1
|
Hermann DM, Wang C, Mohamud Yusuf A, Herz J, Doeppner TR, Giebel B. Extracellular vesicles lay the ground for neuronal plasticity by restoring mitochondrial function, cell metabolism and immune balance. J Cereb Blood Flow Metab 2025:271678X251325039. [PMID: 40072028 PMCID: PMC11904928 DOI: 10.1177/0271678x251325039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/15/2025] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs) convey complex signals between cells that can be used to promote neuronal plasticity and neurological recovery in brain disease models. These EV signals are multimodal and context-dependent, making them unique therapeutic principles. This review analyzes how EVs released from various cell sources control neuronal metabolic function, neuronal survival and plasticity. Preferential sites of EV communication in the brain are interfaces between pre- and postsynaptic neurons at synapses, between astrocytes and neurons at plasma membranes or tripartite synapses, between oligodendrocytes and neurons at axons, between microglial cells/macrophages and neurons, and between cerebral microvascular cells and neurons. At each of these interfaces, EVs support mitochondrial function and cell metabolism under physiological conditions and orchestrate neuronal survival and plasticity in response to brain injury. In the injured brain, the promotion of neuronal survival and plasticity by EVs is tightly linked with EV actions on mitochondrial function, cell metabolism, oxidative stress and immune responses. Via the stabilization of cell metabolism and immune balance, neuronal plasticity responses are activated and functional neurological recovery is induced. As such, EV lay the ground for neuronal plasticity.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thorsten R Doeppner
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Department of Neurology, University Hospital Gießen and Marburg, Justus-Liebig-University Gießen, Gießen, Germany
| |
Collapse
|
2
|
Beltrán FA, Torres-Díaz L L, Troncoso-Escudero P, Villalobos-González J, Mayorga-Weber G, Lara M, Covarrubias-Pinto A, Valdivia S, Vicencio I, Papic E, Paredes-Martínez C, Silva-Januàrio ME, Rojas A, daSilva LLP, Court F, Rosas-Arellano A, Bátiz LF, Rojas P, Rivera FJ, Castro MA. Distinct roles of ascorbic acid in extracellular vesicles and free form: Implications for metabolism and oxidative stress in presymptomatic Huntington's disease. Free Radic Biol Med 2025; 227:521-535. [PMID: 39662690 DOI: 10.1016/j.freeradbiomed.2024.12.001] [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: 10/23/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin gene. The huntingtin protein (Htt) is ubiquitously expressed and localized in several organelles, including endosomes, where it plays an essential role in intracellular trafficking. Presymptomatic HD is associated with a failure in energy metabolism and oxidative stress. Ascorbic acid is a potent antioxidant that plays a key role in modulating neuronal metabolism and is highly concentrated in the brain. During synaptic activity, neurons take up ascorbic acid released by glial cells; however, this process is disrupted in HD. In this study, we aim to elucidate the molecular and cellular mechanisms underlying this dysfunction. Using an electrophysiological approach in presymptomatic YAC128 HD slices, we observed decreased ascorbic acid flux from astrocytes to neurons, which altered neuronal metabolic substrate preferences. Ascorbic acid efflux and recycling were also decreased in cultured astrocytes from YAC128 HD mice. We confirmed our findings using GFAP-HD160Q, an HD mice model expressing mutant N-terminal Htt mainly in astrocytes. For the first time, we demonstrated that ascorbic acid is released from astrocytes via extracellular vesicles (EVs). Decreased number of particles and exosomal markers were observed in EV fractions from cultured YAC128 HD astrocytes and Htt-KD cells. We observed reduced number of multivesicular bodies (MVBs) in YAC128 HD striatum via electron microscopy, suggesting mutant Htt alters MVB biogenesis. EVs containing ascorbic acid effectively reduced reactive oxygen species, whereas "free" ascorbic acid played a role in modulating neuronal metabolic substrate preferences. These findings suggest that the early redox imbalance observed in HD arises from a reduced release of ascorbic acid-containing EVs by astrocytes. Meanwhile, a decrease in "free" ascorbic acid likely contributes to presymptomatic metabolic impairment.
Collapse
Affiliation(s)
- Felipe A Beltrán
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Leandro Torres-Díaz L
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Departamento de Ciencias y Geografía, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Valparaíso, Chile
| | - Paulina Troncoso-Escudero
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Juan Villalobos-González
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Gonzalo Mayorga-Weber
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Marcelo Lara
- Departamento de Biología, Universidad de Santiago de Chile, Santiago, Chile; Escuela de Química y Farmacia, Facultad de Medicina y Ciencia, Universidad San Sebastián, Campus las Tres Pascualas, Concepción, Chile
| | - Adriana Covarrubias-Pinto
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Institute of Biochemistry II, Goethe University School of Medicine, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Sharin Valdivia
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Department of Biological and Chemical Sciences, Faculty of Medicine and Sciences, San Sebastián University, Tres Pascualas Campus, Concepción, Chile
| | - Isidora Vicencio
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Eduardo Papic
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Carolina Paredes-Martínez
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile
| | - Mara E Silva-Januàrio
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Alejandro Rojas
- Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Instituto de Medicina, UACh, Valdivia, Chile
| | - Luis L P daSilva
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Felipe Court
- Center for Aging Research and Healthy Longevity, Faculty of Sciences, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Luis Federico Bátiz
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de Los Andes, Santiago, Chile; Escuela de Medicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Patricio Rojas
- Departamento de Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Francisco J Rivera
- Translational Regenerative Neurobiology Group (TReN), Molecular and Integrative Biosciences Research Program (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, UACh, Valdivia, Chile; Center for Interdisciplinary Studies on Nervous System (CISNe), UACh, Valdivia, Chile; Janelia Research Campus HHMI, Ashburn, VA, USA.
| |
Collapse
|
3
|
Lu Y, Han L, Wang X, Liu X, Jia X, Lan K, Gao S, Feng Z, Yu L, Yang Q, Cui N, Wei YB, Liu JJ. Association between blood mitochondrial DNA copy number and mental disorders: A bidirectional two-sample mendelian randomization study. J Affect Disord 2024; 366:370-378. [PMID: 39197553 DOI: 10.1016/j.jad.2024.08.162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Mitochondria is essential for cellular energy production, oxidative stress, and apoptosis. Mitochondrial DNA (mtDNA) encodes essential proteins for mitochondrial function. Although several studies have explored the association between changes in mtDNA copy number (mtDNA-CN) and risk of mental disorders, the results remain debated. This study used a bidirectional two-sample Mendelian randomization (MR) analysis to examine the genetic causality between mtDNA-CN and mental disorders. METHODS Genome-wide association study (GWAS) data for mtDNA-CN were sourced from UK biobank, involving 383,476 European cases. GWAS data for seven mental disorders-attention deficit/hyperactivity disorder, autism spectrum disorder (ASD), schizophrenia, bipolar disorder, major depressive disorder, anxiety, and obsessive-compulsive disorder-were primarily obtained from the Psychiatric Genomics Consortium. Causal associations were assessed using inverse variance weighting, with sensitivity analyses via the weighted median and MR-Egger methods. Reverse MR considered the seven mental disorders as exposures. All analyses were replicated with additional mtDNA-CN GWAS data from 465,809 individuals in the Heart and Ageing Research in Genomic Epidemiology consortium and the UK Biobank. RESULTS Forward MR observed a 27 % decrease in the risk of ASD per standard deviation increase in genetically determined blood mtDNA-CN (OR = 0.73, 95%CI: 0.58-0.92, p = 0.002), with no causal effects on other disorders. Additionally, reverse MR did not indicate a causal association between any of the mental disorders and mtDNA-CN. Validation analyses corroborated these findings, indicating their robustness. CONCLUSIONS Our study supports the potential causal association between mtDNA-CN and the risk of ASD, suggesting that mtDNA-CN could serve as a promising biomarker for early screening of ASD.
Collapse
Affiliation(s)
- Yan'e Lu
- School of Nursing, Peking University, Beijing 100191, China
| | - Lei Han
- Beijing Key Laboratory of Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Xingxing Wang
- School of Nursing, Peking University, Beijing 100191, China
| | - Xiaotong Liu
- School of Nursing, Peking University, Beijing 100191, China
| | - Xinlei Jia
- School of Nursing, Peking University, Beijing 100191, China
| | - Kunyi Lan
- School of Nursing, Peking University, Beijing 100191, China
| | - Shumin Gao
- Beijing Key Laboratory of Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Zhendong Feng
- Beijing Key Laboratory of Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Lulu Yu
- Mental Health Center, the First Hospital of Hebei Medical University, Hebei Technical Innovation Center for Mental Health Assessment and Intervention, Shijiazhuang, Hebei Province 050031, China
| | - Qian Yang
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Naixue Cui
- School of Nursing and Rehabilitation, Shandong University, Shandong Province 250012, China
| | - Ya Bin Wei
- Beijing Key Laboratory of Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing 100191, China.
| | - Jia Jia Liu
- School of Nursing, Peking University, Beijing 100191, China.
| |
Collapse
|
4
|
Kong L, Liu Y, Li J, Wang Y, Ji P, Shi Q, Han M, Xu H, Li W, Li W. Ginsenoside Rg1 alleviates chronic inflammation-induced neuronal ferroptosis and cognitive impairments via regulation of AIM2 - Nrf2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118205. [PMID: 38641079 DOI: 10.1016/j.jep.2024.118205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng is a valuable herb in traditional Chinese medicine. Modern research has shown that it has various benefits, including tonifying vital energy, nourishing and strengthening the body, calming the mind, improving cognitive function, regulating fluids, and returning blood pressure, etc. Rg1 is a primary active component of ginseng. It protects hippocampal neurons, improves synaptic plasticity, enhances cognitive function, and boosts immunity. Furthermore, it exhibits anti-aging and anti-fatigue properties and holds great potential for preventing and managing neurodegenerative diseases (NDDs). AIM OF THE STUDY The objective of this study was to examine the role of Rg1 in treating chronic inflammatory NDDs and its molecular mechanisms. MATERIALS AND METHODS In vivo, we investigated the protective effects of Rg1 against chronic neuroinflammation and cognitive deficits in mice induced by 200 μg/kg lipopolysaccharide (LPS) for 21 days using behavioral tests, pathological sections, Western blot, qPCR and immunostaining. In vitro experiments involved the stimulation of HT22 cells with 10 μg/ml of LPS, verification of the therapeutic effect of Rg1, and elucidation of its potential mechanism of action using H2DCFDA staining, BODIPY™ 581/591 C11, JC-1 staining, Western blot, and immunostaining. RESULTS Firstly, it was found that Rg1 significantly improved chronic LPS-induced behavioral and cognitive dysfunction in mice. Further studies showed that Rg1 significantly attenuated LPS-induced neuronal damage by reducing levels of IL-6, IL-1β and ROS, and inhibiting AIM2 inflammasome. Furthermore, chronic LPS exposure induced the onset of neuronal ferroptosis by increasing the lipid peroxidation product MDA and regulating the ferroptosis-associated proteins Gpx4, xCT, FSP1, DMT1 and TfR, which were reversed by Rg1 treatment. Additionally, Rg1 was found to activate Nrf2 and its downstream antioxidant enzymes, such as HO1 and NQO1, both in vivo and in vitro. In vitro studies also showed that the Nrf2 inhibitor ML385 could inhibit the anti-inflammatory, antioxidant, and anti-ferroptosis effects of Rg1. CONCLUSIONS This study demonstrated that Rg1 administration ameliorated chronic LPS-induced cognitive deficits and neuronal ferroptosis in mice by inhibiting neuroinflammation and oxidative stress. The underlying mechanisms may be related to the inhibition of AIM2 inflammasome and activation of Nrf2 signaling. These findings provide valuable insights into the treatment of chronic neuroinflammation and associated NDDs.
Collapse
Affiliation(s)
- Liangliang Kong
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Yan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Jingwei Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Yanyan Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Pengmin Ji
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Qifeng Shi
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Min Han
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Hanyang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Weiping Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| | - Weizu Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
5
|
Liang Y, Li Y, Jiao Q, Wei M, Wang Y, Cui A, Li Z, Li G. Axonal mitophagy in retinal ganglion cells. Cell Commun Signal 2024; 22:382. [PMID: 39075570 PMCID: PMC11285280 DOI: 10.1186/s12964-024-01761-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/22/2024] [Indexed: 07/31/2024] Open
Abstract
Neurons, exhibiting unique polarized structures, rely primarily on the mitochondrial production of ATP to maintain their hypermetabolic energy requirements. To maintain a normal energy supply, mitochondria are transported to the distal end of the axon. When mitochondria within the axon are critically damaged beyond their compensatory capacity, they are cleared via autophagosomal phagocytosis, and the degradation products are recycled to replenish energy. When the mitochondria are dysfunctional or their transport processes are blocked, axons become susceptible to degeneration triggered by energy depletion, resulting in neurodegenerative diseases. As the final checkpoint for mitochondrial quality control, axonal mitophagy is vital for neuronal growth, development, injury, and regeneration. Furthermore, abnormal axonal mitophagy is crucial in the pathogenesis of optic nerve-related diseases such as glaucoma. We review recent studies on axonal mitophagy and summarize the progress of research on axonal mitophagy in optic nerve-related diseases to provide insights into diseases associated with axonal damage in optic ganglion cells.
Collapse
Affiliation(s)
- Yang Liang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Yulin Li
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Qing Jiao
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Muyang Wei
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Yan Wang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Aoteng Cui
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhihui Li
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Guangyu Li
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
6
|
Zhang X, Li G, Chen H, Nie XW, Bian JS. Targeting NKAα1 to treat Parkinson's disease through inhibition of mitophagy-dependent ferroptosis. Free Radic Biol Med 2024; 218:190-204. [PMID: 38574977 DOI: 10.1016/j.freeradbiomed.2024.04.002] [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: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Dysfunction of the Na+/K+-ATPase (NKA) has been documented in various neurodegenerative diseases, yet the specific role of NKAα1 in Parkinson's disease (PD) remains incompletely understood. In this investigation, we utilized NKAα1 haploinsufficiency (NKAα1+/-) mice to probe the influence of NKAα1 on dopaminergic (DA) neurodegeneration induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our findings reveal that NKAα1+/- mice displayed a heightened loss of DA neurons and more pronounced motor dysfunction compared to the control group when exposed to MPTP. Intriguingly, this phenomenon coincided with the activation of ferroptosis and impaired mitophagy both in vivo and in vitro. To scrutinize the role and underlying mechanism of NKAα1 in PD, we employed DR-Ab, an antibody targeting the DR-region of the NKA α subunit. Our study demonstrates that the administration of DR-Ab effectively reinstated the membrane abundance of NKAα1, thereby mitigating MPTP-induced DA neuron loss and subsequent improvement in behavioral deficit. Mechanistically, DR-Ab heightened the formation of the surface NKAα1/SLC7A11 complex, inhibiting SLC7A11-dependent ferroptosis. Moreover, DR-Ab disrupted the cytosolic interaction between NKAα1 and Parkin, facilitating the translocation of Parkin to mitochondria and enhancing the process of mitophagy. In conclusion, this study establishes NKAα1 as a key regulator of ferroptosis and mitophagy, identifying its DR-region as a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Guanghong Li
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Hanbin Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Xiao-Wei Nie
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (the First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518055, China.
| | - Jin-Song Bian
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
7
|
Herrera G, Silvero C MJ, Becerra MC, Lasaga M, Scimonelli T. Modulatory role of α-MSH in hippocampal-dependent memory impairment, synaptic plasticity changes, oxidative stress, and astrocyte reactivity induced by short-term high-fat diet intake. Neuropharmacology 2023; 239:109688. [PMID: 37591460 DOI: 10.1016/j.neuropharm.2023.109688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023]
Abstract
High-fat diet (HFD) consumption is associated with cognitive deficits and neurodegenerative diseases. Since the hippocampus is extremely sensitive to pathophysiological changes, neuroinflammation and the concomitant oxidative stress induced by HFD can significantly interfere with hippocampal-dependent functions related to learning and memory. The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) mediates neuroprotective actions in the central nervous system and can reverse the effects of neuroinflammation in cognitive functions that depend on the hippocampus. In this study, we used male Wistar rats to evaluate the effect of short-term HFD intake (5 days) plus a mild immune challenge, Lipopolysaccharide (LPS 10 μg/kg) on contextual fear, changes in structural plasticity, oxidative stress, and astrocyte reactivation in the hippocampus. We also determined the possible modulatory role of α-MSH. HFD consumption was associated with an increase in markers of oxidative stress (Advanced oxidation protein products and Malondialdehyde) in the dorsal hippocampus (DH). We also found changes in hippocampal structural synaptic plasticity, observing a decrease in total spine in the DH after HFD plus LPS. We observed astrocyte proliferation and a significant increase in the percentage of the area occupied by GFAP. Treatment with α-MSH (0.1 μg/0.25 μl) in the DH reversed the effect of short-term HFD plus LPS on contextual fear memory, oxidative stress, and spine density. α-MSH also reduced astrocyte proliferation. Our present results indicate that HFD consumption for a short period sensitizes the central nervous system (CNS) to a subsequent immune challenge and impairs contextual fear memory and that α-MSH could have a modulatory protective effect.
Collapse
Affiliation(s)
- Guadalupe Herrera
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - M Jazmín Silvero C
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M Cecilia Becerra
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica UNITEFA-CONICET. Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Teresa Scimonelli
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET. Departamento de Farmacología Otto Orshinger, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
8
|
Karoglu-Eravsar ET, Tuz-Sasik MU, Karaduman A, Keskus AG, Arslan-Ergul A, Konu O, Kafaligonul H, Adams MM. Long-Term Acetylcholinesterase Depletion Alters the Levels of Key Synaptic Proteins while Maintaining Neuronal Markers in the Aging Zebrafish (Danio rerio) Brain. Gerontology 2023; 69:1424-1436. [PMID: 37793352 PMCID: PMC10711754 DOI: 10.1159/000534343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023] Open
Abstract
INTRODUCTION Interventions targeting cholinergic neurotransmission like acetylcholinesterase (AChE) inhibition distinguish potential mechanisms to delay age-related impairments and attenuate deficits related to neurodegenerative diseases. However, the chronic effects of these interventions are not well described. METHODS In the current study, global levels of cholinergic, cellular, synaptic, and inflammation-mediating proteins were assessed within the context of aging and chronic reduction of AChE activity. Long-term depletion of AChE activity was induced by using a mutant zebrafish line, and they were compared with the wildtype group at young and old ages. RESULTS Results demonstrated that AChE activity was lower in both young and old mutants, and this decrease coincided with a reduction in ACh content. Additionally, an overall age-related reduction in AChE activity and the AChE/ACh ratio was observed, and this decline was more prominent in wildtype groups. The levels of an immature neuronal marker were upregulated in mutants, while a glial marker showed an overall reduction. Mutants had preserved levels of inhibitory and presynaptic elements with aging, whereas glutamate receptor subunit levels declined. CONCLUSION Long-term AChE activity depletion induces synaptic and cellular alterations. These data provide further insights into molecular targets and adaptive responses following the long-term reduction of AChE activity that was also targeted pharmacologically to treat neurodegenerative diseases in human subjects.
Collapse
Affiliation(s)
- Elif Tugce Karoglu-Eravsar
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology (UNAM), Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
- Department of Psychology, Selcuk University, Konya, Turkey
| | - Melek Umay Tuz-Sasik
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology (UNAM), Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
| | - Aysenur Karaduman
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Ayse Gokce Keskus
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology (UNAM), Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
| | - Ayca Arslan-Ergul
- National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology (UNAM), Bilkent University, Ankara, Turkey
| | - Ozlen Konu
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology (UNAM), Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Hulusi Kafaligonul
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology (UNAM), Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Michelle M. Adams
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
- National Nanotechnology Research Center and Institute of Materials Science and Nanotechnology (UNAM), Bilkent University, Ankara, Turkey
- Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
- Department of Psychology, Bilkent University, Ankara, Turkey
| |
Collapse
|
9
|
Al-Kafaji G, Jahrami HA, Alwehaidah MS, Alshammari Y, Husni M. Mitochondrial DNA copy number in autism spectrum disorder and attention deficit hyperactivity disorder: a systematic review and meta-analysis. Front Psychiatry 2023; 14:1196035. [PMID: 37484684 PMCID: PMC10361772 DOI: 10.3389/fpsyt.2023.1196035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Background Several reports suggest that altered mitochondrial DNA copy number (mtDNA-cn), a common biomarker for aberrant mitochondrial function, is implicated in autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), but the results are still elusive. Methods A meta-analysis was performed to summarize the current indication and to provide a more precise assessment of the mtDNA-cn in ASD and ADHD. A search in the MEDLINE-PubMed, Scopus, and EMBASE databases was done to identify related studies up to the end of February 2023. The meta-analysis was conducted according to recommendations of the Cochrane Handbook of Systematic Reviews. Results Fourteen studies involving 666 cases with ASD and ADHD and 585 controls were collected and judged relevant for the systematic review and meta-analysis. The pooled results by a random effects meta-analysis was reported as a geometric mean of the estimated average response ratio and 95% confidence interval. Overall analysis of studies reported differences in mtDNA-cn in blood samples (k = 10) and non-blood samples (brain tissues and oral samples; k = 4) suggested significantly higher mtDNA-cn in patients compared to controls (p = 0.0275). Sub-analysis by stratifying studies based on tissue type, showed no significant increase in mtDNA-cn in blood samples among patients and controls (p = 0.284). Conversely, higher mtDNA-cn was observed in non-blood samples in patients than in controls (p = 0.0122). Further stratified analysis based on blood-cell compositions as potential confounds showed no significant difference in mtDNA-cn in peripheral blood samples of patients comparted to controls (p = 0.074). In addition, stratified analysis of aged-matched ASD and ADHD patients and controls revealed no significant difference in mtDNA-cn in blood samples between patients and controls (p = 0.214), whereas a significant increase in mtDNA-cn was observed in non-blood samples between patients and controls (p < 0.001). Finally, when the mtDNA-cn was analyzed in blood samples of aged-matched patients with ASD (peripheral blood, leukocytes, and PBMCs) or ADHD (peripheral blood), no significant difference in mtDNA-cn was observed between ASD patients and controls (p = 0.385), while a significant increase in mtDNA-cn was found between ADHD patients and controls (p = 0.033). Conclusion In this first meta-analysis of the evaluation of mtDNA-cn in ASD/ADHD, our results show elevated mtDNA-cn in ASD and ADHD, further emphasizing the implication of mitochondrial dysfunction in neurodevelopmental disorders. However, our results indicate that the mtDNA-cn in blood is not reflected in other tissues in ASD/ADHD, and the true relationship between blood-derived mtDNA-cn and ASD/ADHD remains to be defined in future studies. The importance of blood-cell compositions as confounders of blood-based mtDNA-cn measurement and the advantages of salivary mtDNA-cn should be considered in future studies. Moreover, the potential of mtDNA-cn as a biomarker for mitochondrial malfunction in neurodevelopmental disorders deserves further investigations.
Collapse
Affiliation(s)
- Ghada Al-Kafaji
- Department of Molecular Medicine and Al-Jawhara Centre for Molecular Medicine, Genetics, and Inherited Disorders, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Haitham Ali Jahrami
- Department of Psychiatry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
- Government Hospital, Manama, Bahrain
| | - Materah Salem Alwehaidah
- Department of Medical Laboratory, Faculty of Allied Health, Kuwait University, Kuwait City, Kuwait
| | | | - Mariwan Husni
- Department of Psychiatry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
- Department of Psychiatry, Northern Ontarion School of Medicine University, Thunder Bay, ON, Canada
| |
Collapse
|
10
|
Kong L, Sun R, Zhou H, Shi Q, Liu Y, Han M, Li W, Qun S, Li W. Trpc6 knockout improves behavioral dysfunction and reduces Aβ production by inhibiting CN-NFAT1 signaling in T2DM mice. Exp Neurol 2023; 363:114350. [PMID: 36791875 DOI: 10.1016/j.expneurol.2023.114350] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/17/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023]
Abstract
As the prevalence of diabetes and health awareness increase, type 2 diabetes mellitus -associated cognitive dysfunction is receiving increasing attention. However, the pathogenesis is not entirely understood. Transient receptor potential cation channel 6 (TRPC6) is highly correlated with intracellular Ca2+ concentrations, and neuronal calcium overload is an important cause of cognitive dysfunction. In the present study, we investigated the effect and mechanism of Trpc6 knockout in high-fat diet and streptozotocin-induced T2DM mice. The body weight and fasting blood glucose were recorded during the experiment. Behavioral dysfunction was detected using the open field test (OFT), elevated plus maze (EPM), hole-board test (HBT), Morris water maze (MWM) test and contextual fear conditioning (CFC) test. Nissl and H&E staining were used to examine neuronal damage. Western blot, quantitative real-time polymerase chain reaction (q-PCR), and immunofluorescence were performed to detect amyloid beta protein (Aβ) deposition and related indicators of neurological impairments in the cerebral cortex and hippocampus. The results indicated that Trpc6 knockout inhibited body weight loss and fasting blood glucose increase, improved spontaneous activity, learning and memory dysfunction, and alleviated neuroinflammation and neuronal damage in T2DM mice. The further results demonstrated that Trpc6 knockout decreased Aβ generation and deposition, and reduced the expressions of inflammasome-related proteins in T2DM mice. In addition, Trpc6 knockout inhibited intracellular calcium overload in diabetic mice and primary cultured hippocampal neurons, which in turn suppressed CN and NFAT1 expression. These data suggest that Trpc6 knockout may inhibit the CN-NFAT1 signaling pathway by decreasing intracellular calcium overload in the brain of T2DM mice, which consequently reduce Aβ deposition and neuroinflammation, and ultimately delay the development of T2DM-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Liangliang Kong
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Ran Sun
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Huimsin Zhou
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Qifeng Shi
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Yan Liu
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Min Han
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Weiping Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China
| | - Sen Qun
- Stroke Center & Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
11
|
Cross-talk between energy and redox metabolism in astrocyte-neuron functional cooperation. Essays Biochem 2023; 67:17-26. [PMID: 36805653 PMCID: PMC10011404 DOI: 10.1042/ebc20220075] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023]
Abstract
Astrocytes show unique anatomical, morphological, and metabolic features to take up substrates from the blood and metabolize them for local delivery to active synapses to sustain neuron function. In the present review, we specifically focus on key molecular aspects of energy and redox metabolism that facilitate this astrocyte-neuronal coupling in a controlled manner. Basal glycolysis is co-ordinated by the anaphase-promoting complex/cyclosome (APC/C)-Cdh1, a ubiquitin ligase that targets the proglycolytic enzyme 6-phosphofructokinase-2,6-bisphosphastate-3 (PFKFB3) for degradation. APC/C-Cdh1 activity is more robust in neurons than in astrocytes, which determine that PFKFB3 abundance and glycolytic rate are weaker in neurons. The low PFKFB3 activity in neurons facilitates glucose-6-phosphate oxidation via the pentose-phosphate pathway, which promotes antioxidant protection. Conversely, the high PFKFB3 activity in astrocytes allows the production and release of glycolytic lactate, which is taken up by neurons that use it as an oxidizable substrate. Importantly, the mitochondrial respiratory chain is tighter assembled in neurons than in astrocytes, thus the bioenergetic efficiency of mitochondria is higher in neurons. Because of this, the production of reactive oxygen species (mROS) by mitochondrial complex I is very low in neurons and very high in astrocytes. Such a naturally occurring high abundance of mROS in astrocytes physiologically determines a specific transcriptional fingerprint that contributes to sustaining cognitive performance. We conclude that the energy and redox metabolism of astrocytes must complementarily match that of neurons to regulate brain function and animal welfare.
Collapse
|
12
|
TREM2 and Microglia Contribute to the Synaptic Plasticity: from Physiology to Pathology. Mol Neurobiol 2023; 60:512-523. [PMID: 36318443 DOI: 10.1007/s12035-022-03100-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022]
Abstract
Synapses are bridges for information transmission in the central nervous system (CNS), and synaptic plasticity is fundamental for the normal function of synapses, contributing substantially to learning and memory. Numerous studies have proven that microglia can participate in the occurrence and progression of neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD), by regulating synaptic plasticity. In this review, we summarize the main characteristics of synapses and synaptic plasticity under physiological and pathological conditions. We elaborate the origin and development of microglia and the two well-known microglial signaling pathways that regulate synaptic plasticity. We also highlight the unique role of triggering receptor expressed on myeloid cells 2 (TREM2) in microglia-mediated regulation of synaptic plasticity and its relationship with AD. Finally, we propose four possible ways in which TREM2 is involved in regulating synaptic plasticity. This review will help researchers understand how NDDs develop from the perspective of synaptic plasticity.
Collapse
|
13
|
Guo H, Yi J, Wang F, Lei T, Du H. Potential application of heat shock proteins as therapeutic targets in Parkinson's disease. Neurochem Int 2023; 162:105453. [PMID: 36402293 DOI: 10.1016/j.neuint.2022.105453] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/08/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
Parkinson's disease (PD) is a common chronic neurodegenerative disease, and the heat shock proteins (HSPs) are proved to be of great value for PD. In addition, HSPs can maintain protein homeostasis, degrade and inhibit protein aggregation by properly folding and activating intracellular proteins in PD. This study mainly summarizes the important roles of HSPs in PD and explores their feasibility as targets. We introduced the structural and functional characteristics of HSPs and the physiological functions of HSPs in PD. HSPs can protect neurons from damage by degrading aggregates with three mechanisms, including the aggregation and removing α-Synuclein (α-Syn) aggregates, promotion the autophagy of abnormal proteins, and inhibition the apoptosis of degenerated neurons. This study underscores the importance of HSPs as targets in PD and helps to expand new mechanisms in PD treatment strategies.
Collapse
Affiliation(s)
- Haodong Guo
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Jingsong Yi
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Fan Wang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, 100083, China
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing, 100083, China.
| |
Collapse
|
14
|
Billard JM, Freret T. Improved NMDA Receptor Activation by the Secreted Amyloid-Protein Precursor-α in Healthy Aging: A Role for D-Serine? Int J Mol Sci 2022; 23:ijms232415542. [PMID: 36555191 PMCID: PMC9779005 DOI: 10.3390/ijms232415542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Impaired activation of the N-methyl-D-aspartate subtype of glutamate receptors (NMDAR) by D-serine is linked to cognitive aging. Whether this deregulation may be used to initiate pharmacological strategies has yet to be considered. To this end, we performed electrophysiological extracellular recordings at CA3/CA1 synapses in hippocampal slices from young and aged mice. We show that 0.1 nM of the soluble N-terminal recombinant fragment of the secreted amyloid-protein precursor-α (sAPPα) added in the bath significantly increased NMDAR activation in aged but not adult mice without impacting basal synaptic transmission. In addition, sAPPα rescued the age-related deficit of theta-burst-induced long-term potentiation. Significant NMDAR improvement occurred in adult mice when sAPPα was raised to 1 nM, and this effect was drastically reduced in transgenic mice deprived of D-serine through genetic deletion of the synthesizing enzyme serine racemase. Altogether, these results emphasize the interest to consider sAPPα treatment targeting D-serine-dependent NMDAR deregulation to alleviate cognitive aging.
Collapse
|
15
|
Wang H, Fu M, Ma Y, Liu C, Wu M, Nie J. Tobacco smoke exposure and mitochondrial DNA copy number on neurobehavioural performance: A community study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:84180-84190. [PMID: 35776305 DOI: 10.1007/s11356-022-20921-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
The influence of tobacco smoke has been a controversial and very questionable subject within the field of neurological behaviours. To examine the dose-response relationships between tobacco smoke and neurological performance, we investigated whether mitochondrial DNA copy number (mtDNAcn) mediates these relationships. We used restricted cubic spline models to estimate the dose-response relationships. A mediation model was also used to detect the mediating effect. Increased cotinine was negatively associated with auditory memory scores and a 0.51 decrease in mtDNAcn. MtDNAcn acts as a mediator between cotinine and auditory memory. Tobacco smoke levels were inversely associated with mtDNAcn and neurobehavioural changes, and there was a mediation effect between cotinine levels and auditory memory by mtDNAcn.
Collapse
Affiliation(s)
- Huimin Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Mengmeng Fu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yifei Ma
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chenjuan Liu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Min Wu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jisheng Nie
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
16
|
Emerging roles of brain metabolism in cognitive impairment and neuropsychiatric disorders. Neurosci Biobehav Rev 2022; 142:104892. [PMID: 36181925 DOI: 10.1016/j.neubiorev.2022.104892] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022]
Abstract
Here we discuss the role of diverse environmental manipulations affecting cognition with special regard to psychiatric conditions. We present evidence supporting a direct causal correlation between the valence of the environmental stimulation and some psychopathological traits and how the environment influences brain structure and function with special regard to oxidative stress and mitochondrial activity. Increasing experimental evidence supports a role for mitochondrial dysfunctions in neuropsychiatric disorders. Brain mitochondria are considered crucial mediators of allostasis, that is the capability to adapt to stress via a complex interaction between the autonomic, metabolic, and immune systems to maintain cellular homeostasis. In this process, mitochondria act as highly dynamic integrators by sensing and transducing stressors into adaptation mechanisms via metabolic stress mediators, such as glucocorticoids and catecholamines. Alterations in cellular homeostasis induced by chronic stress are thought to predispose to disease by triggering the so-called "mitochondrial allostatic load". This process is characterized by functional and structural changes of the mitochondria, ultimately leading to oxidative stress, inflammation, mitochondrial DNA damage and apoptosis. In this review we discuss the role of diverse environmental manipulations to affect cognition with special regard to psychiatric conditions. How the environment influences brain structure and function, and the interactions between rearing conditions, oxidative stress and mitochondrial activity are fundamental questions that are still poorly understood. As will be discussed, increasing experimental evidence supports a role for mitochondrial dysfunctions in neuropsychiatric disorders. Brain mitochondria are considered crucial mediators of allostasis, that is the capability to adapt to stress via a complex interaction between the autonomic, metabolic, and immune systems to maintain cellular homeostasis. In this process, mitochondria act as highly dynamic integrators by sensing and transducing stressors into adaptation mechanisms via metabolic stress mediators, such as glucocorticoids and catecholamines. Alterations in cellular homeostasis induced by chronic stress are thought to predispose to disease by triggering the so-called "mitochondrial allostatic load". This process is characterized by functional and structural changes of the mitochondria, ultimately leading to oxidative stress, inflammation, mitochondrial DNA damage and apoptosis. The brain requires considerable mitochondrial reserve not only to sustain basal neuronal needs but a also to provide increasing energy demands during stress. Consistently with these high energetic requirements, it is reasonable to hypothesise that the brain is particularly vulnerable to mitochondrial defects. Thus, even subtle metabolic alterations might have a substantial impact on cognitive functions. Over the last decade, several experimental evidence supported the hypothesis that a suboptimal mitochondrial function, which could be of genetic origin or acquired following adverse life events, is a key vulnerability factor for stress-related psychopathologies. Chronic psychological stress is a major promoter of anxiety as well as of oxidative damage, as shown in several studies. Recent evidence from mouse models harbouring mutations in mitochondrial genes demonstrated the role of mitochondria in modulating the response to acute psychological stress. However, it has yet to be determined whether mitochondrial dysfunctions are the cause or the consequence of anxiety. In this review, we discuss how adverse psychosocial environments can impact mitochondrial bioenergetics at the molecular level and we gather evidence from several studies linking energy metabolism and stress resilience/vulnerability. Moreover, we review recent findings supporting that metabolic dysfunction can underlie deficits in complex social behaviours. As will be discussed, aberrations in mitochondrial functionality have been found in the nucleus accumbens of highly anxious mice and mediate low social competitiveness. In addition, alterations in sociability can be reversed by enhancing mitochondrial functions. Recent evidence also demonstrated that a specific mutation in mitochondrial DNA, previously linked to autism spectrum disorder, produces autistic endophenotypes in mice by altering respiration chain and reactive oxygen species (ROS) production. Finally, we discuss a "Negative Enrichment" model that can explain some of the psychopathological conditions relevant to humans. Evidence of a direct causal correlation of valence of environmental stimulation and psychopathological traits will be presented, and possible molecular mechanisms that focus on oxidative stress. Collectively, the findings described here have been achieved with a wide set of behavioural and cognitive tasks with translational validity. Thus, they will be useful for future work aimed to elucidate the fine metabolic alterations in psychopathologies and devise novel approaches targeting mitochondria to alleviate these conditions.
Collapse
|
17
|
Mhatre SD, Iyer J, Petereit J, Dolling-Boreham RM, Tyryshkina A, Paul AM, Gilbert R, Jensen M, Woolsey RJ, Anand S, Sowa MB, Quilici DR, Costes SV, Girirajan S, Bhattacharya S. Artificial gravity partially protects space-induced neurological deficits in Drosophila melanogaster. Cell Rep 2022; 40:111279. [PMID: 36070701 PMCID: PMC10503492 DOI: 10.1016/j.celrep.2022.111279] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 03/16/2022] [Accepted: 08/05/2022] [Indexed: 02/03/2023] Open
Abstract
Spaceflight poses risks to the central nervous system (CNS), and understanding neurological responses is important for future missions. We report CNS changes in Drosophila aboard the International Space Station in response to spaceflight microgravity (SFμg) and artificially simulated Earth gravity (SF1g) via inflight centrifugation as a countermeasure. While inflight behavioral analyses of SFμg exhibit increased activity, postflight analysis displays significant climbing defects, highlighting the sensitivity of behavior to altered gravity. Multi-omics analysis shows alterations in metabolic, oxidative stress and synaptic transmission pathways in both SFμg and SF1g; however, neurological changes immediately postflight, including neuronal loss, glial cell count alterations, oxidative damage, and apoptosis, are seen only in SFμg. Additionally, progressive neuronal loss and a glial phenotype in SF1g and SFμg brains, with pronounced phenotypes in SFμg, are seen upon acclimation to Earth conditions. Overall, our results indicate that artificial gravity partially protects the CNS from the adverse effects of spaceflight.
Collapse
Affiliation(s)
- Siddhita D Mhatre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA; COSMIAC Research Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Janani Iyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA; Universities Space Research Association, Mountain View, CA 94043, USA
| | - Juli Petereit
- Nevada Bioinformatics Center, University of Nevada, Reno, NV 89557, USA
| | - Roberta M Dolling-Boreham
- Department of Electrical and Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; Blue Marble Space Institute of Science, Seattle, WA 94035, USA
| | - Anastasia Tyryshkina
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Universities Space Research Association, Mountain View, CA 94043, USA; Blue Marble Space Institute of Science, Seattle, WA 94035, USA; NASA Postdoctoral Program, Universities Space Research Association, NASA Ames Research Center, Moffett Field, CA 94035, USA; Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL 32114, USA
| | - Rachel Gilbert
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; NASA Postdoctoral Program, Universities Space Research Association, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Matthew Jensen
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | | | - Sulekha Anand
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| | - Marianne B Sowa
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - David R Quilici
- Nevada Proteomics Center, University of Nevada, Reno, NV 89557, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Santhosh Girirajan
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Sharmila Bhattacharya
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Biological and Physical Sciences Division, NASA Headquarters, Washington DC 20024, USA.
| |
Collapse
|
18
|
Sinha T, Ikelle L, Makia MS, Crane R, Zhao X, Kakakhel M, Al-Ubaidi MR, Naash MI. Riboflavin deficiency leads to irreversible cellular changes in the RPE and disrupts retinal function through alterations in cellular metabolic homeostasis. Redox Biol 2022; 54:102375. [PMID: 35738087 PMCID: PMC9233280 DOI: 10.1016/j.redox.2022.102375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 10/25/2022] Open
Abstract
Ariboflavinosis is a pathological condition occurring as a result of riboflavin deficiency. This condition is treatable if detected early enough, but it lacks timely diagnosis. Critical symptoms of ariboflavinosis include neurological and visual manifestations, yet the effects of flavin deficiency on the retina are not well investigated. Here, using a diet induced mouse model of riboflavin deficiency, we provide the first evidence of how retinal function and metabolism are closely intertwined with riboflavin homeostasis. We find that diet induced riboflavin deficiency causes severe decreases in retinal function accompanied by structural changes in the neural retina and retinal pigment epithelium (RPE). This is preceded by increased signs of cellular oxidative stress and metabolic disorder, in particular dysregulation in lipid metabolism, which is essential for both photoreceptors and the RPE. Though many of these deleterious phenotypes can be ameliorated by riboflavin supplementation, our data suggests that some patients may continue to suffer from multiple pathologies at later ages. These studies provide an essential cellular and mechanistic foundation linking defects in cellular flavin levels with the manifestation of functional deficiencies in the visual system and paves the way for a more in-depth understanding of the cellular consequences of ariboflavinosis.
Collapse
Affiliation(s)
- Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Larissa Ikelle
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mustafa S Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Xue Zhao
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mashal Kakakhel
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
19
|
From Molecules to Behavior in Long-Term Inorganic Mercury Intoxication: Unraveling Proteomic Features in Cerebellar Neurodegeneration of Rats. Int J Mol Sci 2021; 23:ijms23010111. [PMID: 35008538 PMCID: PMC8745249 DOI: 10.3390/ijms23010111] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Mercury is a severe environmental pollutant with neurotoxic effects, especially when exposed for long periods. Although there are several evidences regarding mercury toxicity, little is known about inorganic mercury (IHg) species and cerebellum, one of the main targets of mercury associated with the neurological symptomatology of mercurial poisoning. Besides that, the global proteomic profile assessment is a valuable tool to screen possible biomarkers and elucidate molecular targets of mercury neurotoxicity; however, the literature is still scarce. Thus, this study aimed to investigate the effects of long-term exposure to IHg in adult rats’ cerebellum and explore the modulation of the cerebellar proteome associated with biochemical and functional outcomes, providing evidence, in a translational perspective, of new mercury toxicity targets and possible biomarkers. Fifty-four adult rats were exposed to 0.375 mg/kg of HgCl2 or distilled water for 45 days using intragastric gavage. Then, the motor functions were evaluated by rotarod and inclined plane. The cerebellum was collected to quantify mercury levels, to assess the antioxidant activity against peroxyl radicals (ACAPs), the lipid peroxidation (LPO), the proteomic profile, the cell death nature by cytotoxicity and apoptosis, and the Purkinje cells density. The IHg exposure increased mercury levels in the cerebellum, reducing ACAP and increasing LPO. The proteomic approach revealed a total 419 proteins with different statuses of regulation, associated with different biological processes, such as synaptic signaling, energy metabolism and nervous system development, e.g., all these molecular changes are associated with increased cytotoxicity and apoptosis, with a neurodegenerative pattern on Purkinje cells layer and poor motor coordination and balance. In conclusion, all these findings feature a neurodegenerative process triggered by IHg in the cerebellum that culminated into motor functions deficits, which are associated with several molecular features and may be related to the clinical outcomes of people exposed to the toxicant.
Collapse
|
20
|
Pourbagher-Shahri AM, Farkhondeh T, Talebi M, Kopustinskiene DM, Samarghandian S, Bernatoniene J. An Overview of NO Signaling Pathways in Aging. Molecules 2021; 26:4533. [PMID: 34361685 PMCID: PMC8348219 DOI: 10.3390/molecules26154533] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Nitric Oxide (NO) is a potent signaling molecule involved in the regulation of various cellular mechanisms and pathways under normal and pathological conditions. NO production, its effects, and its efficacy, are extremely sensitive to aging-related changes in the cells. Herein, we review the mechanisms of NO signaling in the cardiovascular system, central nervous system (CNS), reproduction system, as well as its effects on skin, kidneys, thyroid, muscles, and on the immune system during aging. The aging-related decline in NO levels and bioavailability is also discussed in this review. The decreased NO production by endothelial nitric oxide synthase (eNOS) was revealed in the aged cardiovascular system. In the CNS, the decline of the neuronal (n)NOS production of NO was related to the impairment of memory, sleep, and cognition. NO played an important role in the aging of oocytes and aged-induced erectile dysfunction. Aging downregulated NO signaling pathways in endothelial cells resulting in skin, kidney, thyroid, and muscle disorders. Putative therapeutic agents (natural/synthetic) affecting NO signaling mechanisms in the aging process are discussed in the present study. In summary, all of the studies reviewed demonstrate that NO plays a crucial role in the cellular aging processes.
Collapse
Affiliation(s)
- Ali Mohammad Pourbagher-Shahri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| | - Marjan Talebi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 1991953381, Iran;
| | - Dalia M. Kopustinskiene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu Pr. 13, LT-50161 Kaunas, Lithuania;
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran
| | - Jurga Bernatoniene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu Pr. 13, LT-50161 Kaunas, Lithuania;
- Department of Drug Technology and Social Pharmacy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu Pr. 13, LT-50161 Kaunas, Lithuania
| |
Collapse
|
21
|
Oyigeya M. Reflex memory theory of acquired involuntary motor and sensory disorders. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2021. [DOI: 10.1186/s41983-021-00307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Explicit and implicit memories are conserved but flexible biological tools that nature uses to regulate the daily behaviors of human beings. An aberrant form of the implicit memory is presumed to exist and may be contributory to the pathophysiology of disorders such as tardive syndromes, phantom phenomena, flashback, posttraumatic stress disorders (PTSD), and related disorders. These disorders have posed significant clinical problems for both patients and physicians for centuries. All extant pathophysiological theories of these disorders have failed to provide basis for effective treatment.
Objective
The objective of this article is to propose an alternative pathophysiological theory that will hopefully lead to new treatment approaches.
Methods
The author sourced over 60 journal articles that treated topics on memory, and involuntary motor and sensory disorders, from open access journals using Google Scholar, and reviewed them and this helped in the formulation of this theory.
Results
From the reviews, the author thinks physical or chemical insult to the nervous system can cause defective circuit remodeling, leading to generation of a variant of implicit (automatic) memory, herein called “reflex memory” and this is encoded interoceptively to contribute to these phenomena states.
Conclusion
Acquired involuntary motor and sensory disorders are caused by defective circuit remodeling involving multiple neural mechanisms. Dysregulation of excitatory neurotransmitters, calcium overload, homeostatic failure, and neurotoxicity are implicated in the process. Sustained effects of these defective mechanisms are encoded interoceptively as abnormal memory in the neurons and the conscious manifestations are these disorders. Extant theories failed to recognize this possibility.
Collapse
|
22
|
Cuttler K, Bignoux MJ, Otgaar TC, Chigumba S, Ferreira E, Weiss SFT. LRP::FLAG Reduces Phosphorylated Tau Levels in Alzheimer's Disease Cell Culture Models. J Alzheimers Dis 2021; 76:753-768. [PMID: 32568204 DOI: 10.3233/jad-200244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque and neurofibrillary tangle formation, respectively. Neurofibrillary tangles form as a result of the intracellular accumulation of hyperphosphorylated tau. Telomerase activity and levels of the human reverse transcriptase (hTERT) subunit of telomerase are significantly decreased in AD. Recently, it has been demonstrated that the 37 kDa/67 kDa laminin receptor (LRP/LR) interacts with telomerase and is implicated in Aβ pathology. Since both LRP/LR and telomerase are known to play a role in the Aβ facet of AD, we hypothesized that they might also play a role in tauopathy. OBJECTIVE This study aimed to determine if LRP/LR has a relationship with tau and whether overexpression of LRP::FLAG has an effect on tauopathy-related proteins. METHODS We employed confocal microscopy and FRET to determine whether LRP/LR and tau co-localize and interact. LRP::FLAG overexpression in HEK-293 and SH-SY5Y cells as well as analysis of tauopathy-related proteins was assessed by western blotting. RESULTS We demonstrate that LRP/LR co-localizes with tau in the perinuclear cell compartment and confirmed a direct interaction between LRP/LR and tau in HEK-293 cells. Overexpression of LRP::FLAG in HEK-293 and SH-SY5Y cells decreased total and phosphorylated tau levels with a concomitant decrease in PrPc levels, a tauopathy-related protein. LRP::FLAG overexpression also resulted in increased hTERT levels. CONCLUSION This data suggest that LRP/LR extends its role in AD through a direct interaction with tau, and recommend LRP::FLAG as a possible alternative AD therapeutic via decreasing phosphorylated tau levels.
Collapse
Affiliation(s)
- Katelyn Cuttler
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa.,Present Address: Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Republic of South Africa
| | - Monique J Bignoux
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Tyrone C Otgaar
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Stephanie Chigumba
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Eloise Ferreira
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Stefan F T Weiss
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Republic of South Africa
| |
Collapse
|
23
|
Karoglu-Eravsar ET, Tuz-Sasik MU, Adams MM. Environmental enrichment applied with sensory components prevents age-related decline in synaptic dynamics: Evidence from the zebrafish model organism. Exp Gerontol 2021; 149:111346. [PMID: 33838219 DOI: 10.1016/j.exger.2021.111346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 10/21/2022]
Abstract
Progression of cognitive decline with or without neurodegeneration varies among elderly subjects. The main aim of the current study was to illuminate the molecular mechanisms that promote and retain successful aging in the context of factors such as environment and gender, both of which alter the resilience of the aging brain. Environmental enrichment (EE) is one intervention that may lead to the maintenance of cognitive processing at older ages in both humans and animal subjects. EE is easily applied to different model organisms, including zebrafish, which show similar age-related molecular and behavioral changes as humans. Global changes in cellular and synaptic markers with respect to age, gender and 4-weeks of EE applied with sensory stimulation were investigated using the zebrafish model organism. Results indicated that EE increases brain weight in an age-dependent manner without affecting general body parameters like body mass index (BMI). Age-related declines in the presynaptic protein synaptophysin, AMPA-type glutamate receptor subunits and a post-mitotic neuronal marker were observed and short-term EE prevents these changes in aged animals, as well as elevates levels of the inhibitory scaffolding protein, gephyrin. Gender-driven alterations were observed in the levels of the glutamate receptor subunits. Oxidative stress markers were significantly increased in the old animals, while exposure to EE did not alter this pattern. These data suggest that EE with sensory stimulation exerts its effects mainly on age-related changes in synaptic dynamics, which likely increase brain resilience through specific cellular mechanisms.
Collapse
Affiliation(s)
- Elif Tugce Karoglu-Eravsar
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey; National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey; Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey; Department of Psychology, Selcuk University, Konya, Turkey
| | - Melek Umay Tuz-Sasik
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey; National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey; Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey
| | - Michelle M Adams
- Interdisciplinary Program in Neuroscience, Aysel Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey; National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, Turkey; Department of Molecular Biology and Genetics, Zebrafish Facility, Bilkent University, Ankara, Turkey; Department of Psychology, Bilkent University, Ankara, Turkey.
| |
Collapse
|
24
|
Hacioglu C, Kar F, Kanbak G. Ex Vivo Investigation of Bexarotene and Nicotinamide Function as a Protectıve Agent on Rat Synaptosomes Treated with Aβ(1-42). Neurochem Res 2021; 46:804-818. [PMID: 33428094 DOI: 10.1007/s11064-020-03216-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 01/09/2023]
Abstract
In this study, we were aimed to investigate the neuroprotective effects of bexarotene and nicotinamide in synaptosomes incubated with amyloid-beta (Aβ). Our study consists of 2 parts, in vivo and in vitro. In the in vivo section, twenty-four Wistar albino male rats were divided into 4 groups (control, dimethyl sulfoxide (DMSO), nicotinamide and bexarotene) with six animals in each group. DMSO(1%), nicotinamide(100 mg/kg) and bexarotene(0.1 mg/kg) were administered intraperitoneally to animals in the experimental groups for seven days. In the in vitro part of our study, three different isolation methods were used to obtain the synaptosomes from the brain tissue. Total antioxidant capacity(TAS), total oxidant capacity(TOS), cleaved caspase 3(CASP3), cytochrome c(Cyt c), sirtuin 1(SIRT1), peroxisome proliferator-activated receptor gamma(PPARγ) and poly(ADP-ribose) polymerase-1(PARP-1) levels in the synaptosomes incubated with a concentration of 10 µM Aβ(1-42) were measured by enzyme-linked immunosorbent assay method. Biochemical analysis and histopathological examinations in serum and brain samples showed that DMSO, nicotinamide and bexarotene treatments did not cause any damage to the rat brain tissue. We found that in vitro Aβ(1-42) administration decreased TAS, SIRT1 and PPARγ levels in synaptosomes while increasing TOS, CASP3, Cyt c, and PARP1 levels. Nicotinamide treatment suppressed oxidative stress and apoptosis by supporting antioxidant capacity and increased PPARγ through SIRT1 activation, causing PARP1 to decrease. On the other hand, bexarotene caused a moderate increase in SIRT1 levels with PPARγ activation. Consequently, we found that nicotinamide can be more effective than bexarotene in AD pathogenesis by regulating mitochondrial functions in synaptosomes.
Collapse
Affiliation(s)
- Ceyhan Hacioglu
- Department of Medical Biochemistry, Faculty of Medicine, Duzce University, Duzce, Turkey.
| | - Fatih Kar
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Gungor Kanbak
- Department of Medical Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
25
|
Salahinejad A, Attaran A, Naderi M, Meuthen D, Niyogi S, Chivers DP. Chronic exposure to bisphenol S induces oxidative stress, abnormal anxiety, and fear responses in adult zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 750:141633. [PMID: 32882496 DOI: 10.1016/j.scitotenv.2020.141633] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/04/2020] [Accepted: 08/09/2020] [Indexed: 06/11/2023]
Abstract
Bisphenol S (BPS) is increasingly used in a wide range of industrial and consumer products, resulting in its ubiquitous distribution across the environment, including aquatic ecosystems. Although it is commonly known as a weak/moderate estrogenic compound, there has been a growing acknowledgment of the potential of BPS to cause toxicity by inducing oxidative stress. Oxidative stress is a major participant in the development of anxiety-like behaviors in humans and animals. Therefore, the present study was designed to examine the impact of BPS on anxiety-like behavior and fear responses in adult zebrafish and also to elucidate the possible linkage between the BPS neurotoxicity and oxidative status of the brain. To this end, adult male and female zebrafish were exposed to 0 (control), 1, 10, and 30 μg/L of BPS and 1 μg/L of 17-β-estradiol (E2) for 75 days. Following exposure, changes in anxiety and fear-related responses were evaluated by applying a novel tank test and by exposing focal fish to chemical alarm cues. Additionally, we evaluated the expression of multiple antioxidant genes in the zebrafish brain. Our results indicate that BPS, irrespective of exposure concentration, and E2 significantly decreased bottom-dwelling behavior and the latency to enter the upper water column. Furthermore, exposure to the highest concentration of BPS and E2 induced a significant decrease in fear-related responses. The impaired anxiety and reduced fear-related responses were associated with a down-regulation in the transcription of genes involved in enzymatic antioxidant defense. Taken together, our results suggest that chronic exposure to BPS impairs anxiety and fear responses in adult zebrafish, possibly by inducing oxidative stress in the brain.
Collapse
Affiliation(s)
- Arash Salahinejad
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada.
| | - Anoosha Attaran
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Mohammad Naderi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Denis Meuthen
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; Evolutionary Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, SK S7N 5B3, Canada
| | - Douglas P Chivers
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| |
Collapse
|
26
|
Golriz Khatami S, Domingo-Fernández D, Mubeen S, Hoyt CT, Robinson C, Karki R, Iyappan A, Kodamullil AT, Hofmann-Apitius M. A Systems Biology Approach for Hypothesizing the Effect of Genetic Variants on Neuroimaging Features in Alzheimer's Disease. J Alzheimers Dis 2021; 80:831-840. [PMID: 33554913 PMCID: PMC8075382 DOI: 10.3233/jad-201397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Neuroimaging markers provide quantitative insight into brain structure and function in neurodegenerative diseases, such as Alzheimer's disease, where we lack mechanistic insights to explain pathophysiology. These mechanisms are often mediated by genes and genetic variations and are often studied through the lens of genome-wide association studies. Linking these two disparate layers (i.e., imaging and genetic variation) through causal relationships between biological entities involved in the disease's etiology would pave the way to large-scale mechanistic reasoning and interpretation. OBJECTIVE We explore how genetic variants may lead to functional alterations of intermediate molecular traits, which can further impact neuroimaging hallmarks over a series of biological processes across multiple scales. METHODS We present an approach in which knowledge pertaining to single nucleotide polymorphisms and imaging readouts is extracted from the literature, encoded in Biological Expression Language, and used in a novel workflow to assist in the functional interpretation of SNPs in a clinical context. RESULTS We demonstrate our approach in a case scenario which proposes KANSL1 as a candidate gene that accounts for the clinically reported correlation between the incidence of the genetic variants and hippocampal atrophy. We find that the workflow prioritizes multiple mechanisms reported in the literature through which KANSL1 may have an impact on hippocampal atrophy such as through the dysregulation of cell proliferation, synaptic plasticity, and metabolic processes. CONCLUSION We have presented an approach that enables pinpointing relevant genetic variants as well as investigating their functional role in biological processes spanning across several, diverse biological scales.
Collapse
Affiliation(s)
- Sepehr Golriz Khatami
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
| | - Sarah Mubeen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Charles Tapley Hoyt
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
| | - Christine Robinson
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Reagon Karki
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Anandhi Iyappan
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Alpha Tom Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
27
|
Bingul D, Kalra K, Murata EM, Belser A, Dash MB. Persistent changes in extracellular lactate dynamics following synaptic potentiation. Neurobiol Learn Mem 2020; 175:107314. [PMID: 32961277 PMCID: PMC7655607 DOI: 10.1016/j.nlm.2020.107314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/27/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
A diverse array of neurometabolic coupling mechanisms exist within the brain to ensure that sufficient metabolite availability is present to meet both acute and chronic energetic demands. Excitatory synaptic activity, which produces the majority of the brain's energetic demands, triggers a rapid metabolic response including a characteristic shift towards aerobic glycolysis. Herein, astrocytically derived lactate appears to serve as an important metabolite to meet the extensive metabolic needs of activated neurons. Despite a wealth of literature characterizing lactate's role in mediating these acute metabolic needs, the extent to which lactate supports chronic energetic demands of neurons remains unclear. We hypothesized that synaptic potentiation, a ubiquitous brain phenomenon that can produce chronic alterations in synaptic activity, could necessitate persistent alterations in brain energetics. In freely-behaving rats, we induced long-term potentiation (LTP) of synapses within the dentate gyrus through high-frequency electrical stimulation (HFS) of the medial perforant pathway. Before, during, and after LTP induction, we continuously recorded extracellular lactate concentrations within the dentate gyrus to assess how changes in synaptic strength alter local glycolytic activity. Synaptic potentiation 1) altered the acute response of extracellular lactate to transient neuronal activation as evident by a larger initial dip and subsequent overshoot and 2) chronically increased local lactate availability. Although synapses were potentiated immediately following HFS, observed changes in lactate dynamics were only evident beginning ~24 h later. Once observed, however, both synaptic potentiation and altered lactate dynamics persisted for the duration of the experiment (~72 h). Persistent alterations in synaptic strength, therefore, appear to be associated with metabolic plasticity in the form of persistent augmentation of glycolytic activity.
Collapse
Affiliation(s)
- D Bingul
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - K Kalra
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - E M Murata
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - A Belser
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - M B Dash
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States; Department of Psychology, Middlebury College, Middlebury, VT 05753, United States.
| |
Collapse
|
28
|
Felix L, Delekate A, Petzold GC, Rose CR. Sodium Fluctuations in Astroglia and Their Potential Impact on Astrocyte Function. Front Physiol 2020; 11:871. [PMID: 32903427 PMCID: PMC7435049 DOI: 10.3389/fphys.2020.00871] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Astrocytes are the main cell type responsible for the regulation of brain homeostasis, including the maintenance of ion gradients and neurotransmitter clearance. These processes are tightly coupled to changes in the intracellular sodium (Na+) concentration. While activation of the sodium-potassium-ATPase (NKA) in response to an elevation of extracellular K+ may decrease intracellular Na+, the cotransport of transmitters, such as glutamate, together with Na+ results in an increase in astrocytic Na+. This increase in intracellular Na+ can modulate, for instance, metabolic downstream pathways. Thereby, astrocytes are capable to react on a fast time scale to surrounding neuronal activity via intracellular Na+ fluctuations and adjust energy production to the demand of their environment. Beside the well-documented conventional roles of Na+ signaling mainly mediated through changes in its electrochemical gradient, several recent studies have identified more atypical roles for Na+, including protein interactions leading to changes in their biochemical activity or Na+-dependent regulation of gene expression. In this review, we will address both the conventional as well as the atypical functions of astrocytic Na+ signaling, presenting the role of transporters and channels involved and their implications for physiological processes in the central nervous system (CNS). We will also discuss how these important functions are affected under pathological conditions, including stroke and migraine. We postulate that Na+ is an essential player not only in the maintenance of homeostatic processes but also as a messenger for the fast communication between neurons and astrocytes, adjusting the functional properties of various cellular interaction partners to the needs of the surrounding network.
Collapse
Affiliation(s)
- Lisa Felix
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Andrea Delekate
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
29
|
Update on Atypicalities of Central Nervous System in Autism Spectrum Disorder. Brain Sci 2020; 10:brainsci10050309. [PMID: 32443912 PMCID: PMC7287879 DOI: 10.3390/brainsci10050309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous, behaviorally defined, neurodevelopmental disorder that has been modeled as a brain-based disease. The behavioral and cognitive features of ASD are associated with pervasive atypicalities in the central nervous system (CNS). To date, the exact mechanisms underlying the pathophysiology of ASD still remain unknown and there is currently no cure or effective treatment for this disorder. Many publications implicated the association of ASD with inflammation, immune dysregulation, neurotransmission dysfunction, mitochondrial impairment and cell signaling dysregulation. This review attempts to highlight evidence of the major pathophysiology of ASD including abnormalities in the brain structure and function, neuroglial activation and neuroinflammation, glutamatergic neurotransmission, mitochondrial dysfunction and mechanistic target of rapamycin (mTOR) signaling pathway dysregulation. Molecular and cellular factors that contributed to the pathogenesis of ASD and how they may affect the development and function of CNS are compiled in this review. However, findings of published studies have been complicated by the fact that autism is a very heterogeneous disorder; hence, we addressed the limitations that led to discrepancies in the reported findings. This review emphasizes the need for future studies to control study variables such as sample size, gender, age range and intelligence quotient (IQ), all of which that could affect the study measurements. Neuroinflammation or immune dysregulation, microglial activation, genetically linked neurotransmission, mitochondrial dysfunctions and mTOR signaling pathway could be the primary targets for treating and preventing ASD. Further research is required to better understand the molecular causes and how they may contribute to the pathophysiology of ASD.
Collapse
|
30
|
Rodriguez C, Agulla J, Delgado-Esteban M. Refocusing the Brain: New Approaches in Neuroprotection Against Ischemic Injury. Neurochem Res 2020; 46:51-63. [PMID: 32189131 DOI: 10.1007/s11064-020-03016-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/28/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
A new era for neuroprotective strategies is emerging in ischemia/reperfusion. This has forced to review the studies existing to date based in neuroprotection against oxidative stress, which have undoubtedly contributed to clarify the brain endogenous mechanisms, as well as to identify possible therapeutic targets or biomarkers in stroke and other neurological diseases. The efficacy of exogenous administration of neuroprotective compounds has been shown in different studies so far. However, something must be missing to get these treatments successfully applied in the clinical environment. Here, the mechanisms involved in neuronal protection against physiological level of ROS and the main neuroprotective signaling pathways induced by excitotoxic and ischemic stimuli are reviewed. Also, the endogenous ischemic tolerance in terms of brain self-protection mechanisms against subsequent cerebral ischemia is revisited to highlight how the preconditioning has emerged as a powerful tool to understand these phenomena. A better understanding of endogenous defense against exacerbated ROS and metabolism in nervous cells will therefore aid to design pharmacological antioxidants targeted specifically against oxidative damage induced by ischemic injury, but also might be very valuable for translational medicine.
Collapse
Affiliation(s)
- Cristina Rodriguez
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain.,Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
| | - Jesús Agulla
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain.,Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
| | - María Delgado-Esteban
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain. .,Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain. .,Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca, Spain.
| |
Collapse
|
31
|
Intertwined ROS and Metabolic Signaling at the Neuron-Astrocyte Interface. Neurochem Res 2020; 46:23-33. [PMID: 31989468 DOI: 10.1007/s11064-020-02965-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/03/2020] [Accepted: 01/16/2020] [Indexed: 12/23/2022]
Abstract
Metabolism and redox signalling share critical nodes in the nervous system. In the last years, a series of major findings have challenged the current vision on how neural reactive oxygen species (ROS) are produced and handled in the nervous system. Once regarded as deleterious by-products, ROS are now shown to be essential for a metabolic and redox crosstalk. In turn, this coupling defines neural viability and function to control behaviour or leading to neurodegeneration when compromised. Findings like a different assembly of mitochondrial respiratory supercomplexes in neurons and astrocytes stands behind a divergent production of ROS in either cell type, more prominent in astrocytes. ROS levels are however tightly controlled by an antioxidant machinery in astrocytes, assumed as more efficient than that of neurons, to regulate redox signalling. By exerting this control in ROS abundance, metabolic functions are finely tuned in both neural cells. Further, a higher engagement of mitochondrial respiration and oxidative function in neurons, underpinned by redox equivalents supplied from the pentose phosphate pathway and from glia, differs from the otherwise strong glycolytic capacity of astrocytes. Here, we recapitulate major findings on how ROS and metabolism differ between neural cells but merge to define reciprocal signalling pathways, ultimately defining neural function and fate.
Collapse
|
32
|
Mounesh, Venugopal Reddy KR. The electrochemical investigation of carboxamide-PEG2-biotin-CoPc using composite MWCNTs on modified GCE: the sensitive detections for glucose and hydrogen peroxide. NEW J CHEM 2020. [DOI: 10.1039/c9nj05807a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The electroanalytical study of a synthesized novel tetra-cobalt(ii) carboxamide-PEG2-biotin phthalocyanine (CoTPEG2BAPc) composite with MWCNTs to create a biosensor with a high response to glucose in the presence of H2O2.
Collapse
Affiliation(s)
- Mounesh
- Department of Chemistry
- Vijayanagara Srikrishnadevaraya University
- Ballari-583 105
- India
| | - K. R. Venugopal Reddy
- Department of Chemistry
- Vijayanagara Srikrishnadevaraya University
- Ballari-583 105
- India
| |
Collapse
|
33
|
Pereda D, Al‐Osta I, Okorocha AE, Easton A, Hartell NA. Changes in presynaptic calcium signalling accompany age-related deficits in hippocampal LTP and cognitive impairment. Aging Cell 2019; 18:e13008. [PMID: 31310431 PMCID: PMC6718530 DOI: 10.1111/acel.13008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/10/2019] [Accepted: 06/26/2019] [Indexed: 11/27/2022] Open
Abstract
The loss of cognitive function accompanying healthy aging is not associated with extensive or characteristic patterns of cell death, suggesting it is caused by more subtle changes in synaptic properties. In the hippocampal CA1 region, long‐term potentiation requires stronger stimulation for induction in aged rats and mice and long‐term depression becomes more prevalent. An age‐dependent impairment of postsynaptic calcium homeostasis may underpin these effects. We have examined changes in presynaptic calcium signalling in aged mice using a transgenic mouse line (SyG37) that expresses a genetically encoded calcium sensor in presynaptic terminals. SyG37 mice showed an age‐dependent decline in cognitive abilities in behavioural tasks that require hippocampal processing including the Barnes maze, T‐maze and object location but not recognition tests. The incidence of LTP was significantly impaired in animals over 18 months of age. These effects of aging were accompanied by a persistent increase in resting presynaptic calcium, an increase in the presynaptic calcium signal following Schaffer collateral fibre stimulation, an increase in postsynaptic fEPSP slope and a reduction in paired‐pulse facilitation. These effects were not caused by synapse proliferation and were of presynaptic origin since they were evident in single presynaptic boutons. Aged synapses behaved like younger ones when the extracellular calcium concentration was reduced. Raising extracellular calcium had little effect on aged synapses but altered the properties of young synapses into those of their aged counterparts. These effects can be readily explained by an age‐dependent change in the properties or numbers of presynaptic calcium channels.
Collapse
Affiliation(s)
- Daniel Pereda
- Department of Neuroscience, Psychology and Behaviour University of Leicester Leicester UK
| | - Ibrahim Al‐Osta
- Department of Neuroscience, Psychology and Behaviour University of Leicester Leicester UK
| | - Albert E. Okorocha
- Department of Neuroscience, Psychology and Behaviour University of Leicester Leicester UK
| | | | - Nicholas A. Hartell
- Department of Neuroscience, Psychology and Behaviour University of Leicester Leicester UK
| |
Collapse
|
34
|
Occelli F, Hasselmann F, Bourien J, Eybalin M, Puel J, Desvignes N, Wiszniowski B, Edeline JM, Gourévitch B. Age-related Changes in Auditory Cortex Without Detectable Peripheral Alterations: A Multi-level Study in Sprague–Dawley Rats. Neuroscience 2019; 404:184-204. [DOI: 10.1016/j.neuroscience.2019.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/21/2019] [Accepted: 02/01/2019] [Indexed: 01/31/2023]
|
35
|
Wei Y, Wang R, Teng J. Inhibition of Calcium/Calmodulin-Dependent Protein Kinase IIα Suppresses Oxidative Stress in Cerebral Ischemic Rats Through Targeting Glucose 6-Phosphate Dehydrogenase. Neurochem Res 2019; 44:1613-1620. [PMID: 30919283 DOI: 10.1007/s11064-019-02785-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is a leading cause of mortality and morbidity worldwide, and oxidative stress plays a significant role in the ischemia stage and reperfusion stage. Previous studies have indicated that both calcium/calmodulin-dependent protein kinase II (CaMKII) and glucose 6-phosphate dehydrogenase (G6PD) are involved in the oxidative stress. Thus, the aim of this study was to investigate the roles of CaMKIIα, an important isoform of CaMKII, and G6PD in a rat model of middle cerebral artery occlusion (MCAO). Intracerebroventricular injection of small interfering ribonucleic acid (siRNA) for CaMKIIα was performed at 48 h pre-MCAO surgery. Immunofluorescence Staining and western blot were performed to detect the expression of p-CaMKIIα and G6PD in the cortices. 2, 3, 5-Triphenyltetrazolium chloride (TTC) staining was performed to investigate the infarct volume. In addition, neurological deficit, reactive oxygen species (ROS), ratio of reduced-to-oxidized glutathione (GSH/GSSG) and ratio of reduced-to-oxidized oxidized nicotinamide adenine dinucleotide phosphate (NADPH/NADP+) were assessed. The results indicated that both p-CaMKIIα and G6PD were widely located in the neurons and astrocytes, and their expression was gradually increased in the cortices after MCAO, which was accompanied by increased level of ROS and decreased levels of GSH/GSSG and NADPH/NADP+. However, after treatment with siRNA for CaMKIIα, p-CaMKIIα expression was decreased and G6PD expression was increased. Moreover, inhibition of CaMKIIα improved the neurological deficit, reduced the infarct volume, decreased the level of ROS and increased the levels of GSH/GSSG and NADPH/NADP+. The results suggested that CaMKIIα inhibition exerted neuroprotective effects through regulating G6PD expression, which provides a new target for prevention and treatment of stroke.
Collapse
Affiliation(s)
- Yamin Wei
- Neurological Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450000, Henan, China
| | - Rui Wang
- Neurological Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450000, Henan, China
| | - Junfang Teng
- Neurological Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
36
|
Soltys DT, Pereira CP, Rowies FT, Farfel JM, Grinberg LT, Suemoto CK, Leite RE, Rodriguez RD, Ericson NG, Bielas JH, Souza-Pinto NC. Lower mitochondrial DNA content but not increased mutagenesis associates with decreased base excision repair activity in brains of AD subjects. Neurobiol Aging 2019; 73:161-170. [DOI: 10.1016/j.neurobiolaging.2018.09.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/13/2018] [Accepted: 09/13/2018] [Indexed: 12/15/2022]
|
37
|
Gowthami N, Sunitha B, Kumar M, Keshava Prasad T, Gayathri N, Padmanabhan B, Srinivas Bharath M. Mapping the protein phosphorylation sites in human mitochondrial complex I (NADH: Ubiquinone oxidoreductase): A bioinformatics study with implications for brain aging and neurodegeneration. J Chem Neuroanat 2019; 95:13-28. [PMID: 29499254 DOI: 10.1016/j.jchemneu.2018.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 12/21/2022]
|
38
|
Ke T, Gonçalves FM, Gonçalves CL, Dos Santos AA, Rocha JBT, Farina M, Skalny A, Tsatsakis A, Bowman AB, Aschner M. Post-translational modifications in MeHg-induced neurotoxicity. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2068-2081. [PMID: 30385410 DOI: 10.1016/j.bbadis.2018.10.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
Mercury (Hg) exposure remains a major public health concern due to its widespread distribution in the environment. Organic mercurials, such as MeHg, have been extensively investigated especially because of their congenital effects. In this context, studies on the molecular mechanism of MeHg-induced neurotoxicity are pivotal to the understanding of its toxic effects and the development of preventive measures. Post-translational modifications (PTMs) of proteins, such as phosphorylation, ubiquitination, and acetylation are essential for the proper function of proteins and play important roles in the regulation of cellular homeostasis. The rapid and transient nature of many PTMs allows efficient signal transduction in response to stress. This review summarizes the current knowledge of PTMs in MeHg-induced neurotoxicity, including the most commonly PTMs, as well as PTMs induced by oxidative stress and PTMs of antioxidant proteins. Though PTMs represent an important molecular mechanism for maintaining cellular homeostasis and are involved in the neurotoxic effects of MeHg, we are far from understanding the complete picture on their role, and further research is warranted to increase our knowledge of PTMs in MeHg-induced neurotoxicity.
Collapse
Affiliation(s)
- Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Filipe Marques Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Cinara Ludvig Gonçalves
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | | | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, 97105900 Santa Maria, RS, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040900 Florianópolis, SC, Brazil
| | - Anatoly Skalny
- Yaroslavl State University, Sovetskaya St., 14, Yaroslavl 150000, Russia; Peoples' Friendship University of Russia (RUDN University), Miklukho-Maklaya St., 6, Moscow 105064, Russia; Orenburg State University, Pobedy Ave., 13, Orenburg 460352, Russia
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
39
|
Yang EJ, Mahmood U, Kim H, Choi M, Choi Y, Lee JP, Cho JY, Hyun JW, Kim YS, Chang MJ, Kim HS. Phloroglucinol ameliorates cognitive impairments by reducing the amyloid β peptide burden and pro-inflammatory cytokines in the hippocampus of 5XFAD mice. Free Radic Biol Med 2018; 126:221-234. [PMID: 30118828 DOI: 10.1016/j.freeradbiomed.2018.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 01/29/2023]
Abstract
Among the various causative factors involved in the pathogenesis of Alzheimer's disease (AD), oxidative stress has emerged as an important factor. Phloroglucinol is a polyphenol component of phlorotannin, which is found at sufficient levels in Ecklonia cava (E. cava). Phloroglucinol has been reported to exert antioxidant activities in various tissues. Previously, we reported that the stereotaxic injection of phloroglucinol regulated synaptic plasticity in an AD mouse model. In this study, we aimed to investigate the effects of oral administration of phloroglucinol in AD. The oral administration of phloroglucinol for 2 months attenuated the impairments in cognitive function observed in 6-month-old 5X familial AD (5XFAD) mice, as assessed with the T-maze and Y-maze tests. The administration of phloroglucinol for 2 months in 5XFAD mice caused a reduction in the number of amyloid plaques and in the protein level of BACE1, a major amyloid precursor protein cleavage enzyme, together with γ-secretase. Phloroglucinol also restored the reduction in dendritic spine density and the number of mature spines in the hippocampi of 5XFAD mice. In addition, phloroglucinol-administered 5XFAD mice displayed lower protein levels of GFAP and Iba-1 and mRNA levels of TNF-α and IL-6 compared with vehicle-administered 5XFAD mice. These results demonstrated that phloroglucinol alleviated the neuropathological features and behavioral phenotypes in the 5XFAD mouse model. Taken together, our results suggest that phloroglucinol has therapeutic potential for AD treatment.
Collapse
Affiliation(s)
- Eun-Jeong Yang
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Usman Mahmood
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Hyunju Kim
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Moonseok Choi
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Yunjung Choi
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Jean-Pyo Lee
- Department of Physiology, Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
| | - Joo-Youn Cho
- Department of Clinical Pharmacology & Therapeutics, College of Medicine, Seoul National University and Hospital, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Yong Sik Kim
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Moon-Jeong Chang
- Department of Foods and Nutrition, College of Natural Science, Kookmin University, Seoul 02707, Republic of Korea.
| | - Hye-Sun Kim
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Seoul National University Bundang Hospital, Seongnam, Sungnam, Bundang-Gu 13620, Republic of Korea; Neuroscience Research Institute, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea.
| |
Collapse
|
40
|
Srinivas Bharath MM. Post-Translational Oxidative Modifications of Mitochondrial Complex I (NADH: Ubiquinone Oxidoreductase): Implications for Pathogenesis and Therapeutics in Human Diseases. J Alzheimers Dis 2018; 60:S69-S86. [PMID: 28582861 DOI: 10.3233/jad-170117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondrial complex I (NADH: ubiquinone oxidoreductase; CI) is central to the electron transport chain (ETC), oxidative phosphorylation, and ATP production in eukaryotes. CI is a multi-subunit complex with a complicated yet organized structure that optimally connects electron transfer with proton translocation and forms higher-order supercomplexes with other ETC complexes. Efforts to understand the molecular genetics, expression profile of subunits, and structure-function relationship of CI have increased over the years due to the direct role of the complex in human diseases. Although mutations in the nuclear and mitochondrial genes of CI and altered expression of subunits could potentially lower CI activity leading to mitochondrial dysfunction in many diseases, oxidative post-translational modifications (PTMs) have emerged as an important mechanism contributing to altered CI activity. These mainly include reversible and irreversible cysteine modifications, tyrosine nitration, carbonylation, and tryptophan oxidation that are generated following exposure to reactive oxygen species/reactive nitrogen species. Interestingly, oxidative PTMs could contribute either to CI damage, mitochondrial dysfunction, and ensuing cell death or a response mechanism with potential cytoprotective effects. This has also emerged as a promising field for structural biologists since analysis of PTMs could assist in understanding the structure-function relationship of the complex and correlate electron transfer mechanism with energy production. However, analysis of PTMs of CI and their contribution to CI function are incomplete in many physiological and pathological conditions. This review aims to highlight the role of oxidative PTMs in modulating CI activity with implications toward pathobiology of CNS diseases and novel therapeutics.
Collapse
Affiliation(s)
- M M Srinivas Bharath
- Department of Neurochemistry and Neurotoxicology Laboratory at the Neurobiology Research Center, National Institute of Mental Health and Neurosciences, Bangalore, India
| |
Collapse
|
41
|
Meunier CJ, Mitchell EC, Roberts JG, Toups JV, McCarty GS, Sombers LA. Electrochemical Selectivity Achieved Using a Double Voltammetric Waveform and Partial Least Squares Regression: Differentiating Endogenous Hydrogen Peroxide Fluctuations from Shifts in pH. Anal Chem 2018; 90:1767-1776. [PMID: 29243477 DOI: 10.1021/acs.analchem.7b03717] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Hydrogen peroxide (H2O2) is a reactive oxygen species that serves as an important signaling molecule in normal brain function. At the same time, excessive H2O2 concentrations contribute to myriad pathological consequences resulting from oxidative stress. Studies to elucidate the diverse roles that H2O2 plays in complex biological environments have been hindered by the lack of robust methods for probing dynamic H2O2 fluctuations in living systems with molecular specificity. Background-subtracted fast-scan cyclic voltammetry at carbon-fiber microelectrodes provides a method of detecting rapid H2O2 fluctuations with high temporal and spatial resolution in brain tissue. However, H2O2 fluctuations can be masked by local changes in pH (ΔpH), because the voltammograms for these species can have significant peak overlap, hindering quantification. We present a method for removing ΔpH-related contributions from complex voltammetric data. By employing two distinct potential waveforms per scan, one in which H2O2 is electrochemically silent and a second in which both ΔpH and H2O2 are redox active, a clear distinction between H2O2 and ΔpH signals is established. A partial least-squares regression (PLSR) model is used to predict the ΔpH signal and subtract it from the voltammetric data. The model has been validated both in vitro and in vivo using k-fold cross-validation. The data demonstrate that the double waveform PLSR model is a powerful tool that can be used to disambiguate and evaluate naturally occurring H2O2 fluctuations in vivo.
Collapse
Affiliation(s)
- Carl J Meunier
- Department of Chemistry, North Carolina State University , Raleigh, North Carolina 27695-8204, United States
| | - Edwin C Mitchell
- Department of Chemistry, North Carolina State University , Raleigh, North Carolina 27695-8204, United States
| | - James G Roberts
- Department of Chemistry, North Carolina State University , Raleigh, North Carolina 27695-8204, United States
| | - Jonathan V Toups
- Department of Chemistry, North Carolina State University , Raleigh, North Carolina 27695-8204, United States
| | - Gregory S McCarty
- Department of Chemistry, North Carolina State University , Raleigh, North Carolina 27695-8204, United States
| | - Leslie A Sombers
- Department of Chemistry, North Carolina State University , Raleigh, North Carolina 27695-8204, United States
| |
Collapse
|
42
|
Pasinetti GM, Singh R, Westfall S, Herman F, Faith J, Ho L. The Role of the Gut Microbiota in the Metabolism of Polyphenols as Characterized by Gnotobiotic Mice. J Alzheimers Dis 2018; 63:409-421. [PMID: 29660942 PMCID: PMC6021178 DOI: 10.3233/jad-171151] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A growing body of experimental data suggests that microbes in the gut influence behavior and can alter brain physiology and neurochemistry. Although promising, researchers are only starting to understand the potential of the gut microbiota for use in neurological disease. Recent evidence demonstrated that gastrointestinal activities are linked to mood disorders such as anxiety, depression, and most recently, cognitive functions in age-related neurodegenerative disorders. Studies from our group and others are uncovering new evidence suggesting that the gut microbiota plays a crucial role in the metabolism and bioavailability of certain dietary compounds and synthetic drugs. Based on this evidence, this review article will discuss the implications of the gut microbiota in mechanisms of bioavailability and biotransformation with an emphasis on dietary polyphenol compounds. This will be followed by a survey of ongoing innovative research identifying the ability of individual gut bacteria to enhance the bioavailability of gut-derived, brain-penetrating, bioactive polyphenol metabolites that ultimately influence mechanisms associated with the promotion of resilience against psychological and cognitive impairment in response to stress. Lastly, current research initiatives aimed at promoting the generation of brain bioactive polyphenol metabolites by specialized gut microbes will be discussed, specifically the use of gnotobiotic mice to develop bioengineered second generation probiotics. We propose that leveraging the gut microbial ecosystem to generate brain targeted bioactive metabolites from dietary polyphenols can attenuate lifestyle risk factors and promote resilience against age-related cognitive decline.
Collapse
Affiliation(s)
- Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Risham Singh
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Westfall
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Francis Herman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeremiah Faith
- Department of Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lap Ho
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
43
|
Joh Y, Choi WS. Mitochondrial Complex I Inhibition Accelerates Amyloid Toxicity. Dev Reprod 2017; 21:417-424. [PMID: 29354787 PMCID: PMC5769135 DOI: 10.12717/dr.2017.21.4.417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 12/21/2017] [Accepted: 12/26/2017] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is neurodegenerative disease, characterized by the
progressive decline of memory, cognitive functions, and changes in personality.
The major pathological features in postmortem brains are neurofibrillary tangles
and amyloid beta (Aβ) deposits. The majority of AD cases are sporadic and
age-related. Although AD pathogenesis has not been established, aging and
declining mitochondrial function has been associated. Mitochondrial dysfunction
has been observed in AD patients' brains and AD mice models, and the mice with a
genetic defect in mitochondrial complex I showed enhanced Aβ level in
vivo. To elucidate the role of mitochondrial complex I in AD, we
used SH-SY5Y cells transfected with DNA constructs expressing human amyloid
precursor protein (APP) or human Swedish APP mutant (APP-swe). The expression of
APP-swe increased the level of Aβ protein in comparison with control. When
complex I was inhibited by rotenone, the increase of ROS level was remarkably
higher in the cells overexpressing APP-swe compared to control. The number of
dead cell was significantly increased in APP-swe-expressing cells by complex I
inhibition. We suggest that complex I dysfunction accelerate amyloid toxicity
and mitochondrial complex I dysfunction in aging may contribute to the
pathogenesis of sporadic AD.
Collapse
Affiliation(s)
- Yechan Joh
- School of Biological Sciences and Technology, College of Natural Sciences, College of Medicine, Chonnam National University, Gwangju 61186, Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, College of Natural Sciences, College of Medicine, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
44
|
Taschetto L, Scaini G, Zapelini HG, Ramos ÂC, Strapazzon G, Andrade VM, Réus GZ, Michels M, Dal-Pizzol F, Quevedo J, Schuck PF, Ferreira GC, Streck EL. Acute and long-term effects of intracerebroventricular administration of α-ketoisocaproic acid on oxidative stress parameters and cognitive and noncognitive behaviors. Metab Brain Dis 2017; 32:1507-1518. [PMID: 28550500 DOI: 10.1007/s11011-017-0035-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/16/2017] [Indexed: 01/07/2023]
Abstract
Maple Syrup Urine Disease (MSUD) is biochemically characterized by elevated levels of leucine, isoleucine and valine, as well as their corresponding transaminated branched-chain α-keto acids in tissue and biological fluids. Neurological symptoms and cerebral abnormalities, whose mechanisms are still unknown, are typical of this metabolic disorder. In the present study, we evaluated the early effects (1 h after injection) and long-term effects (15 days after injection) of a single intracerebroventricular administration of α-ketoisocaproic acid (KIC) on oxidative stress parameters and cognitive and noncognitive behaviors. Our results showed that KIC induced early and long-term effects; we found an increase in TBARS levels, protein carbonyl content and DNA damage in the hippocampus, striatum and cerebral cortex both one hour and 15 days after KIC administration. Moreover, SOD activity increased in the hippocampus and striatum one hour after injection, whereas after 15 days, SOD activity decreased only in the striatum. On the other hand, KIC significantly decreased CAT activity in the striatum one hour after injection, but 15 days after KIC administration, we found a decrease in CAT activity in the hippocampus and striatum. Finally, we showed that long-term cognitive deficits follow the oxidative damage; KIC induced impaired habituation memory and long-term memory impairment. From the biochemical and behavioral findings, it we presume that KIC provokes oxidative damage, and the persistence of brain oxidative stress is associated with long-term memory impairment and prepulse inhibition.
Collapse
Affiliation(s)
- Luciane Taschetto
- Laboratório de Bioenergética e Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
- Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Porto Alegre, RS, Brazil
| | - Giselli Scaini
- Laboratório de Bioenergética e Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil
- Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Porto Alegre, RS, Brazil
| | - Hugo G Zapelini
- Laboratório de Erros Inatos do Metabolismo, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Ândrea C Ramos
- Laboratório de Erros Inatos do Metabolismo, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Giulia Strapazzon
- Laboratório de Biologia Celular e Molecular, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Vanessa M Andrade
- Laboratório de Biologia Celular e Molecular, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gislaine Z Réus
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Monique Michels
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe Dal-Pizzol
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Quevedo
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Patrícia F Schuck
- Laboratório de Erros Inatos do Metabolismo, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gustavo C Ferreira
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emilio L Streck
- Laboratório de Bioenergética e Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105, Criciúma, SC, 88806-000, Brazil.
- Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Porto Alegre, RS, Brazil.
| |
Collapse
|
45
|
Xiao X, Zhang H, Wang H, Li Q, Zhang T. Neuroprotective effect of amantadine on corticosterone-induced abnormal glutamatergic synaptic transmission of CA3-CA1 pathway in rat's hippocampal slices. Synapse 2017; 71. [PMID: 28902436 DOI: 10.1002/syn.22010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/04/2017] [Accepted: 09/08/2017] [Indexed: 01/11/2023]
Abstract
Depression is a psychiatric disorder and chronic stress, leading to altered glucocorticoid secretion patterns, is one of the factors that induce depression. Our previous study showed that amantadine significantly attenuated the impairments of synaptic plasticity and cognitive function a rat model of CUS. However, little is known regarding the underlying mechanism. In the present study, the whole-cell patch-clamp technique was applied to examine the protection effect of amantadine on the hippocampus CA3-CA1 pathway. Evoked excitatory postsynaptic currents (eEPSCs), miniature excitatory postsynaptic currents (mEPSCs), paired-pulse ratio (PPR) and the action potentials of CA3 neurons were recorded. Our data showed that corticosterone increased the amplitude of eEPSCs and decreased the value of paired-pulse ratio (PPR), but both of them were significantly reversed by amantadine. In addition, the frequency of mEPSC was considerably increased by corticosterone, but it was reduced by amantadine. Moreover, we used the Fluo-3/AM image to detect the Ca2+ influx in primary cultured hippocampal neurons. The results showed that the intracellular calcium levels were significantly decreased by amantadine in the corticosterone treated neurons. Additionally, the superoxide dismutase (SOD) and catalase (CAT) activities were reduced by corticosterone, while they were enhanced by either amantadine or low-calcium artificial cerebral spinal fluid (ACSF). These results suggest that amantadine significantly improves corticosterone-induced abnormal glutamatergic synaptic transmission of CA3-CA1 synapses presynaptically and alleviates the activities of antioxidant enzymes via regulating the calcium influx.
Collapse
Affiliation(s)
- Xi Xiao
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Hui Zhang
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Hui Wang
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Qun Li
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Tao Zhang
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| |
Collapse
|
46
|
Amorim JA, Canas PM, Tomé AR, Rolo AP, Agostinho P, Palmeira CM, Cunha RA. Mitochondria in Excitatory and Inhibitory Synapses have Similar Susceptibility to Amyloid-β Peptides Modeling Alzheimer’s Disease. J Alzheimers Dis 2017; 60:525-536. [DOI: 10.3233/jad-170356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- João A. Amorim
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Paula M. Canas
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Angelo R. Tomé
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Anabela P. Rolo
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Paula Agostinho
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| | - Carlos M. Palmeira
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Rodrigo A. Cunha
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| |
Collapse
|
47
|
Ito YA, Di Polo A. Mitochondrial dynamics, transport, and quality control: A bottleneck for retinal ganglion cell viability in optic neuropathies. Mitochondrion 2017; 36:186-192. [PMID: 28866056 DOI: 10.1016/j.mito.2017.08.014] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/11/2017] [Accepted: 08/29/2017] [Indexed: 01/03/2023]
Abstract
Retinal ganglion cells, the neurons that selectively die in glaucoma and other optic neuropathies, are endowed with an exceedingly active metabolism and display a particular vulnerability to mitochondrial dysfunction. Mitochondria are exquisitely dynamic organelles that are continually responding to endogenous and environmental cues to readily meet the energy demand of neuronal networks. The highly orchestrated regulation of mitochondrial biogenesis, fusion, fission, transport and degradation is paramount for the maintenance of energy-expensive synapses at RGC dendrites and axon terminals geared for optimal neurotransmission. The present review focuses on the progress made to date on understanding the biology of mitochondrial dynamics and quality control and how dysregulation of these processes can profoundly affect retinal ganglion cell viability and function in optic nerve diseases.
Collapse
Affiliation(s)
- Yoko A Ito
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Université de Montréal, Montreal, Quebec H2X 1R9, Canada
| | - Adriana Di Polo
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Université de Montréal, Montreal, Quebec H2X 1R9, Canada.
| |
Collapse
|
48
|
Resolving Contributions of Oxygen-Consuming and ROS-Generating Enzymes at the Synapse. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1089364. [PMID: 28003863 PMCID: PMC5149698 DOI: 10.1155/2016/1089364] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/10/2016] [Accepted: 11/06/2016] [Indexed: 01/25/2023]
Abstract
Disruption of cellular redox homeostasis is implicated in a wide variety of pathologic conditions and aging. A fundamental factor that dictates such balance is the ratio between mitochondria-mediated complete oxygen reduction into water and incomplete reduction into superoxide radical by mitochondria and NADPH oxidase (NOX) enzymatic activity. Here we determined mitochondrial as well as NOX-dependent rates of oxygen consumption in parallel with H2O2 generation in freshly isolated synaptosomes using high resolution respirometry combined with fluorescence or electrochemical sensory. Our results indicate that although synaptic mitochondria exhibit substantially higher respiratory activities (8–82-fold greater than NOX oxygen consumption depending on mitochondrial respiratory state), NADPH-dependent oxygen consumption is associated with greater H2O2 production (6-7-fold higher NOX-H2O2). We also show that, in terms of the consumed oxygen, while synaptic mitochondria “leaked” 0.71% ± 0.12 H2O2 during NAD+-linked resting, 0.21% ± 0.04 during NAD+-linked active respiration, and 0.07% ± 0.02 during FAD+-linked active respiration, NOX converted 38% ± 13 of O2 into H2O2. Our results indicate that NOX rather than mitochondria is the major source of synaptic H2O2. The present approach may assist in the identification of redox-modulating synaptic factors that underlie a variety of physiological and pathological processes in neurons.
Collapse
|
49
|
Berezhnoy DS, Fedorova TN, Stvolinskii SL, Inozemtsev AN. Carnosine modulates oxidative homeostasis and levels of neurotransmitters in the brain in models of learning with positive and negative reinforcement. NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416040048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Beckhauser TF, Francis-Oliveira J, De Pasquale R. Reactive Oxygen Species: Physiological and Physiopathological Effects on Synaptic Plasticity. J Exp Neurosci 2016; 10:23-48. [PMID: 27625575 PMCID: PMC5012454 DOI: 10.4137/jen.s39887] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/09/2016] [Accepted: 08/13/2016] [Indexed: 12/18/2022] Open
Abstract
In the mammalian central nervous system, reactive oxygen species (ROS) generation is counterbalanced by antioxidant defenses. When large amounts of ROS accumulate, antioxidant mechanisms become overwhelmed and oxidative cellular stress may occur. Therefore, ROS are typically characterized as toxic molecules, oxidizing membrane lipids, changing the conformation of proteins, damaging nucleic acids, and causing deficits in synaptic plasticity. High ROS concentrations are associated with a decline in cognitive functions, as observed in some neurodegenerative disorders and age-dependent decay of neuroplasticity. Nevertheless, controlled ROS production provides the optimal redox state for the activation of transductional pathways involved in synaptic changes. Since ROS may regulate neuronal activity and elicit negative effects at the same time, the distinction between beneficial and deleterious consequences is unclear. In this regard, this review assesses current research and describes the main sources of ROS in neurons, specifying their involvement in synaptic plasticity and distinguishing between physiological and pathological processes implicated.
Collapse
Affiliation(s)
- Thiago Fernando Beckhauser
- Physiology and Biophysics Department, Biomedical Sciences Institute, Sao Paulo University (USP), Butanta, Sao Paulo, Brazil
| | - José Francis-Oliveira
- Physiology and Biophysics Department, Biomedical Sciences Institute, Sao Paulo University (USP), Butanta, Sao Paulo, Brazil
| | - Roberto De Pasquale
- Physiology and Biophysics Department, Biomedical Sciences Institute, Sao Paulo University (USP), Butanta, Sao Paulo, Brazil
| |
Collapse
|