objectified by an increase in the lipid peroxidation and protein oxidation and a perturbation of antioxidant system in kidney and mitochondria. The activities of NADH-ubiquinone reductase, ubiquinol-cytochrome C reductase and cytochrome C oxidase activities were reduced. These alterations were confirmed by histopathological studies. Co-treatment with EAF improved the antioxidant status and mitochondrial bioenergetics. The nephroprotective effects of EAF could be attributed to its modulation of detoxification enzymes and/or free radical scavenging actions.
Collapse
Affiliation(s)
- Nissaf Aoiadni
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia.
| | - Hajer Jdidi
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia
| | - Abdelfattah El Feki
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia
| | - Hamadi Fetoui
- Laboratory of Toxicology and Environmental Health.LR17ES06, Sciences Faculty of Sfax, University of Sfax, 14 BP1171, 3000, Sfax, Tunisia
| | - Fatma Ghorbel Koubaa
- Laboratory of Animal Eco-Physiology, Faculty of Sciences of Sfax, Tunisia, Street of Soukra Km 3.5, BP 1171, 9 3000, Sfax, CP, Tunisia
| |
Collapse
|
59
|
Chen Y, Qin Q, Zhao W, Luo D, Huang Y, Liu G, Kuang Y, Cao Y, Chen Y. Carnosol Reduced Pathogenic Protein Aggregation and Cognitive Impairment in Neurodegenerative Diseases Models via Improving Proteostasis and Ameliorating Mitochondrial Disorders. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10490-10505. [PMID: 35973126 DOI: 10.1021/acs.jafc.2c02665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's disease, Parkinson's disease, and Huntington's disease are incurable diseases with progressive loss of neural function and require urgent development of effective treatments. Carnosol (CL) reportedly has a pharmacological effect in the prevention of dementia. Nevertheless, the mechanisms of CL's neuroprotection are not entirely clear. The present study aimed to investigate the effects and mechanisms of CL-mediated neuroprotection through Caenorhabditis elegans models. First, CL restored ND protein homeostasis via inhibiting the IIS pathway, regulating MAPK signaling, and simultaneously activating molecular chaperone, thus inhibiting amyloid peptide (Aβ), polyglutamine (polyQ), and α-synuclein (α-syn) deposition and reducing protein disruption-mediated behavioral and cognitive impairments as well as neuronal damages. Furthermore, CL could repair mitochondrial structural damage via improving the mitochondrial membrane protein function and mitochondrial structural homeostasis and improve mitochondrial functional defects via increasing adenosine triphosphate contents, mitochondrial membrane potential, and reactive oxygen species levels, suggesting that CL could improve the ubiquitous mitochondrial defects in NDs. More importantly, we found that CL activated mitochondrial kinetic homeostasis related genes to improve the mitochondrial homeostasis and dysfunction in NDs. Meanwhile, CL up-regulated unc-17, cho-1, and cha-1 genes to alleviate Aβ-mediated cholinergic neurological disorders and activated Notch signaling and the Wnt pathway to diminish polyQ- and α-syn-induced ASH neurons as well as dopaminergic neuron damages. Overall, our study clarified the beneficial anti-ND neuroprotective effects of CL in different aspects and provided new insights into developing CL into products with preventive and therapeutic effects on NDs.
Collapse
Affiliation(s)
- Yun Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Qiao Qin
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Wen Zhao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Danxia Luo
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Yingyin Huang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Guo Liu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Yong Kuang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| | - Yunjiao Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510640 Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510640 Guangdong, China
| |
Collapse
|
60
|
Gu Y, Guerra F, Hu M, Pope A, Sung K, Yang W, Jetha S, Shoff TA, Gunatilake T, Dahlkamp O, Shi LZ, Manganelli F, Nolano M, Zhou Y, Ding J, Bucci C, Wu C. Mitochondria dysfunction in Charcot Marie Tooth 2B Peripheral Sensory Neuropathy. Commun Biol 2022; 5:717. [PMID: 35851620 PMCID: PMC9293960 DOI: 10.1038/s42003-022-03632-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 06/23/2022] [Indexed: 11/21/2022] Open
Abstract
Rab7 GTPase regulates mitochondrial morphology and function. Missense mutation(s) of Rab7 underlies the pathogenesis of Charcot Marie Tooth 2B (CMT2B) peripheral neuropathy. Herein, we investigate how mitochondrial morphology and function are impacted by the CMT2B associated Rab7V162M mutation. In contrast to recent studies of using heterologous overexpression systems, our results demonstrate significant mitochondrial fragmentation in both human CMT2B patient fibroblasts and CMT2B embryonic fibroblasts (MEFs). Primary cultured E18 dorsal root ganglion (DRG) sensory neurons also show mitochondrial fragmentation and altered axonal mitochondrial movement. In addition, we demonstrate that inhibitors to either the mitochondrial fission protein Drp1 or to the nucleotide binding to Rab7 normalize the mitochondrial deficits in both MEFs and E18 cultured DRG neurons. Our study reveals, for the first time, that expression of CMT2B Rab7 mutation at the physiological level enhances Drp1 activity to promote mitochondrial fission, potentially underlying selective vulnerability of peripheral sensory neurons in CMT2B pathogenesis.
Collapse
Affiliation(s)
- Yingli Gu
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
- Department of Neurology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), Università del Salento, Via Provinciale Lecce-Monteroni n. 165, 73100, Lecce, Italy
| | - Mingzheng Hu
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Alexander Pope
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Kijung Sung
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Wanlin Yang
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
- Department of Neurology, Zhujiang Hospital of Southern Medical University Guangzhou, Guangzhou, 510280, Guangdong Sheng, China
| | - Simone Jetha
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Thomas A Shoff
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Tessanya Gunatilake
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Owen Dahlkamp
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA
| | - Linda Zhixia Shi
- Department of Bioengineering, University of California San Diego, La Jolla, 92093, CA, USA
| | - Fiore Manganelli
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Via Sergio Pansini 5, 80131, Naples, Italy
| | - Maria Nolano
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Via Sergio Pansini 5, 80131, Naples, Italy
| | - Yue Zhou
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030, China
| | - Jianqing Ding
- Institute of Neurology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), Università del Salento, Via Provinciale Lecce-Monteroni n. 165, 73100, Lecce, Italy.
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, 92093, CA, USA.
| |
Collapse
|
61
|
Kajjumba GW, Bokota RE, Attene-Ramos M, Marti EJ. Evaluation of disinfection byproducts for their ability to affect mitochondrial function. J Environ Sci (China) 2022; 117:295-304. [PMID: 35725082 DOI: 10.1016/j.jes.2022.05.014] [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: 03/15/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
In the race to deliver clean water to communities through potable water reuse, disinfection and water quality assessment are and will continue to be fundamental factors. There are over 700 disinfection byproducts (DBPs) in water; evaluating each compound is practically impossible and very time consuming. A bioanalytical approach could be an answer to this challenge. In this work, the response of four major classes of DBPs toward mitochondrial membrane potential (ΔΨm) and cytoplasmic adenosine triphosphate (C-ATP) was investigated with human carcinoma (HepG2) cells. Within 90 min of cell exposure, only the haloacetic acid (HAA) mixture caused a cytotoxic response as measured by C-ATP. All four groups (haloacetonitriles (HANs), trihalomethanes (THMs), nitrosamines (NOAs), and HAAs) responded well to ΔΨm, R2 > 0.70. Based on the half-maximum concentration that evoked a 50% response in ΔΨm, the response gradient was HANs >> HAAs ∼ THM > NOAs. The inhibition of the ΔΨm by HANs is driven by dibromoacetonitrile (DBAN), while dichloroacetonitrile (DCAN) did not cause a significant change in the ΔΨm at less than 2000 µM. A mixture of HANs exhibited an antagonistic behavior on the ΔΨm compared to individual compounds. If water samples are concentrated to increase HAN concentrations, especially DBAN, then ΔΨm could be used as a biomonitoring tool for DBP toxicity.
Collapse
Affiliation(s)
| | - Rachael E Bokota
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington DC, USA
| | - Matias Attene-Ramos
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, Washington DC, USA
| | - Erica J Marti
- Department of Civil and Environmental Engineering, University of Nevada, Las Vegas, NV, USA.
| |
Collapse
|
62
|
The Regulation and Characterization of Mitochondrial-Derived Methylmalonic Acid in Mitochondrial Dysfunction and Oxidative Stress: From Basic Research to Clinical Practice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7043883. [PMID: 35656023 PMCID: PMC9155905 DOI: 10.1155/2022/7043883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/16/2022] [Accepted: 04/23/2022] [Indexed: 01/11/2023]
Abstract
Methylmalonic acid (MMA) can act as a diagnosis of hereditary methylmalonic acidemia and assess the status of vitamin B12. Moreover, as a new potential biomarker, it has been widely reported to be associated with the progression and prognosis of chronic diseases such as cardiovascular events, renal insufficiency, cognitive impairment, and cancer. MMA accumulation may cause oxidative stress and impair mitochondrial function, disrupt cellular energy metabolism, and trigger cell death. This review primarily focuses on the mechanisms and epidemiology or progression in the clinical study on MMA.
Collapse
|
63
|
Hawrysh PJ, Myrka AM, Buck LT. Review: A history and perspective of mitochondria in the context of anoxia tolerance. Comp Biochem Physiol B Biochem Mol Biol 2022; 260:110733. [PMID: 35288242 DOI: 10.1016/j.cbpb.2022.110733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 01/01/2023]
Abstract
Symbiosis is found throughout nature, but perhaps nowhere is it more fundamental than mitochondria in all eukaryotes. Since mitochondria were discovered and mechanisms of oxygen reduction characterized, an understanding gradually emerged that these organelles were involved not just in the combustion of oxygen, but also in the sensing of oxygen. While multiple hypotheses exist to explain the mitochondrial involvement in oxygen sensing, key elements are developing that include potassium channels and reactive oxygen species. To understand how mitochondria contribute to oxygen sensing, it is informative to study a model system which is naturally adapted to survive extended periods without oxygen. Amongst air-breathing vertebrates, the most highly adapted are western painted turtles (Chrysemys picta bellii), which overwinter in ice-covered and anoxic water bodies. Through research of this animal, it was postulated that metabolic rate depression is key to anoxic survival and that mitochondrial regulation is a key aspect. When faced with anoxia, excitatory neurotransmitter receptors in turtle brain are inhibited through mitochondrial calcium release, termed "channel arrest". Simultaneously, inhibitory GABAergic signalling contributes to the "synaptic arrest" of excitatory action potential firing through a pathway dependent on mitochondrial depression of ROS generation. While many pathways are implicated in mitochondrial oxygen sensing in turtles, such as those of adenosine, ATP turnover, and gaseous transmitters, an apparent point of intersection is the mitochondria. In this review we will explore how an organelle that was critical for organismal complexity in an oxygenated world has also become a potentially important oxygen sensor.
Collapse
Affiliation(s)
- Peter John Hawrysh
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Alexander Morley Myrka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Leslie Thomas Buck
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
64
|
Reiss AB, Ahmed S, Dayaramani C, Glass AD, Gomolin IH, Pinkhasov A, Stecker MM, Wisniewski T, De Leon J. The role of mitochondrial dysfunction in Alzheimer's disease: A potential pathway to treatment. Exp Gerontol 2022; 164:111828. [DOI: 10.1016/j.exger.2022.111828] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/15/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
|
65
|
Bordoni M, Pansarasa O, Scarian E, Cristofani R, Leone R, Fantini V, Garofalo M, Diamanti L, Bernuzzi S, Gagliardi S, Carelli S, Poletti A, Cereda C. Lysosomes Dysfunction Causes Mitophagy Impairment in PBMCs of Sporadic ALS Patients. Cells 2022; 11:cells11081272. [PMID: 35455952 PMCID: PMC9030813 DOI: 10.3390/cells11081272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023] Open
Abstract
Mitochondria alterations are present in tissues derived from patients and animal models, but no data are available for peripheral blood mononuclear cells (PBMCs) of ALS patients. This work aims to investigate mitophagy in PBMCs of sporadic (sALS) patients and how this pathway can be tuned by using small molecules. We found the presence of morphologically atypical mitochondria by TEM and morphological abnormalities by MitoTracker™. We found a decreased number of healthy mitochondria in sALS PBMCs and an impairment of mitophagy with western blot and immunofluorescence. After rapamycin treatment, we found a higher increase in the LC3 marker in sALS PBMCs, while after NH4Cl treatment, we found a lower increase in the LC3 marker. Finally, mTOR-independent autophagy induction with trehalose resulted in a significant decrease in the lysosomes level sALS PBMCs. Our data suggest that the presence of morphologically altered mitochondria and an inefficient turnover of damaged mitochondria in PBMCs of sALS patients rely on the impairment of the mitophagy pathway. We also found that the induction of the mTOR-independent autophagy pathway leads to a decrease in lysosomes level, suggesting a more sensitivity of sALS PBMCs to trehalose. Such evidence suggests that trehalose could represent an effective treatment for ALS patients.
Collapse
Affiliation(s)
- Matteo Bordoni
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Orietta Pansarasa
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Correspondence: ; Tel.: +0382-380-248
| | - Eveljn Scarian
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | | | - Valentina Fantini
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy;
| | - Maria Garofalo
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Luca Diamanti
- Neuroncology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy;
| | - Stefano Bernuzzi
- Immunohematological and Transfusional Service and Centre of Transplantation Immunology, IRCCS “San Matteo Foundation”, 27100 Pavia, Italy;
| | - Stella Gagliardi
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, 20157 Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | - Cristina Cereda
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| |
Collapse
|
66
|
Fessler E, Krumwiede L, Jae LT. DELE1 tracks perturbed protein import and processing in human mitochondria. Nat Commun 2022; 13:1853. [PMID: 35388015 PMCID: PMC8986780 DOI: 10.1038/s41467-022-29479-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/11/2022] [Indexed: 12/15/2022] Open
Abstract
Protein homeostatic control of mitochondria is key to age-related diseases and organismal decline. However, it is unknown how the diverse types of stress experienced by mitochondria can be integrated and appropriately responded to in human cells. Here we identify perturbations in the ancient conserved processes of mitochondrial protein import and processing as sources of DELE1 activation: DELE1 is continuously sorted across both mitochondrial membranes into the matrix and detects different types of perturbations along the way. DELE1 molecules in transit can become licensed for mitochondrial release and stress signaling through proteolytic removal of N-terminal sorting signals. Import defects that occur at the mitochondrial surface allow DELE1 precursors to bind and activate downstream factor HRI without the need for cleavage. Genome-wide genetics reveal that DELE1 additionally responds to compromised presequence processing by the matrix proteases PITRM1 and MPP, which are mutated in neurodegenerative diseases. These mechanisms rationalize DELE1-dependent mitochondrial stress integration in the human system and may inform future therapies of neuropathies.
Collapse
Affiliation(s)
- Evelyn Fessler
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, 81377, Munich, Germany.
| | - Luisa Krumwiede
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, 81377, Munich, Germany
| | - Lucas T Jae
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Strasse 25, 81377, Munich, Germany.
| |
Collapse
|
67
|
Acute and Delayed Effects of Mechanical Injury on Calcium Homeostasis and Mitochondrial Potential of Primary Neuroglial Cell Culture: Potential Causal Contributions to Post-Traumatic Syndrome. Int J Mol Sci 2022; 23:ijms23073858. [PMID: 35409216 PMCID: PMC8998891 DOI: 10.3390/ijms23073858] [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: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 02/07/2023] Open
Abstract
In vitro models of traumatic brain injury (TBI) help to elucidate the pathological mechanisms responsible for cell dysfunction and death. To simulate in vitro the mechanical brain trauma, primary neuroglial cultures were scratched during different periods of network formation. Fluorescence microscopy was used to measure changes in intracellular free Ca2+ concentration ([Ca2+]i) and mitochondrial potential (ΔΨm) a few minutes later and on days 3 and 7 after scratching. An increase in [Ca2+]i and a decrease in ΔΨm were observed ~10 s after the injury in cells located no further than 150–200 µm from the scratch border. Ca2+ entry into cells during mechanical damage of the primary neuroglial culture occurred predominantly through the NMDA-type glutamate ionotropic channels. MK801, an inhibitor of this type of glutamate receptor, prevented an acute increase in [Ca2+]i in 99% of neurons. Pathological changes in calcium homeostasis persisted in the primary neuroglial culture for one week after injury. Active cell migration in the scratch area occurred on day 11 after neurotrauma and was accompanied by a decrease in the ratio of live to dead cells in the areas adjacent to the injury. Immunohistochemical staining of glial fibrillary acidic protein and β-III tubulin showed that neuronal cells migrated to the injured area earlier than glial cells, but their repair potential was insufficient for survival. Mitochondrial Ca2+ overload and a drop in ΔΨm may cause delayed neuronal death and thus play a key role in the development of the post-traumatic syndrome. Preventing prolonged ΔΨm depolarization may be a promising therapeutic approach to improve neuronal survival after traumatic brain injury.
Collapse
|
68
|
Bakaeva Z, Lizunova N, Tarzhanov I, Boyarkin D, Petrichuk S, Pinelis V, Fisenko A, Tuzikov A, Sharipov R, Surin A. Lipopolysaccharide From E. coli Increases Glutamate-Induced Disturbances of Calcium Homeostasis, the Functional State of Mitochondria, and the Death of Cultured Cortical Neurons. Front Mol Neurosci 2022; 14:811171. [PMID: 35069113 PMCID: PMC8767065 DOI: 10.3389/fnmol.2021.811171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Lipopolysaccharide (LPS), a fragment of the bacterial cell wall, specifically interacting with protein complexes on the cell surface, can induce the production of pro-inflammatory and apoptotic signaling molecules, leading to the damage and death of brain cells. Similar effects have been noted in stroke and traumatic brain injury, when the leading factor of death is glutamate (Glu) excitotoxicity too. But being an amphiphilic molecule with a significant hydrophobic moiety and a large hydrophilic region, LPS can also non-specifically bind to the plasma membrane, altering its properties. In the present work, we studied the effect of LPS from Escherichia coli alone and in combination with the hyperstimulation of Glu-receptors on the functional state of mitochondria and Ca2+ homeostasis, oxygen consumption and the cell survival in primary cultures from the rats brain cerebellum and cortex. In both types of cultures, LPS (0.1–10 μg/ml) did not change the intracellular free Ca2+ concentration ([Ca2+]i) in resting neurons but slowed down the median of the decrease in [Ca2+]i on 14% and recovery of the mitochondrial potential (ΔΨm) after Glu removal. LPS did not affect the basal oxygen consumption rate (OCR) of cortical neurons; however, it did decrease the acute OCR during Glu and LPS coapplication. Evaluation of the cell culture survival using vital dyes and the MTT assay showed that LPS (10 μg/ml) and Glu (33 μM) reduced jointly and separately the proportion of live cortical neurons, but there was no synergism or additive action. LPS-effects was dependent on the type of culture, that may be related to both the properties of neurons and the different ratio between neurons and glial cells in cultures. The rapid manifestation of these effects may be the consequence of the direct effect of LPS on the rheological properties of the cell membrane.
Collapse
Affiliation(s)
- Zanda Bakaeva
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Department of General Biology and Physiology, Kalmyk State University named after B.B. Gorodovikov, Elista, Russia
- *Correspondence: Zanda Bakaeva, ,
| | - Natalia Lizunova
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Department of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ivan Tarzhanov
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Institute of Pharmacy, The Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Dmitrii Boyarkin
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Svetlana Petrichuk
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Vsevolod Pinelis
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Andrey Fisenko
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Alexander Tuzikov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Rinat Sharipov
- Laboratory of Fundamental and Applied Problems of Pain, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Surin
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Laboratory of Fundamental and Applied Problems of Pain, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
69
|
Zhang X, Tomar N, Kandel SM, Audi SH, Cowley AW, Dash RK. Substrate- and Calcium-Dependent Differential Regulation of Mitochondrial Oxidative Phosphorylation and Energy Production in the Heart and Kidney. Cells 2021; 11:131. [PMID: 35011693 PMCID: PMC8750792 DOI: 10.3390/cells11010131] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dehydrogenases are differentially stimulated by Ca2+. Ca2+ has also diverse regulatory effects on mitochondrial transporters and other enzymes. However, the consequences of these regulatory effects on mitochondrial oxidative phosphorylation (OxPhos) and ATP production, and the dependencies of these consequences on respiratory substrates, have not been investigated between the kidney and heart despite the fact that kidney energy requirements are second only to those of the heart. Our objective was, therefore, to elucidate these relationships in isolated mitochondria from the kidney outer medulla (OM) and heart. ADP-induced mitochondrial respiration was measured at different CaCl2 concentrations in the presence of various respiratory substrates, including pyruvate + malate (PM), glutamate + malate (GM), alpha-ketoglutarate + malate (AM), palmitoyl-carnitine + malate (PCM), and succinate + rotenone (SUC + ROT). The results showed that, in both heart and OM mitochondria, and for most complex I substrates, Ca2+ effects are biphasic: small increases in Ca2+ concentration stimulated, while large increases inhibited mitochondrial respiration. Furthermore, significant differences in substrate- and Ca2+-dependent O2 utilization towards ATP production between heart and OM mitochondria were observed. With PM and PCM substrates, Ca2+ showed more prominent stimulatory effects in OM than in heart mitochondria, while with GM and AM substrates, Ca2+ had similar biphasic regulatory effects in both OM and heart mitochondria. In contrast, with complex II substrate SUC + ROT, only inhibitory effects on mitochondrial respiration was observed in both the heart and the OM. We conclude that the regulatory effects of Ca2+ on mitochondrial OxPhos and ATP synthesis are biphasic, substrate-dependent, and tissue-specific.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Sunil M. Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Said H. Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee, WI 53223, USA;
| | - Allen W. Cowley
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
70
|
Tmem160 contributes to the establishment of discrete nerve injury-induced pain behaviors in male mice. Cell Rep 2021; 37:110152. [PMID: 34936870 DOI: 10.1016/j.celrep.2021.110152] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 09/01/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
Chronic pain is a prevalent medical problem, and its molecular basis remains poorly understood. Here, we demonstrate the significance of the transmembrane protein (Tmem) 160 for nerve injury-induced neuropathic pain. An extensive behavioral assessment suggests a pain modality- and entity-specific phenotype in male Tmem160 global knockout (KO) mice: delayed establishment of tactile hypersensitivity and alterations in self-grooming after nerve injury. In contrast, Tmem160 seems to be dispensable for other nerve injury-induced pain modalities, such as non-evoked and movement-evoked pain, and for other pain entities. Mechanistically, we show that global KO males exhibit dampened neuroimmune signaling and diminished TRPA1-mediated activity in cultured dorsal root ganglia. Neither these changes nor altered pain-related behaviors are observed in global KO female and male peripheral sensory neuron-specific KO mice. Our findings reveal Tmem160 as a sexually dimorphic factor contributing to the establishment, but not maintenance, of discrete nerve injury-induced pain behaviors in male mice.
Collapse
|
71
|
Sahu K, Langeh U, Singh C, Singh A. Crosstalk between anticancer drugs and mitochondrial functions. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100047. [PMID: 34909674 PMCID: PMC8663961 DOI: 10.1016/j.crphar.2021.100047] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 01/12/2023] Open
Abstract
Chemotherapy is an important component of cancer treatment, which has side effects like vomiting, peripheral neuropathy, and numerous organ toxicity but the most significant outcomes of chemotherapy are cognitive impairment, which is mainly referred to as chemobrain or CICI (chemotherapy-induced cognitive impairment). It is characterized by difficulty with language, concentrating, processing speed, learning, and memory, as it affects the hippocampus areas of the brain. Mitochondrial dysfunction and oxidative stress are one of the major mechanisms causing chemobrain. The generation of reactive oxygen species (byproducts of oxidative phosphorylation) mainly occurs in mitochondria that play a prominent role in the induction of oxidative stress. The homeostasis of ROS in the mitochondria is maintained by mitochondrial antioxidant mechanism via enzymes like catalase, glutathione, and superoxide dismutase. Lungs and breast cancer are the two most common types of cancer, which are the most leading cancers in the world with about 4.18 million cases. In this review we exposed the current knowledge regarding chemotherapy-induced oxidative stress and mitochondrial dysfunction to cause cognitive impairment.We especially focused on the antineoplastic agent (ADRIAMYCIN, CYCLOPHOSPHAMIDE), platinum group agent CISPLATIN, antimetabolite agents (METHOTREXATE), and nitrogen mustard agent (CARMUSTINE) which increase oxidative stress and inflammatory markers in the PNS (peripheral nervous system) as well as the central nervous system. We also highlight the behavioural and functional changes in the brain.
Collapse
Affiliation(s)
- Kuleshwar Sahu
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Urvashi Langeh
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Charan Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| |
Collapse
|
72
|
Kajjumba GW, Attene-Ramos M, Marti EJ. Toxicity of lanthanide coagulants assessed using four in vitro bioassays. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 800:149556. [PMID: 34399349 DOI: 10.1016/j.scitotenv.2021.149556] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Rare earth element (REE) coagulants are prime contenders in wastewater treatment plants to remove phosphorus; unlike typical coagulants, they are not affected by pH. However, the use of REEs in wastewater treatment could mean increased human exposure to lanthanides (Ln) through wastewater effluent discharge to the environment or through water reuse. Information on the toxicity of lanthanides is scarce and, where available, there are conflicting views. Using in vitro bioassays, we assessed lanthanide toxicity by evaluating four relevant endpoints: the change in mitochondrial membrane potential (Δψm), intracellular adenosine triphosphate (I-ATP), genotoxicity, and cell viability. At less than 5000 μmol-Ln3+/L, lanthanides increased the Δψm, while above 5000 μmol-Ln3+/L, the Δψm level plummeted. The measure of I-ATP indicated constant levels of ATP up to 250 μmol-Ln3+/L, above which the I-ATP decreased steadily; the concentration of La, Ce, Gd, and Lu that triggered half of the cells to become ATP-inactive is 794, 1505, 1488, 1115 μmol-Ln3+/L, respectively. Although La and Lu accelerated cell death in shorter studies (24 h), chronic studies using three cell growth cycles showed cell recovery. Lanthanides exhibited antagonistic toxicity at less than 1000 μmol-Ln3+/L. However, the introduction of heavy REEs in a solution amplified lanthanide toxicity. Tested lanthanides appear to pose little risk, which could pave the way for lanthanide application in wastewater treatment.
Collapse
Affiliation(s)
- George William Kajjumba
- Department of Civil and Environmental Engineering and Construction, University of Nevada, Las Vegas, 4505 S. Maryland Pkwy., Las Vegas, NV 89154, USA.
| | - Matias Attene-Ramos
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, George Washington University, DC, USA
| | - Erica J Marti
- Department of Civil and Environmental Engineering and Construction, University of Nevada, Las Vegas, 4505 S. Maryland Pkwy., Las Vegas, NV 89154, USA.
| |
Collapse
|
73
|
Mitochondrial Phenotypes in Parkinson's Diseases-A Focus on Human iPSC-Derived Dopaminergic Neurons. Cells 2021; 10:cells10123436. [PMID: 34943944 PMCID: PMC8699816 DOI: 10.3390/cells10123436] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Established disease models have helped unravel the mechanistic underpinnings of pathological phenotypes in Parkinson’s disease (PD), the second most common neurodegenerative disorder. However, these discoveries have been limited to relatively simple cellular systems and animal models, which typically manifest with incomplete or imperfect recapitulation of disease phenotypes. The advent of induced pluripotent stem cells (iPSCs) has provided a powerful scientific tool for investigating the underlying molecular mechanisms of both familial and sporadic PD within disease-relevant cell types and patient-specific genetic backgrounds. Overwhelming evidence supports mitochondrial dysfunction as a central feature in PD pathophysiology, and iPSC-based neuronal models have expanded our understanding of mitochondrial dynamics in the development and progression of this devastating disorder. The present review provides a comprehensive assessment of mitochondrial phenotypes reported in iPSC-derived neurons generated from PD patients’ somatic cells, with an emphasis on the role of mitochondrial respiration, morphology, and trafficking, as well as mitophagy and calcium handling in health and disease. Furthermore, we summarize the distinguishing characteristics of vulnerable midbrain dopaminergic neurons in PD and report the unique advantages and challenges of iPSC disease modeling at present, and for future mechanistic and therapeutic applications.
Collapse
|
74
|
Galiakberova AA, Surin AM, Bakaeva ZV, Sharipov RR, Zhang D, Dorovskoy DA, Shakirova KM, Fisenko AP, Dashinimaev EB. IPSC-Derived Human Neurons with GCaMP6s Expression Allow In Vitro Study of Neurophysiological Responses to Neurochemicals. Neurochem Res 2021; 47:952-966. [PMID: 34855047 PMCID: PMC8891101 DOI: 10.1007/s11064-021-03497-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022]
Abstract
The study of human neurons and their interaction with neurochemicals is difficult due to the inability to collect primary biomaterial. However, recent advances in the cultivation of human stem cells, methods for their neuronal differentiation and chimeric fluorescent calcium indicators have allowed the creation of model systems in vitro. In this paper we report on the development of a method to obtain human neurons with the GCaMP6s calcium indicator, based on a human iPSC line with the TetON–NGN2 transgene complex. The protocol we developed allows us quickly, conveniently and efficiently obtain significant amounts of human neurons suitable for the study of various neurochemicals and their effects on specific neurophysiological activity, which can be easily registered using fluorescence microscopy. In the neurons we obtained, glutamate (Glu) induces rises in [Ca2+]i which are caused by ionotropic receptors for Glu, predominantly of the NMDA-type. Taken together, these facts allow us to consider the model we have created to be a useful and successful development of this technology.
Collapse
Affiliation(s)
- A A Galiakberova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, Moscow, Russia, 117997.
- Faculty of Biology, Lomonosov Moscow State University, GSP-1, Leninskie Gory, Moscow, Russia, 119991.
| | - A M Surin
- Laboratory of Neurobiology, "National Medical Research Center of Children's Health", Russian Ministry of Health, Lomonosov Avenue, Moscow, Russia, 119991
- Laboratory of Pathology of Ion Transport and Intracellular Signaling, Institute of General Pathology and Pathophysiology, Baltiyskaya St., Moscow, Russia, 125315
| | - Z V Bakaeva
- Laboratory of Neurobiology, "National Medical Research Center of Children's Health", Russian Ministry of Health, Lomonosov Avenue, Moscow, Russia, 119991
- Department of General Biology and Physiology, Gorodovikov Kalmyk State University, Pushkin St., Elista, Russia, 358000
| | - R R Sharipov
- Laboratory of Pathology of Ion Transport and Intracellular Signaling, Institute of General Pathology and Pathophysiology, Baltiyskaya St., Moscow, Russia, 125315
| | - Dongxing Zhang
- Moscow Institute of Physics and Technology (State University), Institutskiy per., 141701, Dolgoprudny, Russia
| | - D A Dorovskoy
- Moscow Institute of Physics and Technology (State University), Institutskiy per., 141701, Dolgoprudny, Russia
| | - K M Shakirova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, Moscow, Russia, 117997
| | - A P Fisenko
- Laboratory of Neurobiology, "National Medical Research Center of Children's Health", Russian Ministry of Health, Lomonosov Avenue, Moscow, Russia, 119991
| | - E B Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov Street, Moscow, Russia, 117997
- Moscow Institute of Physics and Technology (State University), Institutskiy per., 141701, Dolgoprudny, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov St., Moscow, Russia, 119334
| |
Collapse
|
75
|
Ong HC, Coimbra JTS, Kwek G, Ramos MJ, Xing B, Fernandes PA, García F. Alkyl vs. aryl modifications: a comparative study on modular modifications of triphenylphosphonium mitochondrial vectors. RSC Chem Biol 2021; 2:1643-1650. [PMID: 34977579 PMCID: PMC8637833 DOI: 10.1039/d1cb00099c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/21/2021] [Indexed: 11/21/2022] Open
Abstract
Triphenylphosphonium (TPP+) moieties are commonly conjugated to drug molecules to confer mitochondrial selectivity due to their positive charge and high lipophilicity. Although optimisation of lipophilicity can be achieved by modifying the length of the alkyl linkers between the TPP+ moiety and the drug molecule, it is not always possible. While methylation of the TPP+ moiety is a viable alternative to increase lipophilicity and mitochondrial accumulation, there are no studies comparing these two separate modular approaches. Thus, we have systematically designed, synthesised and tested a range of TPP+ molecules with varying alkyl chain lengths and degree of aryl methylation to compare the two modular methodologies for modulating lipophilicity. The ability of aryl/alkyl modified TPP+ to deliver cargo to the mitochondria was also evaluated by confocal imaging with a TPP+-conjugated fluorescein-based fluorophore. Furthermore, we have employed molecular dynamics simulations to understand the translocation of these molecules through biological membrane model systems. These results provide further insights into the thermodynamics of this process and the effect of alkyl and aryl modular modifications. Alkyl chain extension and aryl methylation can be employed to enhance mitochondrial uptake in triphenylphosphonium vectors. Here we compare these complementary strategies and their mitochondrial-targeting effects using a modular synthetic approach.![]()
Collapse
Affiliation(s)
- How Chee Ong
- School of Physical and Mathematical Sciences, Division of Chemistry and Biological Chemistry, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - João T S Coimbra
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto Rua do Campo Alegre s/n 4169-007 Portugal
| | - Germain Kwek
- School of Physical and Mathematical Sciences, Division of Chemistry and Biological Chemistry, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Maria J Ramos
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto Rua do Campo Alegre s/n 4169-007 Portugal
| | - Bengang Xing
- School of Physical and Mathematical Sciences, Division of Chemistry and Biological Chemistry, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Pedro A Fernandes
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto Rua do Campo Alegre s/n 4169-007 Portugal
| | - Felipe García
- School of Physical and Mathematical Sciences, Division of Chemistry and Biological Chemistry, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| |
Collapse
|
76
|
Markaki M, Tsagkari D, Tavernarakis N. Mitophagy mechanisms in neuronal physiology and pathology during ageing. Biophys Rev 2021; 13:955-965. [DOI: 10.1007/s12551-021-00894-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/01/2021] [Indexed: 12/26/2022] Open
|
77
|
Fibbi B, Marroncini G, Anceschi C, Naldi L, Peri A. Hyponatremia and Oxidative Stress. Antioxidants (Basel) 2021; 10:1768. [PMID: 34829639 PMCID: PMC8614907 DOI: 10.3390/antiox10111768] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/26/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022] Open
Abstract
Hyponatremia, i.e., the presence of a serum sodium concentration ([Na+]) < 136 mEq/L, is the most frequent electrolyte imbalance in the elderly and in hospitalized patients. Symptoms of acute hyponatremia, whose main target is the central nervous system, are explained by the "osmotic theory" and the neuronal swelling secondary to decreased extracellular osmolality, which determines cerebral oedema. Following the description of neurological and systemic manifestations even in mild and chronic hyponatremia, in the last decade reduced extracellular [Na+] was associated with detrimental effects on cellular homeostasis independently of hypoosmolality. Most of these alterations appeared to be elicited by oxidative stress. In this review, we focus on the role of oxidative stress on both osmolality-dependent and -independent impairment of cell and tissue functions observed in hyponatremic conditions. Furthermore, basic and clinical research suggested that oxidative stress appears to be a common denominator of the degenerative processes related to aging, cancer progression, and hyponatremia. Of note, low [Na+] is able to exacerbate multiple manifestations of senescence and to decrease progression-free and overall survival in oncologic patients.
Collapse
Affiliation(s)
- Benedetta Fibbi
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Giada Marroncini
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Cecilia Anceschi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Laura Naldi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Alessandro Peri
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| |
Collapse
|
78
|
Monaco CMF, Tarnopolsky MA, Dial AG, Nederveen JP, Rebalka IA, Nguyen M, Turner LV, Perry CGR, Ljubicic V, Hawke TJ. Normal to enhanced intrinsic mitochondrial respiration in skeletal muscle of middle- to older-aged women and men with uncomplicated type 1 diabetes. Diabetologia 2021; 64:2517-2533. [PMID: 34392397 DOI: 10.1007/s00125-021-05540-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/20/2021] [Indexed: 12/26/2022]
Abstract
AIMS/HYPOTHESIS This study interrogated mitochondrial respiratory function and content in skeletal muscle biopsies of healthy adults between 30 and 72 years old with and without uncomplicated type 1 diabetes. METHODS Participants (12 women/nine men) with type 1 diabetes (48 ± 11 years of age), without overt complications, were matched for age, sex, BMI and level of physical activity to participants without diabetes (control participants) (49 ± 12 years of age). Participants underwent a Bergström biopsy of the vastus lateralis to assess mitochondrial respiratory function using high-resolution respirometry and citrate synthase activity. Electron microscopy was used to quantify mitochondrial content and cristae (pixel) density. RESULTS Mean mitochondrial area density was 27% lower (p = 0.006) in participants with type 1 diabetes compared with control participants. This was largely due to smaller mitochondrial fragments in women with type 1 diabetes (-18%, p = 0.057), as opposed to a decrease in the total number of mitochondrial fragments in men with diabetes (-28%, p = 0.130). Mitochondrial respiratory measures, whether estimated per milligram of tissue (i.e. mass-specific) or normalised to area density (i.e. intrinsic mitochondrial function), differed between cohorts, and demonstrated sexual dimorphism. Mass-specific mitochondrial oxidative phosphorylation (OXPHOS) capacity with the substrates for complex I and complex II (CI + II) was significantly lower (-24%, p = 0.033) in women with type 1 diabetes compared with control participants, whereas mass-specific OXPHOS capacities with substrates for complex I only (pyruvate [CI pyr] or glutamate [CI glu]) or complex II only (succinate [CII succ]) were not different (p > 0.404). No statistical differences (p > 0.397) were found in mass-specific OXPHOS capacity in men with type 1 diabetes compared with control participants despite a 42% non-significant increase in CI glu OXPHOS capacity (p = 0.218). In contrast, intrinsic CI + II OXPHOS capacity was not different in women with type 1 diabetes (+5%, p = 0.378), whereas in men with type 1 diabetes it was 25% higher (p = 0.163) compared with control participants. Men with type 1 diabetes also demonstrated higher intrinsic OXPHOS capacity for CI pyr (+50%, p = 0.159), CI glu (+88%, p = 0.033) and CII succ (+28%, p = 0.123), as well as higher intrinsic respiratory rates with low (more physiological) concentrations of either ADP, pyruvate, glutamate or succinate (p < 0.012). Women with type 1 diabetes had higher (p < 0.003) intrinsic respiratory rates with low concentrations of succinate only. Calculated aerobic fitness (Physical Working Capacity Test [PWC130]) showed a strong relationship with mitochondrial respiratory function and content in the type 1 diabetes cohort. CONCLUSIONS/INTERPRETATION In middle- to older-aged adults with uncomplicated type 1 diabetes, we conclude that skeletal muscle mitochondria differentially adapt to type 1 diabetes and demonstrate sexual dimorphism. Importantly, these cellular alterations were significantly associated with our metric of aerobic fitness (PWC130) and preceded notable impairments in skeletal mass and strength.
Collapse
Affiliation(s)
- Cynthia M F Monaco
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Athan G Dial
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Maria Nguyen
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Lauren V Turner
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Sciences, Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
79
|
Ryan F, Khoshnam SE, Khodagholi F, Ashabi G, Ahmadiani A. How cytosolic compartments play safeguard functions against neuroinflammation and cell death in cerebral ischemia. Metab Brain Dis 2021; 36:1445-1467. [PMID: 34173922 DOI: 10.1007/s11011-021-00770-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 06/06/2021] [Indexed: 11/26/2022]
Abstract
Ischemic stroke is the second leading cause of mortality and disability globally. Neuronal damage following ischemic stroke is rapid and irreversible, and eventually results in neuronal death. In addition to activation of cell death signaling, neuroinflammation is also considered as another pathogenesis that can occur within hours after cerebral ischemia. Under physiological conditions, subcellular organelles play a substantial role in neuronal functionality and viability. However, their functions can be remarkably perturbed under neurological disorders, particularly cerebral ischemia. Therefore, their biochemical and structural response has a determining role in the sequel of neuronal cells and the progression of disease. However, their effects on cell death and neuroinflammation, as major underlying mechanisms of ischemic stroke, are still not understood. This review aims to provide a comprehensive overview of the contribution of each organelle on these pathological processes after ischemic stroke.
Collapse
Affiliation(s)
- Fari Ryan
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Centre, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, PO Box: 1417613151, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
80
|
Lujan BJ, Singh M, Singh A, Renden RB. Developmental shift to mitochondrial respiration for energetic support of sustained transmission during maturation at the calyx of Held. J Neurophysiol 2021; 126:976-996. [PMID: 34432991 PMCID: PMC8560424 DOI: 10.1152/jn.00333.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 11/24/2022] Open
Abstract
A considerable amount of energy is expended following presynaptic activity to regenerate electrical polarization and maintain efficient release and recycling of neurotransmitter. Mitochondria are the major suppliers of neuronal energy, generating ATP via oxidative phosphorylation. However, the specific utilization of energy from cytosolic glycolysis rather than mitochondrial respiration at the presynaptic terminal during synaptic activity remains unclear and controversial. We use a synapse specialized for high-frequency transmission in mice, the calyx of Held, to test the sources of energy used to maintain energy during short activity bursts (<1 s) and sustained neurotransmission (30-150 s). We dissect the role of presynaptic glycolysis versus mitochondrial respiration by acutely and selectively blocking these ATP-generating pathways in a synaptic preparation where mitochondria and synaptic vesicles are prolific, under near-physiological conditions. Surprisingly, if either glycolysis or mitochondrial ATP production is intact, transmission during repetitive short bursts of activity is not affected. In slices from young animals before the onset of hearing, where the synapse is not yet fully specialized, both glycolytic and mitochondrial ATP production are required to support sustained, high-frequency neurotransmission. In mature synapses, sustained transmission relies exclusively on mitochondrial ATP production supported by bath lactate, but not glycolysis. At both ages, we observe that action potential propagation begins to fail before defects in synaptic vesicle recycling. Our data describe a specific metabolic profile to support high-frequency information transmission at the mature calyx of Held, shifting during postnatal synaptic maturation from glycolysis to rely on monocarboxylates as a fuel source.NEW & NOTEWORTHY We dissect the role of presynaptic glycolysis versus mitochondrial respiration in supporting high-frequency neurotransmission, by acutely blocking these ATP-generating pathways at a synapse tuned for high-frequency transmission. We find that massive energy expenditure is required to generate failure when only one pathway is inhibited. Action potential propagation is lost before impaired synaptic vesicle recycling. Synaptic transmission is exclusively dependent on oxidative phosphorylation in mature synapses, indicating presynaptic glycolysis may be dispensable for ATP maintenance.
Collapse
Affiliation(s)
- Brendan J Lujan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada
| | - Mahendra Singh
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada
| | - Abhyudai Singh
- Electrical & Computer Engineering, University of Delaware, Newark, Delaware
| | - Robert B Renden
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada
| |
Collapse
|
81
|
Ding Q, Qi Y, Tsang SY. Mitochondrial Biogenesis, Mitochondrial Dynamics, and Mitophagy in the Maturation of Cardiomyocytes. Cells 2021; 10:cells10092463. [PMID: 34572112 PMCID: PMC8466139 DOI: 10.3390/cells10092463] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 01/01/2023] Open
Abstract
Pluripotent stem cells (PSCs) can undergo unlimited self-renewal and can differentiate into all the cell types present in our body, including cardiomyocytes. Therefore, PSCs can be an excellent source of cardiomyocytes for future regenerative medicine and medical research studies. However, cardiomyocytes obtained from PSC differentiation culture are regarded as immature structurally, electrophysiologically, metabolically, and functionally. Mitochondria are organelles responsible for various cellular functions such as energy metabolism, different catabolic and anabolic processes, calcium fluxes, and various signaling pathways. Cells can respond to cellular needs to increase the mitochondrial mass by mitochondrial biogenesis. On the other hand, cells can also degrade mitochondria through mitophagy. Mitochondria are also dynamic organelles that undergo continuous fusion and fission events. In this review, we aim to summarize previous findings on the changes of mitochondrial biogenesis, mitophagy, and mitochondrial dynamics during the maturation of cardiomyocytes. In addition, we intend to summarize whether changes in these processes would affect the maturation of cardiomyocytes. Lastly, we aim to discuss unanswered questions in the field and to provide insights for the possible strategies of enhancing the maturation of PSC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Qianqian Ding
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
| | - Yanxiang Qi
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China;
| | - Suk-Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China;
- Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong, China
- The Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: ; Tel.: +852-39431020
| |
Collapse
|
82
|
Carmichael K, Sullivan B, Lopez E, Sun L, Cai H. Diverse midbrain dopaminergic neuron subtypes and implications for complex clinical symptoms of Parkinson's disease. AGEING AND NEURODEGENERATIVE DISEASES 2021; 1. [PMID: 34532720 PMCID: PMC8442626 DOI: 10.20517/and.2021.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD), the most common degenerative movement disorder, is clinically manifested with various motor and non-motor symptoms. Degeneration of midbrain substantia nigra pas compacta (SNc) dopaminergic neurons (DANs) is generally attributed to the motor syndrome. The underlying neuronal mechanisms of non-motor syndrome are largely unexplored. Besides SNc, midbrain ventral tegmental area (VTA) DANs also produce and release dopamine and modulate movement, reward, motivation, and memory. Degeneration of VTA DANs also occurs in postmortem brains of PD patients, implying an involvement of VTA DANs in PD-associated non-motor symptoms. However, it remains to be established that there is a distinct segregation of different SNc and VTA DAN subtypes in regulating different motor and non-motor functions, and that different DAN subpopulations are differentially affected by normal ageing or PD. Traditionally, the distinction among different DAN subtypes was mainly based on the location of cell bodies and axon terminals. With the recent advance of single cell RNA sequencing technology, DANs can be readily classified based on unique gene expression profiles. A combination of specific anatomic and molecular markers shows great promise to facilitate the identification of DAN subpopulations corresponding to different behavior modules under normal and disease conditions. In this review, we first summarize the recent progress in characterizing genetically, anatomically, and functionally diverse midbrain DAN subtypes. Then, we provide perspectives on how the preclinical research on the connectivity and functionality of DAN subpopulations improves our current understanding of cell-type and circuit specific mechanisms of the disease, which could be critically informative for designing new mechanistic treatments.
Collapse
Affiliation(s)
- Kathleen Carmichael
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.,The Graduate Partnership Program of NIH and Brown University, National Institutes of Health, Bethesda, MD 20892, USA
| | - Breanna Sullivan
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elena Lopez
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
83
|
Hussain B, Fang C, Chang J. Blood-Brain Barrier Breakdown: An Emerging Biomarker of Cognitive Impairment in Normal Aging and Dementia. Front Neurosci 2021; 15:688090. [PMID: 34489623 PMCID: PMC8418300 DOI: 10.3389/fnins.2021.688090] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) plays a vital role in maintaining the specialized microenvironment of the neural tissue. It separates the peripheral circulatory system from the brain parenchyma while facilitating communication. Alterations in the distinct physiological properties of the BBB lead to BBB breakdown associated with normal aging and various neurodegenerative diseases. In this review, we first briefly discuss the aging process, then review the phenotypes and mechanisms of BBB breakdown associated with normal aging that further cause neurodegeneration and cognitive impairments. We also summarize dementia such as Alzheimer's disease (AD) and vascular dementia (VaD) and subsequently discuss the phenotypes and mechanisms of BBB disruption in dementia correlated with cognition decline. Overlaps between AD and VaD are also discussed. Techniques that could identify biomarkers associated with BBB breakdown are briefly summarized. Finally, we concluded that BBB breakdown could be used as an emerging biomarker to assist to diagnose cognitive impairment associated with normal aging and dementia.
Collapse
Affiliation(s)
- Basharat Hussain
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Fang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
84
|
Woo E, Sansing LH, Arnsten AFT, Datta D. Chronic Stress Weakens Connectivity in the Prefrontal Cortex: Architectural and Molecular Changes. CHRONIC STRESS 2021; 5:24705470211029254. [PMID: 34485797 PMCID: PMC8408896 DOI: 10.1177/24705470211029254] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/14/2021] [Indexed: 12/26/2022]
Abstract
Chronic exposure to uncontrollable stress causes loss of spines and dendrites in the prefrontal cortex (PFC), a recently evolved brain region that provides top-down regulation of thought, action, and emotion. PFC neurons generate top-down goals through recurrent excitatory connections on spines. This persistent firing is the foundation for higher cognition, including working memory, and abstract thought. However, exposure to acute uncontrollable stress drives high levels of catecholamine release in the PFC, which activates feedforward calcium-cAMP signaling pathways to open nearby potassium channels, rapidly weakening synaptic connectivity to reduce persistent firing. Chronic stress exposures can further exacerbate these signaling events leading to loss of spines and resulting in marked cognitive impairment. In this review, we discuss how stress signaling mechanisms can lead to spine loss, including changes to BDNF-mTORC1 signaling, calcium homeostasis, actin dynamics, and mitochondrial actions that engage glial removal of spines through inflammatory signaling. Stress signaling events may be amplified in PFC spines due to cAMP magnification of internal calcium release. As PFC dendritic spine loss is a feature of many cognitive disorders, understanding how stress affects the structure and function of the PFC will help to inform strategies for treatment and prevention.
Collapse
Affiliation(s)
- Elizabeth Woo
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA.,Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Lauren H Sansing
- Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| |
Collapse
|
85
|
Tang J, Zhuo Y, Li Y. Effects of Iron and Zinc on Mitochondria: Potential Mechanisms of Glaucomatous Injury. Front Cell Dev Biol 2021; 9:720288. [PMID: 34447755 PMCID: PMC8383321 DOI: 10.3389/fcell.2021.720288] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/22/2021] [Indexed: 12/26/2022] Open
Abstract
Glaucoma is the most substantial cause of irreversible blinding, which is accompanied by progressive retinal ganglion cell damage. Retinal ganglion cells are energy-intensive neurons that connect the brain and retina, and depend on mitochondrial homeostasis to transduce visual information through the brain. As cofactors that regulate many metabolic signals, iron and zinc have attracted increasing attention in studies on neurons and neurodegenerative diseases. Here, we summarize the research connecting iron, zinc, neuronal mitochondria, and glaucomatous injury, with the aim of updating and expanding the current view of how retinal ganglion cells degenerate in glaucoma, which can reveal novel potential targets for neuroprotection.
Collapse
Affiliation(s)
- Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
86
|
Amruta N, Bix G. ATN-161 Ameliorates Ischemia/Reperfusion-induced Oxidative Stress, Fibro-inflammation, Mitochondrial damage, and Apoptosis-mediated Tight Junction Disruption in bEnd.3 Cells. Inflammation 2021; 44:2377-2394. [PMID: 34420157 PMCID: PMC8380192 DOI: 10.1007/s10753-021-01509-9] [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: 03/23/2021] [Revised: 05/25/2021] [Accepted: 06/27/2021] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated the significance of endothelial cell-expressed α5β1 integrin in ischemic stroke, having shown that α5β1 integrin endothelial cell-selective knockout mice are significantly resistance to ischemic stroke injury via preservation of the tight junction protein claudin-5 and subsequent stabilization of the blood–brain barrier (BBB). In addition, inhibition of α5β1 by the small peptide noncompetitive integrin α5 inhibitor, ATN-161, is beneficial in a mouse model of ischemic stroke through reduction of infarct volume, edema, stabilization of the BBB, and reduced inflammation and immune cell infiltration into the brain. In continuation with our previous findings, we have further evaluated the mechanistic role of ATN-161 in vitro and found that oxygen and glucose deprivation and reperfusion (OGD/R)-induced inflammation, oxidative stress, apoptosis, mitochondrial depolarization, and fibrosis attenuate tight junction integrity via induction of α5, NLRP3, p-FAK, and p-AKT signaling in mouse brain endothelial cells. ATN-161 treatment (10 µM) effectively inhibited OGD/R-induced extracellular matrix (ECM) deposition by reducing integrin α5, MMP-9, and fibronectin expression, as well as reducing oxidative stress by reducing mitochondrial superoxide radicals, intracellular ROS, inflammation by reducing NLRP3 inflammasome, tight junction loss by reducing claudin-5 and ZO-1 expression levels, mitochondrial damage by inhibiting mitochondrial depolarization, and apoptosis via regulation of p-FAK and p-AKT levels. Taken together, our results further support therapeutically targeting α5 integrin with ATN-161, a safe, well-tolerated, and clinically validated peptide, in ischemic stroke.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA. .,Department of Neurology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA. .,Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA.
| |
Collapse
|
87
|
Bcl-xL Is Required by Primary Hippocampal Neurons during Development to Support Local Energy Metabolism at Neurites. BIOLOGY 2021; 10:biology10080772. [PMID: 34440004 PMCID: PMC8389656 DOI: 10.3390/biology10080772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/01/2021] [Accepted: 08/06/2021] [Indexed: 11/17/2022]
Abstract
B-cell lymphoma-extra large (Bcl-xL) is a mitochondrial protein known to inhibit mitochondria-dependent intrinsic apoptotic pathways. An increasing number of studies have demonstrated that Bcl-xL is critical in regulating neuronal energy metabolism and has a protective role in pathologies associated with an energy deficit. However, it is less known how Bcl-xL regulates physiological processes of the brain. In this study, we hypothesize that Bcl-xL is required for neurite branching and maturation during neuronal development by improving local energy metabolism. We found that the absence of Bcl-xL in rat primary hippocampal neurons resulted in mitochondrial dysfunction. Specifically, the ATP/ADP ratio was significantly decreased in the neurites of Bcl-xL depleted neurons. We further found that neurons transduced with Bcl-xL shRNA or neurons treated with ABT-263, a pharmacological inhibitor of Bcl-xL, showed impaired mitochondrial motility. Neurons lacking Bcl-xL had significantly decreased anterograde and retrograde movement of mitochondria and an increased stationary mitochondrial population when Bcl-xL was depleted by either means. These mitochondrial defects, including loss of ATP, impaired normal neurite development. Neurons lacking Bcl-xL showed significantly decreased neurite arborization, growth and complexity. Bcl-xL depleted neurons also showed impaired synapse formation. These neurons showed increased intracellular calcium concentration and were more susceptible to excitotoxic challenge. Bcl-xL may support positioning of mitochondria at metabolically demanding regions of neurites like branching points. Our findings suggest a role for Bcl-xL in physiological regulation of neuronal growth and development.
Collapse
|
88
|
Yoshikawa K. Necdin: A purposive integrator of molecular interaction networks for mammalian neuron vitality. Genes Cells 2021; 26:641-683. [PMID: 34338396 PMCID: PMC9290590 DOI: 10.1111/gtc.12884] [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: 06/04/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 12/29/2022]
Abstract
Necdin was originally found in 1991 as a hypothetical protein encoded by a neural differentiation‐specific gene transcript in murine embryonal carcinoma cells. Virtually all postmitotic neurons and their precursor cells express the necdin gene (Ndn) during neuronal development. Necdin mRNA is expressed only from the paternal allele through genomic imprinting, a placental mammal‐specific epigenetic mechanism. Necdin and its homologous MAGE (melanoma antigen) family, which have evolved presumedly from a subcomplex component of the SMC5/6 complex, are expressed exclusively in placental mammals. Paternal Ndn‐mutated mice totally lack necdin expression and exhibit various types of neuronal abnormalities throughout the nervous system. Ndn‐null neurons are vulnerable to detrimental stresses such as DNA damage. Necdin also suppresses both proliferation and apoptosis of neural stem/progenitor cells. Functional analyses using Ndn‐manipulated cells reveal that necdin consistently exerts antimitotic, anti‐apoptotic and prosurvival effects. Necdin interacts directly with a number of regulatory proteins including E2F1, p53, neurotrophin receptors, Sirt1 and PGC‐1α, which serve as major hubs of protein–protein interaction networks for mitosis, apoptosis, differentiation, neuroprotection and energy homeostasis. This review focuses on necdin as a pleiotropic protein that integrates molecular interaction networks to promote neuronal vitality in modern placental mammals.
Collapse
|
89
|
Cheng H, Yang B, Ke T, Li S, Yang X, Aschner M, Chen P. Mechanisms of Metal-Induced Mitochondrial Dysfunction in Neurological Disorders. TOXICS 2021; 9:142. [PMID: 34204190 PMCID: PMC8235163 DOI: 10.3390/toxics9060142] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 01/31/2023]
Abstract
Metals are actively involved in multiple catalytic physiological activities. However, metal overload may result in neurotoxicity as it increases formation of reactive oxygen species (ROS) and elevates oxidative stress in the nervous system. Mitochondria are a key target of metal-induced toxicity, given their role in energy production. As the brain consumes a large amount of energy, mitochondrial dysfunction and the subsequent decrease in levels of ATP may significantly disrupt brain function, resulting in neuronal cell death and ensuing neurological disorders. Here, we address contemporary studies on metal-induced mitochondrial dysfunction and its impact on the nervous system.
Collapse
Affiliation(s)
- Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
| | - Bobo Yang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China;
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning 530021, China; (H.C.); (X.Y.)
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou 545006, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (B.Y.); (T.K.)
| |
Collapse
|
90
|
Stavsky A, Stoler O, Kostic M, Katoshevsky T, Assali EA, Savic I, Amitai Y, Prokisch H, Leiz S, Daumer-Haas C, Fleidervish I, Perocchi F, Gitler D, Sekler I. Aberrant activity of mitochondrial NCLX is linked to impaired synaptic transmission and is associated with mental retardation. Commun Biol 2021; 4:666. [PMID: 34079053 PMCID: PMC8172942 DOI: 10.1038/s42003-021-02114-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/22/2021] [Indexed: 02/04/2023] Open
Abstract
Calcium dynamics control synaptic transmission. Calcium triggers synaptic vesicle fusion, determines release probability, modulates vesicle recycling, participates in long-term plasticity and regulates cellular metabolism. Mitochondria, the main source of cellular energy, serve as calcium signaling hubs. Mitochondrial calcium transients are primarily determined by the balance between calcium influx, mediated by the mitochondrial calcium uniporter (MCU), and calcium efflux through the sodium/lithium/calcium exchanger (NCLX). We identified a human recessive missense SLC8B1 variant that impairs NCLX activity and is associated with severe mental retardation. On this basis, we examined the effect of deleting NCLX in mice on mitochondrial and synaptic calcium homeostasis, synaptic activity, and plasticity. Neuronal mitochondria exhibited basal calcium overload, membrane depolarization, and a reduction in the amplitude and rate of calcium influx and efflux. We observed smaller cytoplasmic calcium transients in the presynaptic terminals of NCLX-KO neurons, leading to a lower probability of release and weaker transmission. In agreement, synaptic facilitation in NCLX-KO hippocampal slices was enhanced. Importantly, deletion of NCLX abolished long term potentiation of Schaffer collateral synapses. Our results show that NCLX controls presynaptic calcium transients that are crucial for defining synaptic strength as well as short- and long-term plasticity, key elements of learning and memory processes.
Collapse
Affiliation(s)
- Alexandra Stavsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ohad Stoler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Marko Kostic
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tomer Katoshevsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Essam A Assali
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ivana Savic
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yael Amitai
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Holger Prokisch
- Institute of Human Genetics, School of Medicine, Technische Universität München, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Steffen Leiz
- Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany
| | | | - Ilya Fleidervish
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Fabiana Perocchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Daniel Gitler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
91
|
Mukem S, Thongbuakaew T, Khornchatri K. Mito-Tempo suppresses autophagic flux via the PI3K/Akt/mTOR signaling pathway in neuroblastoma SH-SY5Y cells. Heliyon 2021; 7:e07310. [PMID: 34195421 PMCID: PMC8239474 DOI: 10.1016/j.heliyon.2021.e07310] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 06/10/2021] [Indexed: 01/18/2023] Open
Abstract
The generation of excessive mitochondrial reactive oxygen species (mtROS) is associated with glutamate-stimulated neurotoxicity and pathogenesis of Alzheimer's disease (AD). Impaired mitochondrial function is accompanied with oxidative stress that is a significant contributor to initiate autophagy, but the underlying mechanisms are not fully understood. The present study aimed to investigate the neuroprotective effects of Mito-Tempo on glutamate-induced neuroblastoma SH-SY5Y cell toxicity. SH-SY5Y cells were treated with 100 μM glutamate in the presence or absence of 50 and 100 μM Mito-Tempo for 24 h. Changes in cell viability were measured by MTT assay. Cytotoxicity and intracellular ROS accumulation were also evaluated using lactate dehydrogenase (LDH) activity assay and 2,7-dichlorofluorescein diacetate (DCFDA) Reactive Oxygen Species Assay kit, respectively. Mitochondrial membrane potential was analyzed by tetraethylbenzimidazoly-lcarbocyanine iodide (JC-1) staining. Expression of PI3K/AKT/mTOR pathway and autophagy markers, including LC3 (LC3-I/-II) and p62 (SQSTM1) were performed using Western blot analysis. Our results demonstrated that glutamate-exposed cells significantly increased cellular oxidative stress by enhancing ROS production. Glutamate treatment also increased LDH release follows the loss of mitochondrial membrane potential, caused cell viability loss. Treatment with Mito-Tempo not only attenuated the generation of ROS and improved mitochondrial membrane potential but also reduced the neurotoxicity of glutamate in a concentration-dependent manner, which leads to increased cell viability and decreased LDH release. Mito-Tempo has a greater protective effect by enhancing superoxide dismutase (SOD) activity and PI3K/AKT/mTOR phosphorylation. Moreover, Mito-Tempo treatment altered the autophagy process resulting in the decline in the ratio of the autophagy markers LC3-I/-II and p62 (SQSTM1). We propose that Mito-Tempo can improve neuronal properties against glutamate cytotoxicity through its direct free radical scavenging activity and inhibit excessive autophagy signaling pathway, therefore, allow for further studies to investigate the therapeutic potentials of Mito-Tempo in animal disease models and human.
Collapse
Affiliation(s)
- Sirirak Mukem
- School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | | | - Kanjana Khornchatri
- Chulabhorn International College of Medicine, Thammasat University, Rangsit Campus, Pathumthani, 12120, Thailand
| |
Collapse
|
92
|
Kiper K, Freeman JL. Use of Zebrafish Genetic Models to Study Etiology of the Amyloid-Beta and Neurofibrillary Tangle Pathways in Alzheimer's Disease. Curr Neuropharmacol 2021; 20:524-539. [PMID: 34030617 DOI: 10.2174/1570159x19666210524155944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/09/2021] [Accepted: 05/16/2021] [Indexed: 11/22/2022] Open
Abstract
The prevalence of neurodegenerative diseases is increasing globally, with an imperative need to identify and expand the availability of pharmaceutical treatment strategies. Alzheimer's disease is the most common neurodegenerative disease for which there is no cure or has limited treatments. Rodent models are primarily used in Alzheimer's disease research to investigate causes, pathology, molecular mechanisms, and pharmaceutical therapies. However, there is a lack of a comprehensive understanding of Alzheimer's disease causes, pathogenesis, and optimal treatments due in part to some limitations of using rodents, including higher economic cost, which can influence sample size and ultimately statistical power. It is necessary to expand our animal model toolbox to provide alternative strategies in Alzheimer's disease research. The zebrafish application in neurodegenerative disease research and neuropharmacology is greatly expanding due to several vital strengths spanning lower economic costs, the smaller size of the organism, a sequenced characterized genome, and well described anatomical structures. These characteristics are coupled to the conserved molecular function and disease pathways in humans. The existence of orthologs for genes associated with Alzheimer's disease in zebrafish is also confirmed. While wild-type zebrafish appear to lack some of the neuropathological features of Alzheimer's disease, the advent of genetic editing technologies has expanded evaluation of the amyloid and neurofibrillary tangle hypotheses using the zebrafish and exploration of pharmaceutical molecular targets. An overview of how genetic editing technologies are being used with the zebrafish to create models to investigate the causes, pathology, molecular mechanisms, and pharmaceutical targets of Alzheimer's disease is detailed.
Collapse
Affiliation(s)
- Keturah Kiper
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States
| |
Collapse
|
93
|
Visual pH Sensors: From a Chemical Perspective to New Bioengineered Materials. Molecules 2021; 26:molecules26102952. [PMID: 34065629 PMCID: PMC8156760 DOI: 10.3390/molecules26102952] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 02/05/2023] Open
Abstract
Many human activities and cellular functions depend upon precise pH values, and pH monitoring is considered a fundamental task. Colorimetric and fluorescence sensors for pH measurements are chemical and biochemical tools able to sense protons and produce a visible signal. These pH sensors are gaining widespread attention as non-destructive tools, visible to the human eye, that are capable of a real-time and in-situ response. Optical “visual” sensors are expanding researchers’ interests in many chemical contexts and are routinely used for biological, environmental, and medical applications. In this review we provide an overview of trending colorimetric, fluorescent, or dual-mode responsive visual pH sensors. These sensors include molecular synthetic organic sensors, metal organic frameworks (MOF), engineered sensing nanomaterials, and bioengineered sensors. We review different typological chemical entities of visual pH sensors, three-dimensional structures, and signaling mechanisms for pH sensing and applications; developed in the past five years. The progression of this review from simple organic molecules to biological macromolecules seeks to benefit beginners and scientists embarking on a project of pH sensing development, who needs background information and a quick update on advances in the field. Lessons learned from these tools will aid pH determination projects and provide new ways of thinking for cell bioimaging or other cutting-edge in vivo applications.
Collapse
|
94
|
Witt B, Stiboller M, Raschke S, Friese S, Ebert F, Schwerdtle T. Characterizing effects of excess copper levels in a human astrocytic cell line with focus on oxidative stress markers. J Trace Elem Med Biol 2021; 65:126711. [PMID: 33486291 DOI: 10.1016/j.jtemb.2021.126711] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/02/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Being an essential trace element, copper is involved in diverse physiological processes. However, excess levels might lead to adverse effects. Disrupted copper homeostasis, particularly in the brain, has been associated with human diseases including the neurodegenerative disorders Wilson and Alzheimer's disease. In this context, astrocytes play an important role in the regulation of the copper homeostasis in the brain and likely in the prevention against neuronal toxicity, consequently pointing them out as a potential target for the neurotoxicity of copper. Major toxic mechanisms are discussed to be directed against mitochondria probably via oxidative stress. However, the toxic potential and mode of action of copper in astrocytes is poorly understood, so far. METHODS In this study, excess copper levels affecting human astrocytic cell model and their involvement in the neurotoxic mode of action of copper, as well as, effects on the homeostasis of other trace elements (Mn, Fe, Ca and Mg) were investigated. RESULTS Copper induced substantial cytotoxic effects in the human astrocytic cell line following 48 h incubation (EC30: 250 μM) and affected mitochondrial function, as observed via reduction of mitochondrial membrane potential and increased ROS production, likely originating from mitochondria. Moreover, cellular GSH metabolism was altered as well. Interestingly, not only cellular copper levels were affected, but also the homeostasis of other elements (Ca, Fe and Mn) were disrupted. CONCLUSION One potential toxic mode of action of copper seems to be effects on the mitochondria along with induction of oxidative stress in the human astrocytic cell model. Moreover, excess copper levels seem to interact with the homeostasis of other essential elements such as Ca, Fe and Mn. Disrupted element homeostasis might also contribute to the induction of oxidative stress, likely involved in the onset and progression of neurodegenerative disorders. These insights in the toxic mechanisms will help to develop ideas and approaches for therapeutic strategies against copper-mediated diseases.
Collapse
Affiliation(s)
- Barbara Witt
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| | - Michael Stiboller
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Stefanie Raschke
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Sharleen Friese
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Franziska Ebert
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany; German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589, Berlin, Germany
| |
Collapse
|
95
|
Belosludtsev KN, Sharipov RR, Boyarkin DP, Belosludtseva NV, Dubinin MV, Krasilnikova IA, Bakaeva ZV, Zgodova AE, Pinelis VG, Surin AM. The effect of DS16570511, a new inhibitor of mitochondrial calcium uniporter, on calcium homeostasis, metabolism, and functional state of cultured cortical neurons and isolated brain mitochondria. Biochim Biophys Acta Gen Subj 2021; 1865:129847. [PMID: 33453305 DOI: 10.1016/j.bbagen.2021.129847] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/20/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Disorders of mitochondrial Ca2+ homeostasis play a key role in the glutamate excitotoxicity of brain neurons. DS16570511 (DS) is a new penetrating inhibitor of mitochondrial Ca2+ uniporter complex (MCUC). The paper examines the effects of DS on the cultivated cortical neurons and isolated mitochondria of the rat brain. METHODS The functions of neurons and mitochondria were examined using fluorescence microscopy, XF24 microplate-based сell respirometry, ion-selective microelectrodes, spectrophotometry, and polarographic technique. RESULTS At the doses of 30 and 45 μM, DS reliably slowed down the onset of glutamate-induced delayed calcium deregulation of neurons and suppressed their death. 30 μM DS caused hyperpolarization of mitochondria of resting neurons, and 45 μM DS temporarily depolarized neuronal mitochondria. It was also demonstrated that 30-60 μM DS stimulated cellular respiration. DS was shown to suppress Ca2+ uptake by isolated brain mitochondria. In addition, DS inhibited ADP-stimulated mitochondrial respiration and ADP-induced decrease in the mitochondrial membrane potential. It was found that DS inhibited the activity of complex II of the respiratory chain. In the presence of Ca2+, high DS concentrations caused a collapse of the mitochondrial membrane potential. CONCLUSIONS The data obtained indicate that, in addition to the inhibition of MCUC, DS affects the main energy-transducing functions of mitochondria. GENERAL SIGNIFICANCE The using DS as a tool for studying MCUC and its functional role in neuronal cells should be done with care, bearing in mind multiple effects of DS, a proper evaluation of which would require multivariate analysis.
Collapse
Affiliation(s)
- Konstantin N Belosludtsev
- Mari State University, pl. Lenina 1, Yoshkar-Ola, 424001, Russia; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290, Russia.
| | - Rinat R Sharipov
- Institute of General Pathology and Pathophysiology, Baltiyskaya st. 8, Moscow, 125315, Russia
| | - Dmitry P Boyarkin
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Natalia V Belosludtseva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290, Russia
| | | | - Irina A Krasilnikova
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Zanda V Bakaeva
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Arina E Zgodova
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Vsevolod G Pinelis
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Alexander M Surin
- Institute of General Pathology and Pathophysiology, Baltiyskaya st. 8, Moscow, 125315, Russia; National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| |
Collapse
|
96
|
Valencia M, Kim SR, Jang Y, Lee SH. Neuronal Autophagy: Characteristic Features and Roles in Neuronal Pathophysiology. Biomol Ther (Seoul) 2021; 29:605-614. [PMID: 33875624 PMCID: PMC8551733 DOI: 10.4062/biomolther.2021.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/02/2021] [Accepted: 03/23/2021] [Indexed: 11/12/2022] Open
Abstract
Autophagy is an important degradative pathway that eliminates misfolded proteins and damaged organelles from cells. Autophagy is crucial for neuronal homeostasis and function. A lack of or deficiency in autophagy leads to the accumulation of protein aggregates, which are associated with several neurodegenerative diseases. Compared with non-neuronal cells, neurons exhibit rapid autophagic flux because damaged organelles or protein aggregates cannot be diluted in post-mitotic cells; because of this, these cells exhibit characteristic features of autophagy, such as compartment-specific autophagy, which depends on polarized structures and rapid autophagy flux. In addition, neurons exhibit compartment-specific autophagy, which depends on polarized structures. Neuronal autophagy may have additional physiological roles other than amino acid recycling. In this review, we focus on the characteristics and regulatory factors of neuronal autophagy. We also describe intracellular selective autophagy in neurons and its association with neurodegenerative diseases.
Collapse
Affiliation(s)
- McNeil Valencia
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sung Rae Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeseul Jang
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
97
|
Han S, Zhang M, Jeong YY, Margolis DJ, Cai Q. The role of mitophagy in the regulation of mitochondrial energetic status in neurons. Autophagy 2021; 17:4182-4201. [PMID: 33757395 DOI: 10.1080/15548627.2021.1907167] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are the main cellular energy powerhouses and supply most of the energy in the form of ATP to fuel essential neuronal functions through oxidative phosphorylation (OXPHOS). In Alzheimer disease (AD), metabolic and mitochondrial disruptions are an early feature preceding any histopathological and clinical manifestations. Mitochondrial malfunction is also linked to synaptic defects in early AD. Mitophagy serves as a key cellular quality control mechanism involving sequestration of damaged mitochondria within autophagosomes and their subsequent degradation in lysosomes. However, it remains largely unknown whether mitophagy is involved in the regulation of energy metabolism in neurons, and if so, whether metabolic deficiency in AD is attributed to mitophagy dysfunction. Here we reveal that mitophagy is broadly activated in metabolically enhanced neurons upon OXPHOS stimulation, which sustains high energetic activity by increasing mitochondrial turnover and hence facilitating mitochondrial maintenance. Unexpectedly, in AD-related mutant HsAPP Tg mouse brains, early stimulation of OXPHOS activity fails to correct energy deficits but exacerbates synapse loss as a consequence of mitophagy failure. Excitingly, lysosomal enhancement in AD neurons restores impaired metabolic function by promoting elimination of damaged mitochondria, protecting against synaptic damage in AD mouse brains. Taken together, we propose a new mechanism by which mitophagy controls bioenergetic status in neurons, furthering our understanding of the direct impact of mitophagy defects on AD-linked metabolic deficits and shedding light on the development of novel therapeutic strategies to treat AD by the early stimulation of mitochondrial metabolism combined with elevation of lysosomal proteolytic activity.
Collapse
Affiliation(s)
- Sinsuk Han
- Department of Cell Biology and Neuroscience, Division of Life Science, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Mingyang Zhang
- Department of Cell Biology and Neuroscience, Division of Life Science, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Yu Young Jeong
- Department of Cell Biology and Neuroscience, Division of Life Science, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - David J Margolis
- Department of Cell Biology and Neuroscience, Division of Life Science, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Division of Life Science, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
98
|
Brain Insulin Resistance: Focus on Insulin Receptor-Mitochondria Interactions. Life (Basel) 2021; 11:life11030262. [PMID: 33810179 PMCID: PMC8005009 DOI: 10.3390/life11030262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Current hypotheses implicate insulin resistance of the brain as a pathogenic factor in the development of Alzheimer’s disease and other dementias, Parkinson’s disease, type 2 diabetes, obesity, major depression, and traumatic brain injury. A variety of genetic, developmental, and metabolic abnormalities that lead to disturbances in the insulin receptor signal transduction may underlie insulin resistance. Insulin receptor substrate proteins are generally considered to be the node in the insulin signaling system that is critically involved in the development of insulin insensitivity during metabolic stress, hyperinsulinemia, and inflammation. Emerging evidence suggests that lower activation of the insulin receptor (IR) is another common, while less discussed, mechanism of insulin resistance in the brain. This review aims to discuss causes behind the diminished activation of IR in neurons, with a focus on the functional relationship between mitochondria and IR during early insulin signaling and the related roles of oxidative stress, mitochondrial hypometabolism, and glutamate excitotoxicity in the development of IR insensitivity to insulin.
Collapse
|
99
|
Amyotrophic Lateral Sclerosis and Frontotemporal Lobar Degenerations: Similarities in Genetic Background. Diagnostics (Basel) 2021; 11:diagnostics11030509. [PMID: 33805659 PMCID: PMC7998502 DOI: 10.3390/diagnostics11030509] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/06/2021] [Accepted: 03/11/2021] [Indexed: 12/27/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating, uniformly lethal progressive degenerative disorder of motor neurons that overlaps with frontotemporal lobar degeneration (FTLD) clinically, morphologically, and genetically. Although many distinct mutations in various genes are known to cause amyotrophic lateral sclerosis, it remains poorly understood how they selectively impact motor neuron biology and whether they converge on common pathways to cause neuronal degeneration. Many of the gene mutations are in proteins that share similar functions. They can be grouped into those associated with cell axon dynamics and those associated with cellular phagocytic machinery, namely protein aggregation and metabolism, apoptosis, and intracellular nucleic acid transport. Analysis of pathways implicated by mutant ALS genes has provided new insights into the pathogenesis of both familial forms of ALS (fALS) and sporadic forms (sALS), although, regrettably, this has not yet yielded definitive treatments. Many genes play an important role, with TARDBP, SQSTM1, VCP, FUS, TBK1, CHCHD10, and most importantly, C9orf72 being critical genetic players in these neurological disorders. In this mini-review, we will focus on the molecular mechanisms of these two diseases.
Collapse
|
100
|
Li M, Guo J, Wang H, Li Y. Involvement of Mitochondrial Dynamics and Mitophagy in Sevoflurane-Induced Cell Toxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6685468. [PMID: 33728028 PMCID: PMC7937461 DOI: 10.1155/2021/6685468] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
General anesthesia is a powerful and indispensable tool to ensure the accomplishment of surgical procedures or clinical examinations. Sevoflurane as an inhalational anesthetic without unpleasant odor is commonly used in clinical practice, especially for pediatric surgery. However, the toxicity caused by sevoflurane has gained growing attention. Mitochondria play a key role in maintaining cellular metabolism and survival. To maintain the stability of mitochondrial homeostasis, they are constantly going through fusion and fission. Also, damaged mitochondria need to be degraded by autophagy, termed as mitophagy. Accumulating evidence proves that sevoflurane exposure in young age could lead to cell toxicity by triggering the mitochondrial pathway of apoptosis, inducing the abnormalities of mitochondrial dynamics and mitophagy. In the present review, we focus on the current understanding of mitochondrial apoptosis, dynamics and mitophagy in cell function, the implications for cell toxicity in response to sevoflurane, and their underlying potential mechanisms.
Collapse
Affiliation(s)
- Ming Li
- School of Basic Medical Sciences, Hebei University, Baoding, Hebei Province, China
| | - Jiguang Guo
- School of Basic Medical Sciences, Hebei University, Baoding, Hebei Province, China
| | - Hongjie Wang
- School of Basic Medical Sciences, Hebei University, Baoding, Hebei Province, China
- Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Yuzhen Li
- Department of Pathophysiology, Graduate School of PLA General Hospital, Beijing, China
| |
Collapse
|