1
|
Walls AB, Andersen JV, Waagepetersen HS, Bak LK. Fueling Brain Inhibition: Integrating GABAergic Neurotransmission and Energy Metabolism. Neurochem Res 2025; 50:136. [PMID: 40189668 DOI: 10.1007/s11064-025-04384-0] [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: 01/29/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025]
Abstract
Despite decades of research in brain energy metabolism and to what extent different cell types utilize distinct substrates for their energy metabolism, this topic remains a vibrant area of neuroscience research. In this review, we focus on the substrates utilized by the inhibitory GABAergic neurons, which has been less explored than glutamatergic neurons. First, we discuss how GABAergic neurons may utilize both glucose, lactate, or ketone bodies under different functional conditions, and provide some preliminary data suggesting that unlike glutamatergic neurons, GABAergic neurons work well when substrate supply is restricted to lactate. We end by discussing the role of GABAergic neuron energy metabolism in pathologies where failure of inhibitory function play a central role, namely epilepsy, hepatic encephalopathy, and Alzheimer's disease.
Collapse
Affiliation(s)
- Anne B Walls
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Capital Region Hospital Pharmacy, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Lasse K Bak
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
- Translational Research Center (TRACE), Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark.
| |
Collapse
|
2
|
Salarvandian S, Digaleh H, Khodagholi F, Javadpour P, Asadi S, Zaman AAO, Dargahi L. Harmonic activity of glutamate dehydrogenase and neuroplasticity: The impact on aging, cognitive dysfunction, and neurodegeneration. Behav Brain Res 2025; 480:115399. [PMID: 39675635 DOI: 10.1016/j.bbr.2024.115399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
In recent years, glutamate has attracted significant attention for its roles in various brain processes. However, one of its key regulators, glutamate dehydrogenase (GDH), remains understudied despite its pivotal role in several biochemical pathways. Dysfunction or dysregulation of GDH has been implicated in aging and various neurological disorders, such as Alzheimer's disease and Parkinson's disease. In this review, the impact of GDH on aging, cognitive impairment, and neurodegenerative conditions, as exemplars of the phenomena that may affected by neuroplasticity, has been reviewed. Despite extensive research on synaptic plasticity, the precise influence of GDH on brain structure and function remains undiscovered. This review of existing literature on GDH and neuroplasticity reveals diverse and occasionally conflicting effects. Future research endeavors should aim to describe the precise mechanisms by which GDH influences neuroplasticity (eg. synaptic plasticity and neurogenesis), particularly in the context of human aging and disease progression. Studies on GDH activity have been limited by factors such as insufficient sample sizes and varying experimental conditions. Researchers should focus on investigating the molecular mechanisms by which GDH modulates neuroplasticity, utilizing various animal strains and species, ages, sexes, GDH isoforms, brain regions, and cell types. Understanding GDH's role in neuroplasticity may offer innovative therapeutic strategies for neurodegenerative and psychiatric diseases, potentially slowing the aging process and promoting brain regeneration.
Collapse
Affiliation(s)
- Shakiba Salarvandian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Digaleh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pegah Javadpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sareh Asadi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ali Orang Zaman
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Choi H, An YK, Lee CJ, Song CU, Kim EJ, Lee CE, Cho SJ, Eyun SI. Genome assembly, gene content, and plastic gene expression responses to salinity changes in the Brackishwater Clam (Corbicula japonica) from a dynamic estuarine environment. JOURNAL OF HAZARDOUS MATERIALS 2025; 483:136627. [PMID: 39616841 DOI: 10.1016/j.jhazmat.2024.136627] [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: 07/01/2024] [Revised: 11/01/2024] [Accepted: 11/21/2024] [Indexed: 01/28/2025]
Abstract
Estuaries are dynamic transition zones between marine and freshwater environments, where salinity varies greatly on spatial and temporal scales. The temporal salinity fluctuations of these habitats require organisms to rapidly regulate ionic concentrations and osmotic pressure to survive in these dynamic conditions. Understanding the extent of plasticity of euryhaline animals is vital for predicting their responses and resilience to salinity change. We generated the first high-resolution genome and transcriptome sequences of C. japonica. In comparison with 11 other molluscan genomes, the C. japonica genome displayed striking expansions of putative neuron-related genes and gene families. The involvement of these genes in the glutamate/GABA-glutamine and glycine cycle suggests a possible contribution to the excitation of neuronal networks, particularly under high salinity conditions. This study contributes to our understanding of mechanisms underlying the rapid responses of estuarine species to changing conditions and raises many intriguing hypotheses and questions for future investigation.
Collapse
Affiliation(s)
- Hyeongwoo Choi
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Yun Keun An
- Division of Marine Technology, Chonnam National University, Yeosu 59626, Korea
| | - Chan-Jun Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| | - Chi-Une Song
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Eun-Jeong Kim
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Carol Eunmi Lee
- Department of Integrative Biology, University of Wisconsin, Madison, WI 53706, USA
| | - Sung-Jin Cho
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju 28644, Korea.
| | - Seong-Il Eyun
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea.
| |
Collapse
|
4
|
Ilyaskina D, Nakadera Y, Lamoree MH, Koene JM, Leonards PEG. Impact of fluoxetine exposure on Lymnaea stagnalis and its developing eggs: integrating untargeted lipidomics, targeted metabolomics, and classical risk assessment. Front Pharmacol 2025; 16:1536438. [PMID: 39968180 PMCID: PMC11832466 DOI: 10.3389/fphar.2025.1536438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Pharmaceuticals such as selective serotonin reuptake inhibitors (SSRIs), are increasingly detected in aquatic environments, posing potential risks to non-target organisms, because many of those substances are widely shared neuromodulator. In this study, we investigated the effects of SSRI antidepressant, namely, fluoxetine, exposure on the freshwater snail L. stagnalis, focusing on egg development, neurochemical pathways, and lipid metabolism. Snails were exposed to a range of 51-434 µg fluoxetine L⁻1 for 7 days, followed by analysis of survival, feeding behaviour, reproduction, and metabolomic changes in the central nervous system (CNS), albumen gland, and eggs. Although no significant effects were observed on survival or fecundity, fluoxetine exposure significantly impaired egg development in a dose-dependent manner, reducing hatching rates with an EC50 of 126 µg fluoxetine L⁻1. Removal of eggs from the contaminated environment partially reversed these developmental effects, suggesting potential recovery if fluoxetine levels decrease. Molecular analysis revealed several neurochemical and lipidomic alterations. In the CNS, elevated levels of catecholamines, phosphatidylcholines (PC), and ceramides were linked to disruptions in neurotransmission, membrane integrity, and impaired embryo development. In the albumen gland, we detected a decrease of key lipid classes, including sphingomyelins and fatty acids, which can be linked with impaired egg quality. Additionally, a decrease in histamine in both the albumen gland and eggs suggested further disruption of egg development, potentially affecting metamorphosis success. Moreover, the dose-dependent increase in choline, along with PC and oxidized PC, indicated oxidative stress and lipid peroxidation in the CNS and exposed eggs of Lymnaea stagnalis. Our findings highlight the benefits of combining behavioral assessments with metabolomic profiling to better understand the mechanistic pathways underlying fluoxetine's adverse effects.
Collapse
Affiliation(s)
- Diana Ilyaskina
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Yumi Nakadera
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Senckenberg Ocean Species Alliance, Senckenberg Research Institute and Natural History Museum Frankfurt, Frankfurt, Germany
| | - Marja H. Lamoree
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Joris M. Koene
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Pim E. G. Leonards
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
5
|
Li M, Yuan H, Yang X, Lei Y, Lian J. Glutamine-glutamate centered metabolism as the potential therapeutic target against Japanese encephalitis virus-induced encephalitis. Cell Biosci 2025; 15:6. [PMID: 39844330 PMCID: PMC11755858 DOI: 10.1186/s13578-024-01340-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Japanese encephalitis (JE) induced by Japanese encephalitis virus (JEV) infection is the most prevalent diagnosed epidemic viral encephalitis globally. The underlying pathological mechanisms remain largely unknown. Given that viruses are obligate intracellular parasites, cellular metabolic reprogramming triggered by viral infection is intricately related to the establishment of infection and progression of disease. Therefore, uncovering and manipulating the metabolic reprogramming that underlies viral infection will help elucidate the pathogenic mechanisms and develop novel therapeutic strategies. METHODS Metabolomics analysis was performed to comprehensively delineate the metabolic profiles in JEV-infected mice brains and neurons. Metabolic flux analysis, quantitative real-time PCR, western blotting and fluorescence immunohistochemistry were utilized to describe detailed glutamine-glutamate metabolic profiles during JEV infection. Exogenous addition of metabolites and associated compounds and RNA interference were employed to manipulate glutamine-glutamate metabolism to clarify its effects on viral replication. The survival rate, severity of neuroinflammation, and levels of viral replication were assessed to determine the efficacy of glutamine supplementation in JEV-challenged mice. RESULTS Here, we have delineated a novel perspective on the pathogenesis of JE by identifying an aberrant low flux in glutamine-glutamate metabolism both in vivo and in vitro, which was critical in the establishment of JEV infection and progression of JE. The perturbed glutamine-glutamate metabolism induced neurotransmitter imbalance and created an immune-inhibitory state with increased gamma-aminobutyric acid/glutamate ratio, thus facilitating efficient viral replication both in JEV-infected neurons and the brain of JEV-infected mice. In addition, viral infection restrained the utilization of glutamine via the glutamate-α-ketoglutaric acid axis in neurons, thus avoiding the adverse effects of glutamine oxidation on viral propagation. As the conversion of glutamine to glutamate was inhibited after JEV infection, the metabolism of glutathione (GSH) was simultaneously impaired, exacerbating oxidative stress in JEV-infected neurons and mice brains and promoting the progression of JE. Importantly, the supplementation of glutamine in vivo alleviated the intracranial inflammation and enhanced the survival of JEV-challenged mice. CONCLUSION Altogether, our study highlights an aberrant glutamine-glutamate metabolism during JEV infection and unveils how this facilitates viral replication and promotes JE progression. Manipulation of these metabolic alterations may potentially be exploited to develop therapeutic approaches for JEV infection.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Hang Yuan
- Pathogenic Biology, Medical College of Yan'an University, Yan'an, 716000, China
| | - Xiaofei Yang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yingfeng Lei
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China.
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
6
|
Selvaraj S, Weerasinghe L. The Role of Nanotechnology in Understanding the Pathophysiology of Traumatic Brain Injury. Cent Nerv Syst Agents Med Chem 2025; 25:20-38. [PMID: 38676493 DOI: 10.2174/0118715249291999240418112531] [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: 11/09/2023] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024]
Abstract
Recently, traumatic brain injury (TBI) has been a growing disorder due to frequent brain dysfunction. The Glasgow Coma Scale expresses TBI as classified as having mild, moderate, or severe brain effects, according to the effects on the brain. Brain receptors undergo various modifications in their pathology through chemical synaptic pathways, leading to depression, Alzheimer's, and Parkinson's disease. These brain disorders can be controlled using central receptors such as dopamine, glutamate, and γ-aminobutyric acid, which are clearly explained in this review. Furthermore, there are many complications in TBI's clinical trials and diagnostics, leading to insignificant treatment, causing permanent neuro-damage, physical disability, and even death. Bio-screening and conventional molecular-based therapies are inappropriate due to poor preclinical testing and delayed recovery. Hence, modern nanotechnology utilizing nanopulsed laser therapy and advanced nanoparticle insertion will be suitable for TBI's diagnostics and treatment. In recent days, nanotechnology has an important role in TBI control and provides a higher success rate than conventional therapies. This review highlights the pathophysiology of TBI by comprising the drawbacks of conventional techniques and supports suitable modern alternates for treating TBI.
Collapse
Affiliation(s)
- Saranya Selvaraj
- Department of Chemistry, Faculty of Applied sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Laksiri Weerasinghe
- Department of Chemistry, Faculty of Applied sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| |
Collapse
|
7
|
Brandão‐Teles C, Carregari VC, Reis‐de‐Oliveira G, Smith BJ, Chaves Y, Sousa Santos AV, Pinheiro EMDC, Oliveira CC, Vieira AS, Crunfli F, Martins‐de‐Souza D. Impacts of hnRNP A1 Splicing Inhibition on the Brain Remyelination Proteome. J Neurochem 2025; 169:e16304. [PMID: 39840781 PMCID: PMC11752419 DOI: 10.1111/jnc.16304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/26/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025]
Abstract
Oligodendrocytes, the myelinating cells in the central nervous system, are implicated in several neurological disorders marked by dysfunctional RNA-binding proteins (RBPs). The present study aimed at investigating the role of hnRNP A1 in the proteome of the corpus callosum, prefrontal cortex, and hippocampus of a murine cuprizone-induced demyelination model. Right after the cuprizone insult, we administered an hnRNP A1 splicing activity inhibitor and analyzed its impact on brain remyelination by nanoESI-LC-MS/MS label-free proteomic analysis to assess the biological processes affected in these brain regions. Significant alterations in essential myelination proteins highlighted the involvement of hnRNP A1 in maintaining myelin integrity. Pathways related to sphingolipid and endocannabinoid signaling were affected, as well as the synaptic vesicle cycle and GABAergic synapses. Although behavioral impairments were not observed, molecular changes suggest potential links to memory, synaptic function, and neurotransmission processes. These findings enhance our understanding of the multifaceted roles of hnRNP A1 in the central nervous system, providing valuable insights for future investigations and therapeutic interventions in neurodegenerative and demyelinating diseases.
Collapse
Affiliation(s)
- Caroline Brandão‐Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Victor Corasolla Carregari
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Guilherme Reis‐de‐Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Bradley J. Smith
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Yane Chaves
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Aline Valéria Sousa Santos
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | | | - Caio C. Oliveira
- Institute of ChemistryUniversity of CampinasCampinasSão PauloBrazil
| | - Andre Schwambach Vieira
- Laboratory of Electrophysiology, Neurobiology and Behaviour, Department of Functional and Structural Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
| | - Fernanda Crunfli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Daniel Martins‐de‐Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of BiologyUniversity of CampinasCampinasBrazil
- Experimental Medicine Research Cluster (EMRC)University of CampinasCampinasSPBrazil
- D'Or Institute for Research and Education (IDOR)São PauloBrazil
- Instituto Nacional de Biomarcadores Em Neuropsiquiatria, Conselho Nacional de Desenvolvimento Científico e TecnológicoSão PauloBrazil
- INCT in Modelling Human Complex Diseases With 3D Platforms (Model3D), Conselho Nacional de Desenvolvimento Científico e TecnológicoSão PauloBrazil
| |
Collapse
|
8
|
Zhang Y, Wang Z, Xu F, Liu Z, Zhao Y, Yang LZ, Fang W. Progress of Astrocyte-Neuron Crosstalk in Central Nervous System Diseases. Neurochem Res 2024; 49:3187-3207. [PMID: 39292330 DOI: 10.1007/s11064-024-04241-6] [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: 07/05/2024] [Revised: 08/08/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
Neurons are the primary cells responsible for information processing in the central nervous system (CNS). However, they are vulnerable to damage and insult in a variety of neurological disorders. As the most abundant glial cells in the brain, astrocytes provide crucial support to neurons and participate in synapse formation, synaptic transmission, neurotransmitter recycling, regulation of metabolic processes, and the maintenance of the blood-brain barrier integrity. Though astrocytes play a significant role in the manifestation of injury and disease, they do not work in isolation. Cellular interactions between astrocytes and neurons are essential for maintaining the homeostasis of the CNS under both physiological and pathological conditions. In this review, we explore the diverse interactions between astrocytes and neurons under physiological conditions, including the exchange of neurotrophic factors, gliotransmitters, and energy substrates, and different CNS diseases such as Alzheimer's disease, Parkinson's disease, stroke, traumatic brain injury, and multiple sclerosis. This review sheds light on the contribution of astrocyte-neuron crosstalk to the progression of neurological diseases to provide potential therapeutic targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Ziyu Wang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Fenglian Xu
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Zijun Liu
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Yu Zhao
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China
| | - Lele Zixin Yang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, PA, 19107, USA
| | - Weirong Fang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, 210009, P. R. China.
| |
Collapse
|
9
|
Gateva P, Hristov M, Ivanova N, Vasileva D, Ivanova A, Sabit Z, Bogdanov T, Apostolova S, Tzoneva R. Antinociceptive Behavior, Glutamine/Glutamate, and Neopterin in Early-Stage Streptozotocin-Induced Diabetic Neuropathy in Liraglutide-Treated Mice under a Standard or Enriched Environment. Int J Mol Sci 2024; 25:10786. [PMID: 39409118 PMCID: PMC11477071 DOI: 10.3390/ijms251910786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
Diabetic neuropathy (DN) is a common complication of long-lasting type 1 and type 2 diabetes, with no curative treatment available. Here, we tested the effect of the incretin mimetic liraglutide in DN in mice with early-stage type 1 diabetes bred in a standard laboratory or enriched environment. With a single i.p. injection of streptozotocin 150 mg/kg, we induced murine diabetes. Liraglutide (0.4 mg/kg once daily, i.p. for ten days since the eighth post-streptozotocin day) failed to decrease the glycemia in the diabetic mice; however, it alleviated their antinociceptive behavior, as tested with formalin. The second phase of the formalin test had significantly lower results in liraglutide-treated mice reared in the enriched environment vs. liraglutide-treated mice under standard conditions [2.00 (0.00-11.00) vs. 29.00 (2.25-41.50) s, p = 0.016]. Liraglutide treatment, however, decreased the threshold of reactivity in the von Fray test. A significantly higher neopterin level was demonstrated in the diabetic control group compared to treatment-naïve controls and the liraglutide-treated diabetic mice (p < 0.001). The glutamine/glutamate ratio in both liraglutide-treated groups, either reared under standard conditions (p = 0.003) or an enriched environment (p = 0.002), was significantly higher than in the diabetic controls. This study demonstrates an early liraglutide effect on pain sensation in two streptozotocin-induced diabetes mouse models by reducing some inflammatory and oxidative stress parameters.
Collapse
Affiliation(s)
- Pavlina Gateva
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria; (M.H.); (N.I.); (D.V.); (A.I.)
| | - Milen Hristov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria; (M.H.); (N.I.); (D.V.); (A.I.)
| | - Natasha Ivanova
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria; (M.H.); (N.I.); (D.V.); (A.I.)
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Debora Vasileva
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria; (M.H.); (N.I.); (D.V.); (A.I.)
| | - Alexandrina Ivanova
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria; (M.H.); (N.I.); (D.V.); (A.I.)
| | - Zafer Sabit
- Department of Pathophysiology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Todor Bogdanov
- Department of Medical Physics and Biophysics, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Sonia Apostolova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (S.A.); (R.T.)
| | - Rumiana Tzoneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (S.A.); (R.T.)
| |
Collapse
|
10
|
Jiang G, Wang C, Wang Y, Wang J, Xue Y, Lin Y, Hu X, Lv Y. Exogenous putrescine plays a switch-like influence on the pH stress adaptability of biofilm-based activated sludge. Appl Environ Microbiol 2024; 90:e0056924. [PMID: 38916292 PMCID: PMC11267902 DOI: 10.1128/aem.00569-24] [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: 03/26/2024] [Accepted: 06/04/2024] [Indexed: 06/26/2024] Open
Abstract
Microbial community adaptability to pH stress plays a crucial role in biofilm formation. This study aims to investigate the regulatory mechanisms of exogenous putrescine on pH stress, as well as enhance understanding and application for the technical measures and molecular mechanisms of biofilm regulation. Findings demonstrated that exogenous putrescine acted as a switch-like distributor affecting microorganism pH stress, thus promoting biofilm formation under acid conditions while inhibiting it under alkaline conditions. As pH decreases, the protonation degree of putrescine increases, making putrescine more readily adsorbed. Protonated exogenous putrescine could increase cell membrane permeability, facilitating its entry into the cell. Subsequently, putrescine consumed intracellular H+ by enhancing the glutamate-based acid resistance strategy and the γ-aminobutyric acid metabolic pathway to reduce acid stress on cells. Furthermore, putrescine stimulated ATPase expression, allowing for better utilization of energy in H+ transmembrane transport and enhancing oxidative phosphorylation activity. However, putrescine protonation was limited under alkaline conditions, and the intracellular H+ consumption further exacerbated alkali stress and inhibits cellular metabolic activity. Exogenous putrescine promoted the proportion of fungi and acidophilic bacteria under acidic stress and alkaliphilic bacteria under alkali stress while having a limited impact on fungi in alkaline biofilms. Increasing Bdellovibrio under alkali conditions with putrescine further aggravated the biofilm decomposition. This research shed light on the unclear relationship between exogenous putrescine, environmental pH, and pH stress adaptability of biofilm. By judiciously employing putrescine, biofilm formation could be controlled to meet the needs of engineering applications with different characteristics.IMPORTANCEThe objective of this study is to unravel the regulatory mechanism by which exogenous putrescine influences biofilm pH stress adaptability and understand the role of environmental pH in this intricate process. Our findings revealed that exogenous putrescine functioned as a switch-like distributor affecting the pH stress adaptability of biofilm-based activated sludge, which promoted energy utilization for growth and reproduction processes under acidic conditions while limiting biofilm development to conserve energy under alkaline conditions. This study not only clarified the previously ambiguous relationship between exogenous putrescine, environmental pH, and biofilm pH stress adaptability but also offered fresh insights into enhancing biofilm stability within extreme environments. Through the modulation of energy utilization, exerting control over biofilm growth and achieving more effective engineering goals could be possible.
Collapse
Affiliation(s)
- Guanyu Jiang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| | - Can Wang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| | - Yongchao Wang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| | - Jiayi Wang
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| | - Yimei Xue
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| | - Yuting Lin
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| | - Xurui Hu
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| | - Yahui Lv
- School of Environmental Science and Engineering, Tianjin University, Tianjin, China
- Tianjin Key Lab of Indoor Air Environmental Quality Control, Tianjin, China
| |
Collapse
|
11
|
Wei L, Chen S, Deng X, Liu Y, Wang H, Gao X, Huang Y. Metabolomic discoveries for early diagnosis and traditional Chinese medicine efficacy in ischemic stroke. Biomark Res 2024; 12:63. [PMID: 38902829 PMCID: PMC11188286 DOI: 10.1186/s40364-024-00608-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
Ischemic stroke (IS), a devastating cerebrovascular accident, presents with high mortality and morbidity. Following IS onset, a cascade of pathological changes, including excitotoxicity, inflammatory damage, and blood-brain barrier disruption, significantly impacts prognosis. However, current clinical practices struggle with early diagnosis and identifying these alterations. Metabolomics, a powerful tool in systems biology, offers a promising avenue for uncovering early diagnostic biomarkers for IS. By analyzing dynamic metabolic profiles, metabolomics can not only aid in identifying early IS biomarkers but also evaluate Traditional Chinese Medicine (TCM) efficacy and explore its mechanisms of action in IS treatment. Animal studies demonstrate that TCM interventions modulate specific metabolite levels, potentially reflecting their therapeutic effects. Identifying relevant metabolites in cerebral ischemia patients holds immense potential for early diagnosis and improved outcomes. This review focuses on recent metabolomic discoveries of potential early diagnostic biomarkers for IS. We explore variations in metabolites observed across different ages, genders, disease severity, and stages. Additionally, the review examines how specific TCM extracts influence IS development through metabolic changes, potentially revealing their mechanisms of action. Finally, we emphasize the importance of integrating metabolomics with other omics approaches for a comprehensive understanding of IS pathophysiology and TCM efficacy, paving the way for precision medicine in IS management.
Collapse
Affiliation(s)
- Liangzhe Wei
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Siqi Chen
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China
| | - Xinpeng Deng
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Yuchun Liu
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Haifeng Wang
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Xiang Gao
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China.
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China.
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China.
| | - Yi Huang
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China.
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China.
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China.
| |
Collapse
|
12
|
Cataldo PG, Urquiza Martínez MP, Villena J, Kitazawa H, Saavedra L, Hebert EM. Comprehensive characterization of γ-aminobutyric acid (GABA) production by Levilactobacillus brevis CRL 2013: insights from physiology, genomics, and proteomics. Front Microbiol 2024; 15:1408624. [PMID: 38962125 PMCID: PMC11219586 DOI: 10.3389/fmicb.2024.1408624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024] Open
Abstract
Introduction Levilactobacillus brevis CRL 2013, a plant-derived lactic acid bacterium (LAB) with immunomodulatory properties, has emerged as an efficient producer of γ-aminobutyric acid (GABA). Notably, not all LAB possess the ability to produce GABA, highlighting the importance of specific genetic and environmental conditions for GABA synthesis. This study aimed to elucidate the intriguing GABA-producing machinery of L. brevis CRL 2013 and support its potential for safe application through comprehensive genome analysis. Methods A comprehensive genome analysis of L. brevis CRL 2013 was performed to identify the presence of antibiotic resistance genes, virulence markers, and genes associated with the glutamate decarboxylase system, which is essential for GABA biosynthesis. Then, an optimized chemically defined culture medium (CDM) was supplemented with monosodium glutamate (MSG) and yeast extract (YE) to analyze their influence on GABA production. Proteomic and transcriptional analyses were conducted to assess changes in protein and gene expression related to GABA production. Results The absence of antibiotic resistance genes and virulence markers in the genome of L. brevis CRL 2013 supports its safety for potential probiotic applications. Genes encoding the glutamate decarboxylase system, including two gad genes (gadA and gadB) and the glutamate antiporter gene (gadC), were identified. The gadB gene is located adjacent to gadC, while gadA resides separately on the chromosome. The transcriptional regulator gadR was found upstream of gadC, with transcriptional analyses demonstrating cotranscription of gadR with gadC. Although MSG supplementation alone did not activate GABA synthesis, the addition of YE significantly enhanced GABA production in the optimized CDM containing glutamate. Proteomic analysis revealed minimal differences between MSG-supplemented and non-supplemented CDM cultures, whereas YE supplementation resulted in significant proteomic changes, including upregulation of GadB. Transcriptional analysis confirmed increased expression of gadB and gadR upon YE supplementation, supporting its role in activating GABA production. Conclusion These findings provide valuable insights into the influence of nutrient composition on GABA production. Furthermore, they unveil the potential of L. brevis CRL 2013 as a safe, nonpathogenic strain with valuable biotechnological traits which can be further leveraged for its probiotic potential in the food industry.
Collapse
Affiliation(s)
- Pablo G. Cataldo
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | | | - Julio Villena
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Lucila Saavedra
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Elvira M. Hebert
- Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina
| |
Collapse
|
13
|
Bae J, Moniruzzaman M, Je HW, Lee S, Choi W, Min T, Kim KW, Bai SC. Evaluation of Gamma-Aminobutyric Acid (GABA) as a Functional Feed Ingredient on Growth Performance, Immune Enhancement, and Disease Resistance in Olive Flounder ( Paralichthys olivaceus) under High Stocking Density. Antioxidants (Basel) 2024; 13:647. [PMID: 38929086 PMCID: PMC11201082 DOI: 10.3390/antiox13060647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is a non-protein amino acid that is found in the brain and central nervous system of animals as an inhibitory neurotransmitter. It has been shown to have a variety of physiological functions, including stress reduction and immune enhancement. This study investigated the effects of dietary supplementation with GABA on growth, serum biochemistry, innate immunity, and disease resistance in juvenile olive flounders (Paralichthys olivaceus) challenged with Edwardsiella tarda under high-stocking density. A control diet and three experimental diets were prepared, with 150 mg/kg (GABA150), 200 mg/kg (GABA200), and 250 mg/kg (GABA250) of GABA added to each diet, respectively. Each experimental diet was fed to olive flounders in triplicate with an initial weight of 12.75 g ± 0.3 g in 40 L tanks at two stocking densities: normal density (20 fish/tank) and high density (40 fish/tank). After 8 weeks of the feeding trial, growth, feed utilization, whole-body proximate compositions, blood analyses, and non-specific immune responses were measured, and challenge tests were performed. There were no significant differences in the weight gain (WG) and specific growth rate (SGR) among fish fed the GABA-supplemented diets at the two stocking densities. However, the normal-density groups showed significantly higher WG and SGR than the high-density groups (p < 0.05). There was no significant difference in feed efficiency and protein efficiency ratio among all groups. Moreover, there was no significant difference in the whole-body proximate composition analysis (p > 0.05). There were no significant differences in cortisol levels in fish fed the GABA at both densities, but the high-density group showed a significantly higher cortisol than the low-density group. Blood GABA significantly increased in a dose-dependent manner regardless of the density groups (p < 0.05). Superoxide dismutase activity showed significantly higher levels than the control group, but there was no significant effect of the stocking densities in fish fed the GABA diets (p < 0.05). Myeloperoxidase activities in fish fed the GABA200 and GABA250 diets showed significantly higher levels at both of the stocking densities (p < 0.05). Lysozyme activity was significantly higher in the GABA150 group than in the CON, GABA200, and GABA250 groups (p < 0.05). After 15 days of challenge tests with Edwardsiella tarda, the cumulative survival rates of the GABA150, GABA200, and GABA250 groups were significantly higher than that of the CON group (p < 0.05). The results suggested that the optimal dietary GABA level for juvenile olive flounder culture is 150 mg/kg, regardless of rearing density, to enhance growth, immunity, and disease resistance.
Collapse
Affiliation(s)
- Jinho Bae
- Aquafeed Research Center, National Institute of Fisheries Science, Pohang 37517, Republic of Korea; (J.B.); (K.-W.K.)
| | - Mohammad Moniruzzaman
- Department of Animal Biotechnology, Jeju International Animal Research Center (JIA), Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju 63243, Republic of Korea;
| | - Hyeong-Woo Je
- Department of Fisheries Biology, Pukyong National University, 45, Yongso-ro, Nam-gu, Busan 48513, Republic of Korea;
| | - Seunghan Lee
- Department of Aquaculture and Aquatic Science, Kunsan National University, Gunsan 54150, Republic of Korea;
| | - Wonsuk Choi
- CJ Feed & Care, AN R&D Center, 170, Eulji-ro, Jung-gu, Seoul 04548, Republic of Korea;
| | - Taesun Min
- Department of Animal Biotechnology, Bio-Resources Computing Research Center, Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju 63243, Republic of Korea;
| | - Kang-Woong Kim
- Aquafeed Research Center, National Institute of Fisheries Science, Pohang 37517, Republic of Korea; (J.B.); (K.-W.K.)
| | - Sungchul C. Bai
- Feeds & Foods Nutrition Research Center, Pukyong National University, Busan 48547, Republic of Korea
| |
Collapse
|
14
|
Benarroch E. What Is the Role of Lactate in Brain Metabolism, Plasticity, and Neurodegeneration? Neurology 2024; 102:e209378. [PMID: 38574305 DOI: 10.1212/wnl.0000000000209378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
|
15
|
Li D, Gao X, Ma X, Wang M, Cheng C, Xue T, Gao F, Shen Y, Zhang J, Liu Q. Aging-induced tRNA Glu-derived fragment impairs glutamate biosynthesis by targeting mitochondrial translation-dependent cristae organization. Cell Metab 2024; 36:1059-1075.e9. [PMID: 38458203 DOI: 10.1016/j.cmet.2024.02.011] [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: 05/14/2023] [Revised: 10/31/2023] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Mitochondrial cristae, infoldings of the mitochondrial inner membrane, undergo aberrant changes in their architecture with age. However, the underlying molecular mechanisms and their contribution to brain aging are largely elusive. Here, we observe an age-dependent accumulation of Glu-5'tsRNA-CTC, a transfer-RNA-derived small RNA (tsRNA), derived from nuclear-encoded tRNAGlu in the mitochondria of glutaminergic neurons. Mitochondrial Glu-5'tsRNA-CTC disrupts the binding of mt-tRNALeu and leucyl-tRNA synthetase2 (LaRs2), impairing mt-tRNALeu aminoacylation and mitochondria-encoded protein translation. Mitochondrial translation defects disrupt cristae organization, leading to damaged glutaminase (GLS)-dependent glutamate formation and reduced synaptosomal glutamate levels. Moreover, reduction of Glu-5'tsRNA-CTC protects aged brains from age-related defects in mitochondrial cristae organization, glutamate metabolism, synaptic structures, and memory. Thus, beyond illustrating a physiological role for normal mitochondrial cristae ultrastructure in maintaining glutamate levels, our study defines a pathological role for tsRNAs in brain aging and age-related memory decline.
Collapse
Affiliation(s)
- Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xinyi Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaolin Ma
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ming Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chuandong Cheng
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tian Xue
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Feng Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Yong Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China; Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
16
|
Witkin JM, Shafique H, Cerne R, Smith JL, Marini AM, Lipsky RH, Delery E. Mechanistic and therapeutic relationships of traumatic brain injury and γ-amino-butyric acid (GABA). Pharmacol Ther 2024; 256:108609. [PMID: 38369062 DOI: 10.1016/j.pharmthera.2024.108609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
Traumatic brain injury (TBI) is a highly prevalent medical condition for which no medications specific for the prophylaxis or treatment of the condition as a whole exist. The spectrum of symptoms includes coma, headache, seizures, cognitive impairment, depression, and anxiety. Although it has been known for years that the inhibitory neurotransmitter γ-amino-butyric acid (GABA) is involved in TBI, no novel therapeutics based upon this mechanism have been introduced into clinical practice. We review the neuroanatomical, neurophysiological, neurochemical, and neuropharmacological relationships of GABA neurotransmission to TBI with a view toward new potential GABA-based medicines. The long-standing idea that excitatory and inhibitory (GABA and others) balances are disrupted by TBI is supported by the experimental data but has failed to invent novel methods of restoring this balance. The slow progress in advancing new treatments is due to the complexity of the disorder that encompasses multiple dynamically interacting biological processes including hemodynamic and metabolic systems, neurodegeneration and neurogenesis, major disruptions in neural networks and axons, frank brain lesions, and a multitude of symptoms that have differential neuronal and neurohormonal regulatory mechanisms. Although the current and ongoing clinical studies include GABAergic drugs, no novel GABA compounds are being explored. It is suggested that filling the gap in understanding the roles played by specific GABAA receptor configurations within specific neuronal circuits could help define new therapeutic approaches. Further research into the temporal and spatial delivery of GABA modulators should also be useful. Along with GABA modulation, research into the sequencing of GABA and non-GABA treatments will be needed.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | | | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA; Department of Anatomy and Cell Biology, Indiana University/Purdue University, Indianapolis, IN, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA
| | - Ann M Marini
- Department of Neurology, Program in Neuroscience, and Molecular and Cellular Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert H Lipsky
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Elizabeth Delery
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA.
| |
Collapse
|
17
|
Pushchina EV, Bykova ME, Varaksin AA. Post-Traumatic Expressions of Aromatase B, Glutamine Synthetase, and Cystathionine-Beta-Synthase in the Cerebellum of Juvenile Chum Salmon, Oncorhynchus keta. Int J Mol Sci 2024; 25:3299. [PMID: 38542274 PMCID: PMC10970380 DOI: 10.3390/ijms25063299] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/11/2024] [Accepted: 03/11/2024] [Indexed: 04/29/2025] Open
Abstract
In adult fish, neurogenesis occurs in many areas of the brain, including the cerebellum, with the ratio of newly formed cells relative to the total number of brain cells being several orders of magnitude greater than in mammals. Our study aimed to compare the expressions of aromatase B (AroB), glutamine synthetase (GS), and cystathionine-beta-synthase (CBS) in the cerebellum of intact juvenile chum salmon, Oncorhynchus keta. To identify the dynamics that determine the involvement of AroB, GS, and CBS in the cellular mechanisms of regeneration, we performed a comprehensive assessment of the expressions of these molecular markers during a long-term primary traumatic brain injury (TBI) and after a repeated acute TBI to the cerebellum of O. keta juveniles. As a result, in intact juveniles, weak or moderate expressions of AroB, GS, and CBS were detected in four cell types, including cells of the neuroepithelial type, migrating, and differentiated cells (graphic abstract, A). At 90 days post injury, local hypercellular areas were found in the molecular layer containing moderately labeled AroB+, GS+, and CBS+ cells of the neuroepithelial type and larger AroB+, GS+, and CBS+ cells (possibly analogous to the reactive glia of mammals); patterns of cells migration and neovascularization were also observed. A repeated TBI caused the number of AroB+, GS+, and CBS+ cells to further increase; an increased intensity of immunolabeling was recorded from all cell types (graphic abstract, C). Thus, the results of this study provide a better understanding of adult neurogenesis in teleost fishes, which is expected to clarify the issue of the reactivation of adult neurogenesis in mammalian species.
Collapse
Affiliation(s)
- Evgeniya V. Pushchina
- Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia; (M.E.B.); (A.A.V.)
| | | | | |
Collapse
|
18
|
Hu E, Tang T, Li Y, Li T, Zhu L, Ding R, Wu Y, Huang Q, Zhang W, Wu Q, Wang Y. Spatial amine metabolomics and histopathology reveal localized brain alterations in subacute traumatic brain injury and the underlying mechanism of herbal treatment. CNS Neurosci Ther 2024; 30:e14231. [PMID: 37183394 PMCID: PMC10915989 DOI: 10.1111/cns.14231] [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] [Indexed: 05/16/2023] Open
Abstract
INTRODUCTION Spatial changes of amine metabolites and histopathology of the whole brain help to reveal the mechanism of traumatic brain injury (TBI) and treatment. METHODS A newly developed liquid microjunction surface sampling-tandem mass tag-ultra performance liquid chromatography-mass spectrometry technique is applied to profile brain amine metabolites in five brain regions after impact-induced TBI at the subacute stage. H&E, Nissl, and immunofluorescence staining are performed to spatially correlate microscopical changes to metabolic alterations. Then, bioinformatics, molecular docking, ELISA, western blot, and immunofluorescence are integrated to uncover the mechanism of Xuefu Zhuyu decoction (XFZYD) against TBI. RESULTS Besides the hippocampus and cortex, the thalamus, caudate-putamen, and fiber tracts also show differentiated metabolic changes between the Sham and TBI groups. Fourteen amine metabolites (including isomers such as L-leucine and L-isoleucine) are significantly altered in specific regions. The metabolic changes are well matched with the degree of neuronal damage, glia activation, and neurorestoration. XFZYD reverses the dysregulation of several amine metabolites, such as hippocampal Lys-Phe/Phe-Lys and dopamine. Also, XFZYD enhances post-TBI angiogenesis in the hippocampus and the thalamus. CONCLUSION This study reveals the local amine-metabolite and histological changes in the subacute stage of TBI. XFZYD may promote TBI recovery by normalizing amine metabolites and spatially promoting dopamine production and angiogenesis.
Collapse
Affiliation(s)
- En Hu
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Tao Tang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - You‐mei Li
- College of Chemistry and Chemical EngineeringCentral South UniversityChangshaHunanChina
| | - Teng Li
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Lin Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Ruo‐qi Ding
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Yao Wu
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Qing Huang
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
- Department of NeurologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Qian Wu
- College of Chemistry and Chemical EngineeringCentral South UniversityChangshaHunanChina
| | - Yang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
19
|
Martin SP, Leeman-Markowski BA. Proposed mechanisms of tau: relationships to traumatic brain injury, Alzheimer's disease, and epilepsy. Front Neurol 2024; 14:1287545. [PMID: 38249745 PMCID: PMC10797726 DOI: 10.3389/fneur.2023.1287545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Traumatic brain injury (TBI), Alzheimer's disease (AD), and epilepsy share proposed mechanisms of injury, including neuronal excitotoxicity, cascade signaling, and activation of protein biomarkers such as tau. Although tau is typically present intracellularly, in tauopathies, phosphorylated (p-) and hyper-phosphorylated (hp-) tau are released extracellularly, the latter leading to decreased neuronal stability and neurofibrillary tangles (NFTs). Tau cleavage at particular sites increases susceptibility to hyper-phosphorylation, NFT formation, and eventual cell death. The relationship between tau and inflammation, however, is unknown. In this review, we present evidence for an imbalanced endoplasmic reticulum (ER) stress response and inflammatory signaling pathways resulting in atypical p-tau, hp-tau and NFT formation. Further, we propose tau as a biomarker for neuronal injury severity in TBI, AD, and epilepsy. We present a hypothesis of tau phosphorylation as an initial acute neuroprotective response to seizures/TBI. However, if the underlying seizure pathology or TBI recurrence is not effectively treated, and the pathway becomes chronically activated, we propose a "tipping point" hypothesis that identifies a transition of tau phosphorylation from neuroprotective to injurious. We outline the role of amyloid beta (Aβ) as a "last ditch effort" to revert the cell to programmed death signaling, that, when fails, transitions the mechanism from injurious to neurodegenerative. Lastly, we discuss targets along these pathways for therapeutic intervention in AD, TBI, and epilepsy.
Collapse
Affiliation(s)
- Samantha P. Martin
- Comprehensive Epilepsy Center, New York University Langone Health, New York, NY, United States
- Department of Neurology, New York University Langone Health, New York, NY, United States
- New York University Grossman School of Medicine, New York, NY, United States
- VA New York Harbor Healthcare System, New York, NY, United States
| | - Beth A. Leeman-Markowski
- Comprehensive Epilepsy Center, New York University Langone Health, New York, NY, United States
- Department of Neurology, New York University Langone Health, New York, NY, United States
- VA New York Harbor Healthcare System, New York, NY, United States
| |
Collapse
|
20
|
Vaidya B, Padhy DS, Joshi HC, Sharma SS, Singh JN. Ion Channels and Metal Ions in Parkinson's Disease: Historical Perspective to the Current Scenario. Methods Mol Biol 2024; 2761:529-557. [PMID: 38427260 DOI: 10.1007/978-1-0716-3662-6_36] [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] [Indexed: 03/02/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative condition linked to the deterioration of motor and cognitive performance. It produces degeneration of the dopaminergic neurons along the nigrostriatal pathway in the central nervous system (CNS), which leads to symptoms such as bradykinesias, tremors, rigidity, and postural instability. There are several medications currently approved for the therapy of PD, but a permanent cure for it remains elusive. With the aging population set to increase, a number of PD cases are expected to shoot up in the coming times. Hence, there is a need to look for new molecular targets that could be investigated both preclinically and clinically for PD treatment. Among these, several ion channels and metal ions are being studied for their effects on PD pathology and the functioning of dopaminergic neurons. Ion channels such as N-methyl-D-aspartate (NMDA), γ-aminobutyric acid A (GABAA), voltage-gated calcium channels, potassium channels, HCN channels, Hv1 proton channels, and voltage-gated sodium channels and metal ions such as mercury, zinc, copper, iron, manganese, calcium, and lead showed prominent involvement in PD. Pharmacological agents have been used to target these ion channels and metal ions to prevent or treat PD. Hence, in the present review, we summarize the pathophysiological events linked to PD with an emphasis on the role of ions and ion channels in PD pathology, and pharmacological agents targeting these ion channels have also been listed.
Collapse
Affiliation(s)
- Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Dibya S Padhy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Hem C Joshi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India.
| | - Jitendra Narain Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India.
| |
Collapse
|
21
|
Pagowski V. A description of the bat star nervous system throughout larval ontogeny. Evol Dev 2024; 26:e12468. [PMID: 38108150 DOI: 10.1111/ede.12468] [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: 08/09/2023] [Revised: 11/13/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Larvae represent a distinct life history stage in which animal morphology and behavior contrast strongly to adult organisms. This life history stage is a ubiquitous aspect of animal life cycles, particularly in the marine environment. In many species, the structure and function of the nervous system differ significantly between metamorphosed juveniles and larvae. However, the distribution and diversity of neural cell types in larval nervous systems remains incompletely known. Here, the expression of neurotransmitter and neuropeptide synthesis and transport genes in the bat star Patiria miniata is examined throughout larval development. This characterization of nervous system structure reveals three main neural regions with distinct but overlapping territories. These regions include a densely innervated anterior region, an enteric neural plexus, and neurons associated with the ciliary band. In the ciliary band, cholinergic cells are pervasive while dopaminergic, noradrenergic, and GABAergic cells show regional differences in their localization patterns. Furthermore, the distribution of some neural subtypes changes throughout larval development, suggesting that changes in nervous system structure align with shifting ecological priorities during different larval stages, before the development of the adult nervous system. While past work has described aspects of P. miniata larval nervous system structure, largely focusing on early developmental timepoints, this work provides a comprehensive description of neural cell type localization throughout the extensive larval period.
Collapse
Affiliation(s)
- Veronica Pagowski
- Hopkins Marine Station of Stanford University, Pacific Grove, California, USA
| |
Collapse
|
22
|
Erickson JD, Kyllo T, Wulff H. Ca 2+-regulated expression of high affinity methylaminoisobutryic acid transport in hippocampal neurons inhibited by riluzole and novel neuroprotective aminothiazoles. Curr Res Physiol 2023; 6:100109. [PMID: 38107787 PMCID: PMC10724208 DOI: 10.1016/j.crphys.2023.100109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 12/19/2023] Open
Abstract
High affinity methylaminoisobutyric acid(MeAIB)/glutamine(Gln) transport activity regulated by neuronal firing occurs at the plasma membrane in mature rat hippocampal neuron-enriched cultures. Spontaneous Ca2+-regulated transport activity was similarly inhibited by riluzole, a benzothiazole anticonvulsant agent, and by novel naphthalenyl substituted aminothiazole derivatives such as SKA-378. Here, we report that spontaneous transport activity is stimulated by 4-aminopyridine (4-AP) and that phorbol-myristate acetate (PMA) increases high K+ stimulated transport activity that is inhibited by staurosporine. 4-AP-stimulated spontaneous and PMA-stimulated high K+-induced transport is not present at 7 days in vitro (DIV) and is maximal by DIV∼21. The relative affinity for MeAIB is similar for spontaneous and high K+-stimulated transport (Km ∼ 50 μM) suggesting that a single transporter is involved. While riluzole and SKA-378 inhibit spontaneous transport with equal potency (IC50 ∼ 1 μM), they exhibit decreased (∼3-5 X) potency for 4-AP-stimulated spontaneous transport. Interestingly, high K+-stimulated MeAIB transport displays lower and differential sensitivity to the two compounds. SKA-378-related halogenated derivatives of SKA-75 (SKA-219, SKA-377 and SKA-375) preferentially inhibit high K+-induced expression of MeAIB transport activity at the plasma membrane (IC50 < 25 μM), compared to SKA-75 and riluzole (IC50 > 100 μM). Ca2+-dependent spontaneous and high K+-stimulated MeAIB transport activity is blocked by ω-conotoxin MVIIC, ω-agatoxin IVA, ω-agatoxin TK (IC50 ∼ 500 nM) or cadmium ion (IC50 ∼ 20 μM) demonstrating that P/Q-type CaV channels that are required for activity-regulated presynaptic vesicular glutamate (Glu) release are also required for high-affinity MeAIB transport expression at the plasma membrane. We suggest that neural activity driven and Ca2+ dependent trafficking of the high affinity MeAIB transporter to the plasma membrane is a unique target to understand mechanisms of Glu/Gln recycling in synapses and acute neuroprotection against excitotoxic presynaptic Glu induced neural injury.
Collapse
Affiliation(s)
- Jeffrey D. Erickson
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, USA
| | - Thomas Kyllo
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, USA
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California-Davis, Davis, CA, USA
| |
Collapse
|
23
|
Rodríguez-Vega A, Dutra-Tavares AC, Souza TP, Semeão KA, Filgueiras CC, Ribeiro-Carvalho A, Manhães AC, Abreu-Villaça Y. Nicotine Exposure in a Phencyclidine-Induced Mice Model of Schizophrenia: Sex-Selective Medial Prefrontal Cortex Protein Markers of the Combined Insults in Adolescent Mice. Int J Mol Sci 2023; 24:14634. [PMID: 37834084 PMCID: PMC10572990 DOI: 10.3390/ijms241914634] [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: 07/19/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Tobacco misuse as a comorbidity of schizophrenia is frequently established during adolescence. However, comorbidity markers are still missing. Here, the method of label-free proteomics was used to identify deregulated proteins in the medial prefrontal cortex (prelimbic and infralimbic) of male and female mice modelled to schizophrenia with a history of nicotine exposure during adolescence. Phencyclidine (PCP), used to model schizophrenia (SCHZ), was combined with an established model of nicotine minipump infusions (NIC). The combined insults led to worse outcomes than each insult separately when considering the absolute number of deregulated proteins and that of exclusively deregulated ones. Partially shared Reactome pathways between sexes and between PCP, NIC and PCPNIC groups indicate functional overlaps. Distinctively, proteins differentially expressed exclusively in PCPNIC mice reveal unique effects associated with the comorbidity model. Interactome maps of these proteins identified sex-selective subnetworks, within which some proteins stood out: for females, peptidyl-prolyl cis-trans isomerase (Fkbp1a) and heat shock 70 kDa protein 1B (Hspa1b), both components of the oxidative stress subnetwork, and gamma-enolase (Eno2), a component of the energy metabolism subnetwork; and for males, amphiphysin (Amph), a component of the synaptic transmission subnetwork. These are proposed to be further investigated and validated as markers of the combined insult during adolescence.
Collapse
Affiliation(s)
- Andrés Rodríguez-Vega
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Ana Carolina Dutra-Tavares
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Thainá P. Souza
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Keila A. Semeão
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Claudio C. Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores da Universidade do Estado do Rio de Janeiro, São Gonçalo 24435-005, RJ, Brazil;
| | - Alex C. Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20550-170, RJ, Brazil; (A.R.-V.); (A.C.D.-T.); (T.P.S.); (K.A.S.); (C.C.F.); (A.C.M.)
| |
Collapse
|
24
|
Neves D, Salazar IL, Almeida RD, Silva RM. Molecular mechanisms of ischemia and glutamate excitotoxicity. Life Sci 2023; 328:121814. [PMID: 37236602 DOI: 10.1016/j.lfs.2023.121814] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
Excitotoxicity is classically defined as the neuronal damage caused by the excessive release of glutamate, and subsequent activation of excitatory plasma membrane receptors. In the mammalian brain, this phenomenon is mainly driven by excessive activation of glutamate receptors (GRs). Excitotoxicity is common to several chronic disorders of the Central Nervous System (CNS) and is considered the primary mechanism of neuronal loss of function and cell death in acute CNS diseases (e.g. ischemic stroke). Multiple mechanisms and pathways lead to excitotoxic cell damage including pro-death signaling cascade events downstream of glutamate receptors, calcium (Ca2+) overload, oxidative stress, mitochondrial impairment, excessive glutamate in the synaptic cleft as well as altered energy metabolism. Here, we review the current knowledge on the molecular mechanisms that underlie excitotoxicity, emphasizing the role of Nicotinamide Adenine Dinucleotide (NAD) metabolism. We also discuss novel and promising therapeutic strategies to treat excitotoxicity, highlighting recent clinical trials. Finally, we will shed light on the ongoing search for stroke biomarkers, an exciting and promising field of research, which may improve stroke diagnosis, prognosis and allow better treatment options.
Collapse
Affiliation(s)
- Diogo Neves
- iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Ivan L Salazar
- Multidisciplinary Institute of Ageing, MIA - Portugal, University of Coimbra, Coimbra, Portugal
| | - Ramiro D Almeida
- iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | - Raquel M Silva
- iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; Universidade Católica Portuguesa, Faculdade de Medicina Dentária, Centro de Investigação Interdisciplinar em Saúde, Viseu, Portugal.
| |
Collapse
|
25
|
Tarazi D, Maynes JT. Impact of Opioids on Cellular Metabolism: Implications for Metabolic Pathways Involved in Cancer. Pharmaceutics 2023; 15:2225. [PMID: 37765194 PMCID: PMC10534826 DOI: 10.3390/pharmaceutics15092225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Opioid utilization for pain management is prevalent among cancer patients. There is significant evidence describing the many effects of opioids on cancer development. Despite the pivotal role of metabolic reprogramming in facilitating cancer growth and metastasis, the specific impact of opioids on crucial oncogenic metabolic pathways remains inadequately investigated. This review provides an understanding of the current research on opioid-mediated changes to cellular metabolic pathways crucial for oncogenesis, including glycolysis, the tricarboxylic acid cycle, glutaminolysis, and oxidative phosphorylation (OXPHOS). The existing literature suggests that opioids affect energy production pathways via increasing intracellular glucose levels, increasing the production of lactic acid, and reducing ATP levels through impediment of OXPHOS. Opioids modulate pathways involved in redox balance which may allow cancer cells to overcome ROS-mediated apoptotic signaling. The majority of studies have been conducted in healthy tissue with a predominant focus on neuronal cells. To comprehensively understand the impact of opioids on metabolic pathways critical to cancer progression, research must extend beyond healthy tissue and encompass patient-derived cancer tissue, allowing for a better understanding in the context of the metabolic reprogramming already undergone by cancer cells. The current literature is limited by a lack of direct experimentation exploring opioid-induced changes to cancer metabolism as they relate to tumor growth and patient outcome.
Collapse
Affiliation(s)
- Doorsa Tarazi
- Department of Biochemistry, University of Toronto, Toronto, ON M5G 1A8, Canada;
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Jason T. Maynes
- Department of Biochemistry, University of Toronto, Toronto, ON M5G 1A8, Canada;
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON M5G 1E2, Canada
| |
Collapse
|
26
|
Fang M, Liu W, Tuo J, Liu M, Li F, Zhang L, Yu C, Xu Z. Advances in understanding the pathogenesis of post-traumatic epilepsy: a literature review. Front Neurol 2023; 14:1141434. [PMID: 37638179 PMCID: PMC10449544 DOI: 10.3389/fneur.2023.1141434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Severe head trauma can lead to seizures. Persistent epileptic seizures and their progression are associated with the severity of trauma. Although case reports have revealed that early use of anti-seizure drugs after trauma can prevent epilepsy, clinical case-control studies have failed to confirm this phenomenon. To date, many brain trauma models have been used to study the correlation between post-traumatic seizures and related changes in neural circuit function. According to these studies, neuronal and glial responses are activated immediately after brain trauma, usually leading to significant cell loss in injured brain regions. Over time, long-term changes in neural circuit tissues, especially in the neocortex and hippocampus, lead to an imbalance between excitatory and inhibitory neurotransmission and an increased risk of spontaneous seizures. These changes include alterations in inhibitory interneurons and the formation of new, over-recurrent excitatory synaptic connections. In this study, we review the progress of research related to post-traumatic epilepsy to better understand the mechanisms underlying the initiation and development of post-traumatic seizures and to provide theoretical references for the clinical treatment of post-traumatic seizures.
Collapse
Affiliation(s)
- Mingzhu Fang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Sichuan Provincial People’s Hospital Medical Group Chuantou Xichang Hospital, Xichang, China
| | - Wanyu Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jinmei Tuo
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Fangjing Li
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lijia Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
27
|
Tzanetakos D, Kyrozis A, Karavasilis E, Velonakis G, Tzartos JS, Toulas P, Sotirli SA, Evdokimidis I, Tsivgoulis G, Potagas C, Kilidireas C, Andreadou E. Early metabolic alterations in the normal‑appearing grey and white matter of patients with clinically isolated syndrome suggestive of multiple sclerosis: A proton MR spectroscopic study. Exp Ther Med 2023; 26:349. [PMID: 37324507 PMCID: PMC10265702 DOI: 10.3892/etm.2023.12048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/18/2023] [Indexed: 06/17/2023] Open
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) is an advanced method of examining metabolic profiles. The present study aimed to assess in vivo metabolite levels in areas of normal-appearing grey (thalamus) and white matter (centrum semiovale) using 1H-MRS in patients with clinically isolated syndrome (CIS) suggestive of multiple sclerosis and compare them to healthy controls (HCs). Data from 35 patients with CIS (CIS group), of which 23 were untreated (CIS-untreated group) and 12 were treated (CIS-treated group) with disease-modifying-therapies (DMTs) at the time of 1H-MRS, and from 28 age- and sex-matched HCs were collected using a 3.0 T MRI and single-voxel 1H-MRS (point resolved spectroscopy sequence; repetition time, 2,000 msec; time to echo, 35 msec). Concentrations and ratios of total N-acetyl aspartate (tNAA), total creatine (tCr), total choline (tCho), myoinositol, glutamate (Glu), glutamine (Gln), Glu + Gln (Glx) and glutathione (Glth) were estimated in the thalamic-voxel (th) and centrum semiovale-voxel (cs). For the CIS group, the median duration from the first clinical attack to 1H-MRS was 102 days (interquartile range, 89.5.-131.5). Compared with HCs, significantly lower Glx(cs) (P=0.014) and ratios of tCho/tCr(th) (P=0.026), Glu/tCr(cs) (P=0.040), Glx/tCr(cs) (P=0.004), Glx/tNAA(th) (P=0.043) and Glx/tNAA(cs) (P=0.015) were observed in the CIS group. No differences in tNAA levels were observed between the CIS and the HC groups; however, tNAA(cs) was higher in the CIS-treated than in the CIS-untreated group (P=0.028). Compared with those in HC group, decreased Glu(cs) (P=0.019) and Glx(cs) levels (P=0.014) and lower ratios for tCho/tCr(th) (P=0.015), Gln/tCr(th) (P=0.004), Glu/tCr(cs) (P=0.021), Glx/tCr(th) (P=0.041), Glx/tCr(cs) (P=0.003), Glx/tNAA(th) (P=0.030) and Glx/tNAA(cs) (P=0.015) were found in the CIS-untreated group. The present findings showed alterations in the normal-appearing grey and white matter of patients with CIS; moreover, the present results suggested an early indirect treatment effect of DMTs on the brain metabolic profile of these patients.
Collapse
Affiliation(s)
- Dimitrios Tzanetakos
- Second Department of Neurology, ‘Attikon’ University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Andreas Kyrozis
- First Department of Neurology, Eginition Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Efstratios Karavasilis
- Research Unit of Radiology, Second Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
- Medical Physics Laboratory, School of Medicine, Democritus University of Thrace, 68100 Alexandroupoli, Greece
| | - Georgios Velonakis
- Research Unit of Radiology, Second Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - John S. Tzartos
- Second Department of Neurology, ‘Attikon’ University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Panagiotis Toulas
- Research Unit of Radiology, Second Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Stefania Alexia Sotirli
- MS Center and Other Neurodegenerative diseases, Metropolitan General Hospital, 15562 Holargos, Athens, Greece
| | - Ioannis Evdokimidis
- First Department of Neurology, Eginition Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, ‘Attikon’ University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Constantin Potagas
- First Department of Neurology, Eginition Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Costantinos Kilidireas
- First Department of Neurology, Eginition Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Elisabeth Andreadou
- First Department of Neurology, Eginition Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
28
|
Cui Q, Jiang D, Zhang Y, Chen C. The tumor-nerve circuit in breast cancer. Cancer Metastasis Rev 2023; 42:543-574. [PMID: 36997828 PMCID: PMC10349033 DOI: 10.1007/s10555-023-10095-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/16/2023] [Indexed: 04/01/2023]
Abstract
It is well established that innervation is one of the updated hallmarks of cancer and that psychological stress promotes the initiation and progression of cancer. The breast tumor environment includes not only fibroblasts, adipocytes, endothelial cells, and lymphocytes but also neurons, which is increasingly discovered important in breast cancer progression. Peripheral nerves, especially sympathetic, parasympathetic, and sensory nerves, have been reported to play important but different roles in breast cancer. However, their roles in the breast cancer progression and treatment are still controversial. In addition, the brain is one of the favorite sites of breast cancer metastasis. In this review, we first summarize the innervation of breast cancer and its mechanism in regulating cancer growth and metastasis. Next, we summarize the neural-related molecular markers in breast cancer diagnosis and treatment. In addition, we review drugs and emerging technologies used to block the interactions between nerves and breast cancer. Finally, we discuss future research directions in this field. In conclusion, the further research in breast cancer and its interactions with innervated neurons or neurotransmitters is promising in the clinical management of breast cancer.
Collapse
Affiliation(s)
- Qiuxia Cui
- Affiliated Hospital of Guangdong Medical University Science & Technology of China, Zhanjiang, 524000, China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Dewei Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Yuanqi Zhang
- Affiliated Hospital of Guangdong Medical University Science & Technology of China, Zhanjiang, 524000, China.
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China.
- The Third Affiliated Hospital, Kunming Medical University, Kunming, 650118, China.
| |
Collapse
|
29
|
Ham S, Kim HJ, Shin N, Hwang JH, Oh SJ, Park JY, Joo JC, Kim HT, Bhatia SK, Yang YH. Continuous production of gamma aminobutyric acid by engineered and immobilized Escherichia coli whole-cells in a small-scale reactor system. Enzyme Microb Technol 2023; 168:110258. [PMID: 37210798 DOI: 10.1016/j.enzmictec.2023.110258] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
γ-Amino butyric acid (GABA) is a non-proteinogenic amino acid and a human neurotransmitter. Recently, increasing demand for food additives and biodegradable bioplastic monomers, such as nylon 4, has been reported. Consequently, considerable efforts have been made to produce GABA through fermentation and bioconversion. To realize bioconversion, wild-type or recombinant strains harboring glutamate decarboxylase were paired with the cheap starting material monosodium glutamate, resulting in less by-product formation and faster production compared to fermentation. To increase the reusability and stability of whole-cell production systems, this study used an immobilization and continuous production system with a small-scale continuous reactor for gram-scale production. The cation type, alginate concentration, barium concentration, and whole-cell concentration in the beads were optimized and this optimization resulted in more than 95 % conversion of 600 mM monosodium glutamate to GABA in 3 h and reuse of the immobilized cells 15 times, whereas free cells lost all activity after the ninth reaction. When a continuous production system was applied after optimizing the buffer concentration, substrate concentration, and flow rate, 165 g of GABA was produced after 96 h of continuous operation in a 14-mL scale reactor. Our work demonstrates the efficient and economical production of GABA by immobilization and continuous production in a small-scale reactor.
Collapse
Affiliation(s)
- Sion Ham
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Hyun Jin Kim
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Nara Shin
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Jeong Hyeon Hwang
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Suk Jin Oh
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Jun Young Park
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Jeong Chan Joo
- Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Republic of Korea
| | - Hee Taek Kim
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Chungnam National University, Chungchung nam-do, Republic of Korea
| | - Shashi Kant Bhatia
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea.
| | - Yung-Hun Yang
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Elsaid S, Truong P, Sailasuta N, Le Foll B. Evaluating Back-to-Back and Day-to-Day Reproducibility of Cortical GABA+ Measurements Using Proton Magnetic Resonance Spectroscopy ( 1H MRS). Int J Mol Sci 2023; 24:ijms24097713. [PMID: 37175420 PMCID: PMC10178500 DOI: 10.3390/ijms24097713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
γ-aminobutyric acid (GABA) is a major inhibitory neurotransmitter implicated in neuropsychiatric disorders. The best method for quantifying GABA is proton magnetic resonance spectroscopy (1H MRS). Considering that accurate measurements of GABA are affected by slight methodological alterations, demonstrating GABA reproducibility in healthy volunteers is essential before implementing the changes in vivo. Thus, our study aimed to evaluate the back-to-back (B2B) and day-to-day (D2D) reproducibility of GABA+ macromolecules (GABA+) using a 3 Tesla MRI scanner, the new 32-channel head coil (CHC), and Mescher-Garwood Point Resolved Spectroscopy (MEGA-PRESS) technique with the scan time (approximately 10 min), adequate for psychiatric patients. The dorsomedial pre-frontal cortex/anterior cingulate cortex (dmPFC/ACC) was scanned in 29 and the dorsolateral pre-frontal cortex (dlPFC) in 28 healthy volunteers on two separate days. Gannet 3.1 was used to quantify GABA+. The reproducibility was evaluated by Pearson's r correlation, the interclass-correlation coefficient (ICC), and the coefficient of variation (CV%) (r/ICC/CV%). For Day 1, B2B reproducibility was 0.59/0.60/5.02% in the dmPFC/ACC and 0.74/0.73/5.15% for dlPFC. For Day 2, it was 0.60/0.59/6.26% for the dmPFC/ACC and 0.54/0.54/6.89 for dlPFC. D2D reproducibility of averaged GABA+ was 0.62/0.61/4.95% for the dmPFC/ACC and 0.58/0.58/5.85% for dlPFC. Our study found excellent GABA+ repeatability and reliability in the dmPFC/ACC and dlPFC.
Collapse
Affiliation(s)
- Sonja Elsaid
- Translation Addiction Research Laboratory (TARL), Centre for Addiction and Mental Health (CAMH), Toronto, ON M5S 2S1, Canada
- Institute of Medical Science (IMS), Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Brain Health Imaging Centre (BHIC), Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
| | - Peter Truong
- Brain Health Imaging Centre (BHIC), Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Sunnybrook Health Science Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Napapon Sailasuta
- Brain Health Imaging Centre (BHIC), Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Departments of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Bernard Le Foll
- Translation Addiction Research Laboratory (TARL), Centre for Addiction and Mental Health (CAMH), Toronto, ON M5S 2S1, Canada
- Institute of Medical Science (IMS), Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Brain Health Imaging Centre (BHIC), Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Addictions Division, Centre for Addiction and Mental Health (CAMH), Toronto, ON M6J 1H4, Canada
- Waypoint Centre for Mental Health Care, Waypoint Research Institute, Penetanguishene, ON L9M 1G3, Canada
| |
Collapse
|
31
|
Tang Y, Yan Y, Mao J, Ni J, Qing H. The hippocampus associated GABAergic neural network impairment in early-stage of Alzheimer's disease. Ageing Res Rev 2023; 86:101865. [PMID: 36716975 DOI: 10.1016/j.arr.2023.101865] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is the commonest neurodegenerative disease with slow progression. Pieces of evidence suggest that the GABAergic system is impaired in the early stage of AD, leading to hippocampal neuron over-activity and further leading to memory and cognitive impairment in patients with AD. However, the precise impairment mechanism of the GABAergic system on the pathogenesis of AD is still unclear. The impairment of neural networks associated with the GABAergic system is tightly associated with AD. Therefore, we describe the roles played by hippocampus-related GABAergic circuits and their impairments in AD neuropathology. In addition, we give our understand on the process from GABAergic circuit impairment to cognitive and memory impairment, since recent studies on astrocyte in AD plays an important role behind cognition dysfunction caused by GABAergic circuit impairment, which helps better understand the GABAergic system and could open up innovative AD therapy.
Collapse
Affiliation(s)
- Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jian Mao
- Zhengzhou Tobacco Institute of China National Tobacco Company, Zhengzhou 450001, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China.
| |
Collapse
|
32
|
Meguid NA, Hashem HS, Ghanem MH, Helal SA, Semenova Y, Hashem S, Hashish A, Chirumbolo S, Elwan AM, Bjørklund G. Evaluation of Branched-Chain Amino Acids in Children with Autism Spectrum Disorder and Epilepsy. Mol Neurobiol 2023; 60:1997-2004. [PMID: 36600079 DOI: 10.1007/s12035-022-03202-w] [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: 10/10/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023]
Abstract
Autism spectrum disorder (ASD) and epilepsy run hand-to-hand in their pathophysiology. Epilepsy is not an uncommon finding in patients with ASD. The aim of the present study was to identify the metabolic abnormalities of BCAAs (leucine, isoleucine, and valine) in children with ASD with and without seizures in comparison with neurotypical controls. Also, this study aimed to investigate the presence of epileptiform discharges on electroencephalography (EEG) in ASD patients and to describe the types and frequency of seizures observed. The study included 90 children aged 2-7 years, 30 of whom were diagnosed with both ASD and epilepsy. The other 30 children were diagnosed as ASD without epilepsy, and a comparable 30 normally developed children served as a control group. The groups were matched by age and gender. All patients were referred to the Autism Disorders Clinic for interviews and examinations. The Childhood Autism Rating Scale (CARS) was applied to all study participants to assess the degree of autism. The present study results show that all types of seizures may be identified in ASD children. The median serum levels of BCAAs were lower in ASD children with and without epilepsy than in neurotypical controls. This opens the door for discussion about new etiologies and better categorizations of ASD based on genotype and genetic abnormalities detected. More studies with larger samples are needed to understand ASD better and to more reliable evaluate the association between ASD, EEG changes, seizures, and BCAAs.
Collapse
Affiliation(s)
- Nagwa A Meguid
- Research On Children With Special Needs Department, National Research Centre, Giza, Egypt.,CONEM Egypt Child Brain Research Group, National Research Centre, Giza, Egypt
| | - Heba S Hashem
- Research On Children With Special Needs Department, National Research Centre, Giza, Egypt
| | - Mohamed H Ghanem
- Faculty of Medicine, Department of Neurology and Psychiatry, Ain Shams University, El-Abaseya, Egypt
| | - Samia A Helal
- Faculty of Medicine, Department of Neurology and Psychiatry, Ain Shams University, El-Abaseya, Egypt
| | - Yuliya Semenova
- Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Saher Hashem
- Department of Neurology, Cairo University, Cairo, Egypt
| | - Adel Hashish
- Research On Children With Special Needs Department, National Research Centre, Giza, Egypt
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,CONEM Scientific Secretary, Verona, Italy
| | - Ahmed M Elwan
- Research On Children With Special Needs Department, National Research Centre, Giza, Egypt
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo I Rana, Norway.
| |
Collapse
|
33
|
Zhang SN, Li HM, Liu Q, Li XZ, Yang WD, Zhou Y. Omics combined with network pharmacology reveal the neuroprotective mechanism of Sophora tonkinensis based on the biolabel research pattern: The treatment of Parkinson's disease against oxidative stress and neuroexcitatory toxicity. Biomed Chromatogr 2023; 37:e5557. [PMID: 36453605 DOI: 10.1002/bmc.5557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/19/2022] [Accepted: 11/26/2022] [Indexed: 12/04/2022]
Abstract
Based on the biolabel research pattern, omics and network pharmacology were used for exploring the neuroprotection of Sophora tonkinensis (ST) in the treatment of brain diseases. Multi-omics were applied to investigate biolabels for ST intervention in brain tissue. Based on biolabels, the therapeutic potential, mechanism and material basis of ST for treating brain diseases were topologically analyzed by network pharmacology. A Parkinson's disease (PD) mouse model was used to validate biolabel analysis results. Four proteins and three metabolites were involved in two key pathways (alanine, aspartate and glutamate metabolism and arginine biosynthesis) and considered as biolabels. Network pharmacology showed that ST has the potential to treat some brain diseases, especially PD. Eight compounds (including caffeic acid, gallic acid and cinnamic acid) may serve as the material basis of ST treating brain diseases via the mediation of three biolabels. In the PD model, ST and its active compounds (caffeic acid and gallic acid) may protect dopaminergic neurons (maximum recovery rate for dopamine, 49.5%) from oxidative stress (E3 ubiquitin-protein ligase parkin, reactive oxygen species, nitric oxide, etc.) and neuroexcitatory toxicity (glutamate dehydrogenase, glutamine, glutamic acid, etc.). These findings indicated that omics and network pharmacology may contribute to the achievement of the objectives of this study based on the biolabel research pattern.
Collapse
Affiliation(s)
- Shuai-Nan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, China
| | - Hong-Mei Li
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, China
| | - Qi Liu
- The Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Xu-Zhao Li
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, China
| | - Wu-de Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, China
| | - Ying Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, China
| |
Collapse
|
34
|
Henao‐Restrepo J, López‐Murillo C, Valderrama‐Carmona P, Orozco‐Santa N, Gomez J, Gutiérrez‐Vargas J, Moraga R, Toledo J, Littau JL, Härtel S, Arboleda‐Velásquez JF, Sepulveda‐Falla D, Lopera F, Cardona‐Gómez GP, Villegas A, Posada‐Duque R. Gliovascular alterations in sporadic and familial Alzheimer's disease: APOE3 Christchurch homozygote glioprotection. Brain Pathol 2023; 33:e13119. [PMID: 36130084 PMCID: PMC10041169 DOI: 10.1111/bpa.13119] [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: 01/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
In response to brain insults, astrocytes become reactive, promoting protection and tissue repair. However, astroglial reactivity is typical of brain pathologies, including Alzheimer's disease (AD). Considering the heterogeneity of the reactive response, the role of astrocytes in the course of different forms of AD has been underestimated. Colombia has the largest human group known to have familial AD (FAD). This group carries the autosomal dominant and fully penetrant mutation E280A in PSEN1, which causes early-onset AD. Recently, our group identified an E280A carrier who did not develop FAD. The individual was homozygous for the Christchurch mutation R136S in APOE3 (APOEch). Remarkably, APOE is the main genetic risk factor for developing sporadic AD (SAD) and most of cerebral ApoE is produced by astroglia. Here, we characterized astrocyte properties related to reactivity, glutamate homeostasis, and structural integrity of the gliovascular unit (GVU), as factors that could underlie the pathogenesis or protection of AD. Specifically, through histological and 3D microscopy analyses of postmortem samples, we briefly describe the histopathology and cytoarchitecture of the frontal cortex of SAD, FAD, and APOEch, and demonstrate that, while astrodegeneration and vascular deterioration are prominent in SAD, FAD is characterized by hyperreactive-like glia, and APOEch displays the mildest astrocytic and vascular alterations despite having the highest burden of Aβ. Notably, astroglial, gliovascular, and vascular disturbances, as well as brain cell death, correlate with the specific astrocytic phenotypes identified in each condition. This study provides new insights into the potential relevance of the gliovasculature in the development and protection of AD. To our knowledge, this is the first study assessing the components of the GVU in human samples of SAD, FAD, and APOEch.
Collapse
Affiliation(s)
- Julián Henao‐Restrepo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Carolina López‐Murillo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Pablo Valderrama‐Carmona
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Natalia Orozco‐Santa
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Johana Gomez
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Johanna Gutiérrez‐Vargas
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Health Sciences FacultyRemington University CorporationMedellínColombia
| | - Renato Moraga
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jorge Toledo
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jessica Lisa Littau
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Steffen Härtel
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Joseph F. Arboleda‐Velásquez
- Schepens Eye Research Institute of Mass Eye and Ear, Department of OphthalmologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Diego Sepulveda‐Falla
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Gloria Patricia Cardona‐Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Andrés Villegas
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Rafael Posada‐Duque
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| |
Collapse
|
35
|
Chakraborty P, Dey A, Gopalakrishnan AV, Swati K, Ojha S, Prakash A, Kumar D, Ambasta RK, Jha NK, Jha SK, Dewanjee S. Glutamatergic neurotransmission: A potential pharmacotherapeutic target for the treatment of cognitive disorders. Ageing Res Rev 2023; 85:101838. [PMID: 36610558 DOI: 10.1016/j.arr.2022.101838] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
In the mammalian brain, glutamate is regarded to be the primary excitatory neurotransmitter due to its widespread distribution and wide range of metabolic functions. Glutamate plays key roles in regulating neurogenesis, synaptogenesis, neurite outgrowth, and neuron survival in the brain. Ionotropic and metabotropic glutamate receptors, neurotransmitters, neurotensin, neurosteroids, and others co-ordinately formulate a complex glutamatergic network in the brain that maintains optimal excitatory neurotransmission. Cognitive activities are potentially synchronized by the glutamatergic activities in the brain via restoring synaptic plasticity. Dysfunctional glutamate receptors and other glutamatergic components are responsible for the aberrant glutamatergic activity in the brain that cause cognitive impairments, loss of synaptic plasticity, and neuronal damage. Thus, controlling the brain's glutamatergic transmission and modifying glutamate receptor function could be a potential therapeutic strategy for cognitive disorders. Certain drugs that regulate glutamate receptor activities have shown therapeutic promise in improving cognitive functions in preclinical and clinical studies. However, several issues regarding precise functional information of glutamatergic activity are yet to be comprehensively understood. The present article discusses the scope of developing glutamatergic systems as prospective pharmacotherapeutic targets to treat cognitive disorders. Special attention has been given to recent developments, challenges, and future prospects.
Collapse
Affiliation(s)
- Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Anand Prakash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand 248007, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India.
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
36
|
Kim K, Yoon H. Gamma-Aminobutyric Acid Signaling in Damage Response, Metabolism, and Disease. Int J Mol Sci 2023; 24:ijms24054584. [PMID: 36902014 PMCID: PMC10003236 DOI: 10.3390/ijms24054584] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Gamma-aminobutyric acid (GABA) plays a crucial role in signal transduction and can function as a neurotransmitter. Although many studies have been conducted on GABA in brain biology, the cellular function and physiological relevance of GABA in other metabolic organs remain unclear. Here, we will discuss recent advances in understanding GABA metabolism with a focus on its biosynthesis and cellular functions in other organs. The mechanisms of GABA in liver biology and disease have revealed new ways to link the biosynthesis of GABA to its cellular function. By reviewing what is known about the distinct effects of GABA and GABA-mediated metabolites in physiological pathways, we provide a framework for understanding newly identified targets regulating the damage response, with implications for ameliorating metabolic diseases. With this review, we suggest that further research is necessary to develop GABA's beneficial and toxic effects on metabolic disease progression.
Collapse
|
37
|
The Long Noncoding RNA Cytoskeleton Regulator RNA (CYTOR)/miRNA-24-3p Axis Facilitates Nasopharyngeal Carcinoma Progression by Modulating GAD1 Expression. JOURNAL OF ONCOLOGY 2023; 2023:6027860. [PMID: 36814556 PMCID: PMC9940962 DOI: 10.1155/2023/6027860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/11/2022] [Accepted: 11/24/2022] [Indexed: 02/16/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is a head and neck epithelial carcinoma that is unusually prevalent in Southeast Asia. Noncoding RNAs, including lncRNA and miRNA, and their target genes are considered vital regulators of tumorigenesis and the progression of NPC. However, the detailed underlying mechanisms of GAD1 involved in the regulation of NPC need to be further elucidated. In the present study, we identified that GAD1 was significantly upregulated in NPC tissues. GAD1 overexpression is promoted, while genetic knockdown of GAD1 suppresses proliferation, colony formation, migration, and invasion of NPC cells. Bioinformatics analysis and a luciferase reporter assay demonstrated that GAD1 is a direct target gene of miR-24-3p. In NPC tissues, miR-24-3p was downregulated and the lncRNA CYTOR was upregulated. CYTOR was sponged to suppress the function of miR-24-3p. CYTOR regulates GAD1 expression via modulating miR-24-3p. The CYTOR/miR-24-3p/GAD1 axis is converged to modulate the growth, migration, and invasion of NPC cells. In conclusion, the study identified a novel axis for the regulation of NPC cell growth, providing new insights into the understanding of NPC.
Collapse
|
38
|
Ncho CM, Goel A, Gupta V, Jeong CM, Choi YH. Effect of in ovo feeding of γ-aminobutyric acid combined with embryonic thermal manipulation on hatchability, growth, and hepatic gene expression in broilers. Anim Biosci 2023; 36:284-294. [PMID: 35798039 PMCID: PMC9834729 DOI: 10.5713/ab.22.0099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE This study investigated the effects of in ovo feeding of γ-aminobutyric acid (GABA) and embryonic thermal manipulation (ETM) on growth performance, organ indices, plasma biochemical parameters, hepatic antioxidant levels, and expression of lipid metabolism-related genes in broilers. METHODS Two hundred and fifty eggs were assigned to one of four treatments: control eggs incubated under standard conditions (CON); eggs that received an in ovo injection of 10% GABA on day 17.5 of incubation (G10); thermally manipulated eggs between days 10 and 18 of incubation at 39.6°C for 6 h daily (TM); and eggs that received both treatments during incubation (G10+TM). After 28 days of rearing, five birds per treatment were selected for blood and organ sampling. RESULTS No differences were found in hatchability or growth parameters among different treatment groups. Hepatic gene expression of catalase (CAT) and glutathione peroxidase 1 (GPx1) was upregulated (p = 0.046 and p = 0.006, respectively) in the G10+TM group, while that of nuclear factor erythroid 2-related factor 2 (NRF2) was upregulated (p = 0.039) in the G10 group. In addition, the relative gene expression of NADPH oxidase 1 (NOX1) was significantly lower (p = 0.007) in all treatment groups than that in the CON group. Hepatic fatty acid synthase (FAS) levels and average daily feed intake (ADFI) of last week showed a positive correlation (r = 0.50, p = 0.038). In contrast, the relative gene expression of the extracellular fatty acid-binding protein (EXFAB) and peroxisome proliferator-activated receptor-γ (PPAR-γ) were positively correlated (r = 0.48, p = 0.042 and r = 0.50, p = 0.031) with the overall ADFI of birds. CONCLUSION Taken together, the results of this study suggest that the combination of in ovo feeding of GABA and ETM can enhance hepatic antioxidant function in broilers.
Collapse
Affiliation(s)
- Chris Major Ncho
- Department of Animal Science, Gyeongsang National University, Jinju 52828,
Korea,Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828,
Korea
| | - Akshat Goel
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828,
Korea
| | - Vaishali Gupta
- Division of Applied Life Sciences (BK21 Plus Program), Gyeongsang National University, Jinju 52828,
Korea
| | - Chae-Mi Jeong
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828,
Korea,Division of Applied Life Sciences (BK21 Plus Program), Gyeongsang National University, Jinju 52828,
Korea
| | - Yang-Ho Choi
- Department of Animal Science, Gyeongsang National University, Jinju 52828,
Korea,Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828,
Korea,Division of Applied Life Sciences (BK21 Plus Program), Gyeongsang National University, Jinju 52828,
Korea,Corresponding Author: Yang-Ho Choi, Tel: +82-55-772-1946, Fax: +82-55-772-1949, E-mail:
| |
Collapse
|
39
|
Demchenko IT, Suliman HB, Zhilyaey SY, Alekseeva OS, Platonova TF, Makowski MS, Piantadosi CA, Gasier HG. GAT inhibition preserves cerebral blood flow and reduces oxidant damage to mitochondria in rodents exposed to extreme hyperbaric oxygen. Front Mol Neurosci 2023; 15:1062410. [PMID: 36704328 PMCID: PMC9871636 DOI: 10.3389/fnmol.2022.1062410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Oxygen breathing at elevated partial pressures (PO2's) at or more than 3 atmospheres absolute (ATA) causes a reduction in brain γ-aminobutyric acid (GABA) levels that impacts the development of central nervous system oxygen toxicity (CNS-OT). Drugs that increase brain GABA content delay the onset of CNS-OT, but it is unknown if oxidant damage is lessened because brain tissue PO2 remains elevated during hyperbaric oxygen (HBO2) exposures. Experiments were performed in rats and mice to measure brain GABA levels with or without GABA transporter inhibitors (GATs) and its influence on cerebral blood flow, oxidant damage, and aspects of mitochondrial quality control signaling (mitophagy and biogenesis). In rats pretreated with tiagabine (GAT1 inhibitor), the tachycardia, secondary rise in mean arterial blood pressure, and cerebral hyperemia were prevented during HBO2 at 5 and 6 ATA. Tiagabine and the nonselective GAT inhibitor nipecotic acid similarly extended HBO2 seizure latencies. In mice pretreated with tiagabine and exposed to HBO2 at 5 ATA, nuclear and mitochondrial DNA oxidation and astrocytosis was attenuated in the cerebellum and hippocampus. Less oxidant injury in these regions was accompanied by reduced conjugated microtubule-associated protein 1A/1B-light chain 3 (LC3-II), an index of mitophagy, and phosphorylated cAMP response element binding protein (pCREB), an initiator of mitochondrial biogenesis. We conclude that GABA prevents cerebral hyperemia and delays neuroexcitation under extreme HBO2, limiting oxidant damage in the cerebellum and hippocampus, and likely lowering mitophagy flux and initiation of pCREB-initiated mitochondrial biogenesis.
Collapse
Affiliation(s)
- Ivan T. Demchenko
- The Duke Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, NC, United States
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Hagir B. Suliman
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Sergey Y. Zhilyaey
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga S. Alekseeva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Tatyana F. Platonova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Matthew S. Makowski
- The Duke Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, NC, United States
| | - Claude A. Piantadosi
- The Duke Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, NC, United States
| | - Heath G. Gasier
- The Duke Center for Hyperbaric Medicine and Environmental Physiology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
40
|
Fathi M, Saeedyan S, Kaoosi M. Gamma-amino butyric acid (GABA) supplementation alleviates dexamethasone treatment-induced oxidative stress and inflammation response in broiler chickens. Stress 2023; 26:2185861. [PMID: 36861448 DOI: 10.1080/10253890.2023.2185861] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
This experiment was conducted to investigate the effect of Gamma-amino butyric acid (GABA) on growth performance, serum and liver antioxidant status, inflammation response and hematological changes, in male broiler chickens under experimentally induced stress via in-feed dexamethasone (DEX). A total of 300 male chicks (Ross 308) on day 7 after hatching, were randomly selected into four groups which were positive control group (PC, without any treatment), negative control (NC, with 1 mg/kg DEX), a third group received 1 mg/kg DEX and 100 mg/kg GABA (DG +) and the last one was (DG ++) which received 1 mg/kg DEX and 200 mg/kg GABA. Each group has five replicates (15 birds/replicate). Dietary GABA modulated DEX-induced adverse effects on body weight, feed intake, and feed conversion ratio. The DEX-induced effect of serum levels of IL-6 and IL-10 was reduced by dietary GABA supplementation. The activity of serum and liver superoxide dismutase, catalase, glutathione peroxidase were enhanced and malondialdehyde was reduced by GABA supplementation. The serum levels of total cholesterol & triglyceride were higher while low-density lipoprotein & high-density lipoprotein were lower in GABA groups than NC group. GABA supplementation also significantly decreased the heterophil, heterophil/lymphocyte ratio and elevated the activities of aspartate aminotransferase (AST), alanine transaminase (ALT) and alkaline phosphatase (ALP) than NC group. In conclusion, dietary GABA supplementation can alleviate DEX stress-induced oxidative stress and inflammation response.
Collapse
Affiliation(s)
- Mokhtar Fathi
- Department of Animal Science, Payam Noor University, Tehran, Iran
| | | | - Majid Kaoosi
- Department of Biology, Payam Noor University, Tehran, Iran
| |
Collapse
|
41
|
Zalachoras I, Ramos-Fernández E, Hollis F, Trovo L, Rodrigues J, Strasser A, Zanoletti O, Steiner P, Preitner N, Xin L, Astori S, Sandi C. Glutathione in the nucleus accumbens regulates motivation to exert reward-incentivized effort. eLife 2022; 11:77791. [DOI: 10.7554/elife.77791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
Emerging evidence is implicating mitochondrial function and metabolism in the nucleus accumbens in motivated performance. However, the brain is vulnerable to excessive oxidative insults resulting from neurometabolic processes, and whether antioxidant levels in the nucleus accumbens contribute to motivated performance is not known. Here, we identify a critical role for glutathione (GSH), the most important endogenous antioxidant in the brain, in motivation. Using proton magnetic resonance spectroscopy at ultra-high field in both male humans and rodent populations, we establish that higher accumbal GSH levels are highly predictive of better, and particularly, steady performance over time in effort-related tasks. Causality was established in in vivo experiments in rats that, first, showed that downregulating GSH levels through micro-injections of the GSH synthesis inhibitor buthionine sulfoximine in the nucleus accumbens impaired effort-based reward-incentivized performance. In addition, systemic treatment with the GSH precursor N-acetyl-cysteine increased accumbal GSH levels in rats and led to improved performance, potentially mediated by a cell-type-specific shift in glutamatergic inputs to accumbal medium spiny neurons. Our data indicate a close association between accumbal GSH levels and an individual’s capacity to exert reward-incentivized effort over time. They also suggest that improvement of accumbal antioxidant function may be a feasible approach to boost motivation.
Collapse
Affiliation(s)
- Ioannis Zalachoras
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Eva Ramos-Fernández
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Fiona Hollis
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine
| | - Laura Trovo
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé SA, Vers-chez-les-Blanc
| | - João Rodrigues
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Alina Strasser
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Pascal Steiner
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé SA, Vers-chez-les-Blanc
| | - Nicolas Preitner
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé SA, Vers-chez-les-Blanc
| | - Lijing Xin
- Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging (CIBM), EPFL
| | - Simone Astori
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Carmen Sandi
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| |
Collapse
|
42
|
Divergent Cellular Energetics, Glutamate Metabolism, and Mitochondrial Function Between Human and Mouse Cerebral Cortex. Mol Neurobiol 2022; 59:7495-7512. [PMID: 36201140 DOI: 10.1007/s12035-022-03053-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 09/24/2022] [Indexed: 10/10/2022]
Abstract
Disruptions of brain energy and neurotransmitter metabolism are associated with several pathological conditions including neurodegenerative diseases such as Alzheimer's disease. Transgenic rodent models, and in vitro preparations hereof, are often applied for studying pathological aspects of brain metabolism. However, despite the conserved cerebral development across mammalian species, distinct differences in cellular composition and structure may influence metabolism of the rodent and human brain. To address this, we investigated the metabolic function of acutely isolated brain slices and non-synaptic mitochondria obtained from the cerebral cortex of mice and neurosurgically resected neocortical tissue of humans. Utilizing dynamic isotope labeling with 13C-enriched metabolic substrates, we show that metabolism of glucose, acetate, β-hydroxybutyrate, and glutamine operates at lower rates in human cerebral cortical slices when compared to mouse slices. In contrast, human cerebral cortical slices display a higher capacity for converting exogenous glutamate into glutamine, which subsequently supports neuronal GABA synthesis, whereas mouse slices primarily convert glutamate into aspartate. In line with the reduced metabolic rate of the human brain slices, isolated non-synaptic mitochondria of the human cerebral cortex have a lower oxygen consumption rate when provided succinate as substrate. However, when provided pyruvate and malate, human mitochondria display a higher coupled respiration and lower proton leak, signifying a more efficient mitochondrial coupling compared to mouse mitochondria. This study reveals key differences between mouse and human brain metabolism concerning both neurons and astrocytes, which must be taken into account when applying in vitro rodent preparations as a model system of the human brain.
Collapse
|
43
|
Ilieva M, Aldana BI, Vinten KT, Hohmann S, Woofenden TW, Lukjanska R, Waagepetersen HS, Michel TM. Proteomic phenotype of cerebral organoids derived from autism spectrum disorder patients reveal disrupted energy metabolism, cellular components, and biological processes. Mol Psychiatry 2022; 27:3749-3759. [PMID: 35618886 DOI: 10.1038/s41380-022-01627-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/04/2022] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
The way in which brain morphology and proteome are remodeled during embryonal development, and how they are linked to the cellular metabolism, could be a key for elucidating the pathological mechanisms of certain neurodevelopmental disorders. Cerebral organoids derived from autism spectrum disorder (ASD) patients were generated to capture critical time-points in the neuronal development, and metabolism and protein expression were investigated. The early stages of development, when neurogenesis commences (day in vitro 39), appeared to be a critical timepoint in pathogenesis. In the first month of development, increased size in ASD-derived organoids were detected in comparison to the controls. The size of the organoids correlates with the number of proliferating cells (Ki-67 positive cells). A significant difference in energy metabolism and proteome phenotype was also observed in ASD organoids at this time point, specifically, prevalence of glycolysis over oxidative phosphorylation, decreased ATP production and mitochondrial respiratory chain activity, differently expressed cell adhesion proteins, cell cycle (spindle formation), cytoskeleton, and several transcription factors. Finally, ASD patients and controls derived organoids were clustered based on a differential expression of ten proteins-heat shock protein 27 (hsp27) phospho Ser 15, Pyk (FAK2), Elk-1, Rac1/cdc42, S6 ribosomal protein phospho Ser 240/Ser 244, Ha-ras, mTOR (FRAP) phospho Ser 2448, PKCα, FoxO3a, Src family phospho Tyr 416-at day 39 which could be defined as potential biomarkers and further investigated for potential drug development.
Collapse
Affiliation(s)
- Mirolyuba Ilieva
- Department of Psychiatry, Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Psychiatry in the Region of Southern Denmark, Odense University Hospital, Odense, Denmark. .,Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen SV, Denmark.
| | - Blanca Irene Aldana
- Neurometabolism Research Group, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Tore Vinten
- Neurometabolism Research Group, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sonja Hohmann
- Department of Psychiatry, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Psychiatry in the Region of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Thomas William Woofenden
- Neurometabolism Research Group, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Renate Lukjanska
- Department of Psychiatry, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Psychiatry in the Region of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Helle S Waagepetersen
- Neurometabolism Research Group, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tanja Maria Michel
- Department of Psychiatry, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Psychiatry in the Region of Southern Denmark, Odense University Hospital, Odense, Denmark
| |
Collapse
|
44
|
Li J, Zou Y, Yang J, Li Q, Bourne DG, Sweet M, Liu C, Guo A, Zhang S. Cultured Bacteria Provide Insight into the Functional Potential of the Coral-Associated Microbiome. mSystems 2022; 7:e0032722. [PMID: 35695425 PMCID: PMC9426491 DOI: 10.1128/msystems.00327-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/20/2022] [Indexed: 01/07/2023] Open
Abstract
Improving the availability of representative isolates from the coral microbiome is essential for investigating symbiotic mechanisms and applying beneficial microorganisms to improve coral health. However, few studies have explored the diversity of bacteria which can be isolated from a single species. Here, we isolated a total of 395 bacterial strains affiliated with 49 families across nine classes from the coral Pocillopora damicornis. Identification results showed that most of the strains represent potential novel bacterial species or genera. We also sequenced and assembled the genomes of 118 of these isolates, and then the putative functions of these isolates were identified based on genetic signatures derived from the genomes and this information was combined with isolate-specific phenotypic data. Genomic information derived from the isolates identified putative functions including nitrification and denitrification, dimethylsulfoniopropionate transformation, and supply of fixed carbon, amino acids, and B vitamins which may support their eukaryotic partners. Furthermore, the isolates contained genes associated with chemotaxis, biofilm formation, quorum sensing, membrane transport, signal transduction, and eukaryote-like repeat-containing and cell-cell attachment proteins, all of which potentially help the bacterium establish association with the coral host. Our work expands on the existing culture collection of coral-associated bacteria and provides important information on the metabolic potential of these isolates which can be used to refine understanding of the role of bacteria in coral health and are now available to be applied to novel strategies aimed at improving coral resilience through microbiome manipulation. IMPORTANCE Microbes underpin the health of corals which are the building blocks of diverse and productive reef ecosystems. Studying the culturable fraction of coral-associated bacteria has received less attention in recent times than using culture-independent molecular methods. However, the genomic and phenotypic characterization of isolated strains allows assessment of their functional role in underpinning coral health and identification of beneficial microbes for microbiome manipulation. Here, we isolated 395 bacterial strains from tissues of Pocillopora damicornis with many representing potentially novel taxa and therefore providing a significant contribution to coral microbiology through greatly enlarging the existing cultured coral-associated bacterial bank. Through analysis of the genomes obtained in this study for the coral-associated bacteria and coral host, we elucidate putative metabolic linkages and symbiotic establishment. The results of this study will help to elucidate the role of specific isolates in coral health and provide beneficial microbes for efforts aimed at improving coral health.
Collapse
Affiliation(s)
- Jie Li
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Marine Biotechnology of Hainan Province, Sanya Institute of Oceanology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Sanya National Marine Ecosystem Research Station, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Yiyang Zou
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Jian Yang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Marine Biotechnology of Hainan Province, Sanya Institute of Oceanology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Sanya National Marine Ecosystem Research Station, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Qiqi Li
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - David G. Bourne
- College of Science and Engineering, James Cook University, Townsville, Queensland, Australia
- Australian Institute of Marine Science, Townsville, Queensland, Australia
| | - Michael Sweet
- Aquatic Research Facility, Environmental Sustainability Research Centre, University of Derby, Derby, United Kingdom
| | - Cong Liu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Anjie Guo
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Si Zhang
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Marine Biotechnology of Hainan Province, Sanya Institute of Oceanology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Sanya National Marine Ecosystem Research Station, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
The effects of hypothermia on glutamate and γ-aminobutyric acid metabolism during ischemia in monkeys: a repeated-measures ANOVA study. Sci Rep 2022; 12:14470. [PMID: 36008544 PMCID: PMC9411555 DOI: 10.1038/s41598-022-18783-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
During an ischemic stroke, the brain releases various factors, including glutamate and γ-aminobutyric acid. Glutamate can cause neurotoxic effects through certain receptors and exacerbate neurological damage, while γ-aminobutyric acid as an inhibitory neurotransmitter can antagonize the excitotoxic effects of glutamate and enhance the tolerance of neurons to ischemia. Therefore, in this study, the content of amino acid neurotransmitters in brain tissue before ischemia, after 10 min of ischemia, hypothermic perfusion, and rewarming were analyzed by high-performance liquid chromatography-UV in an animal model of ischemic stroke generated by blocking the bilateral common carotid arteries of rhesus monkeys. The changes in amino acid neurotransmitters in the rhesus monkey brain during post-ischemia hypothermic perfusion and rewarming were investigated by statistical methods of repeated measures ANOVA, showing that the concentration change of glutamate had not only a temporal factor but also was influenced by temperature, and there was an interaction effect between the two. Time but not temperature affected the change in γ-aminobutyric acid concentration, and there was an interaction effect between the two. Accordingly, hypoperfusion exerts a protective effect during ischemia by inhibiting the release of excitatory amino acid neurotransmitters, while the antagonistic effect of GABA on Glu is not significant.
Collapse
|
46
|
Telegina DV, Antonenko AK, Fursova AZ, Kolosova NG. The glutamate/GABA system in the retina of male rats: effects of aging, neurodegeneration, and supplementation with melatonin and antioxidant SkQ1. Biogerontology 2022; 23:571-585. [PMID: 35969289 DOI: 10.1007/s10522-022-09983-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/20/2022] [Indexed: 11/02/2022]
Abstract
Glutamate and -aminobutyric acid (GABA) are the most abundant amino acids in the retina. An imbalance of the glutamate/GABA system is involved in the pathogenesis of various neurodegenerative disorders. Here we for the first time analyzed alterations of expression of glutamate- and GABA-synthesizing enzymes, transporters, and relevant receptors in the retina with age in Wistar rats and in senescence-accelerated OXYS rats who develop AMD-like retinopathy. We noted consistent age-dependent expression changes of GABAergic-system proteins (GAD67, GABA-T, and GAT1) in OXYS and Wistar rats: upregulation by age 3 months and downregulation at age 18 months. At a late stage of AMD-like retinopathy in OXYS rats (18 months), there was significant upregulation of glutaminase and downregulation of glutamine synthetase, possibly indicating an increasing level of glutamate in the retina. AMD-like-retinopathy development in the OXYS strain was accompanied by underexpression of glutamate transporter GLAST. Prolonged supplementation with both melatonin and SkQ1 (separately) suppressed the progression of the AMD-like pathology in OXYS rats without affecting the glutamate/GABA system but worsened the condition of the Wistar rat's retina during normal aging. We observed decreasing protein levels of glutamine synthetase, GLAST, and GABAAR1 and an increasing level of glutaminase in Wistar rats. In summary, both melatonin and mitochondrial antioxidant SkQ1 had different effect on the retinal glutamate / GABA in healthy Wistar and senescence-accelerated OXYS rats.
Collapse
|
47
|
Scotti-Muzzi E, Chile T, Vallada H, Otaduy MCG, Soeiro-de-Souza MG. Association between CACNA1C gene rs100737 polymorphism and glutamatergic neurometabolites in bipolar disorder. Eur Neuropsychopharmacol 2022; 59:26-35. [PMID: 35544990 DOI: 10.1016/j.euroneuro.2022.04.001] [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: 10/30/2021] [Revised: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 11/25/2022]
Abstract
Abnormalities in Ca2+ homeostasis in Bipolar Disorders (BD) have been associated with impairments in glutamatergic receptors and voltage-gated calcium channels. Increased anterior cingulate cortex (ACC) glutamatergic neurometabolites have been consistently disclosed in BD by proton magnetic resonance spectroscopy (1H-MRS). A single nucleotide polymorphism (SNP) in the CACNA1C gene (rs1006737), which encodes the alpha 1-C subunit of the L-type calcium channel, has been associated with BD and is reported to modulate intra-cellular Ca2+. Thus, this study aimed to explore the association of the CACNA1C genotype with ACC glutamatergic metabolites measured by 1H-MRS in both BD and HC subjects. A total of 194 subjects (121 euthymic BD type I patients and 73 healthy controls (HC) were genotyped for CACNA1C rs1006737, underwent a 3-Tesla 1H-MRS imaging examination and ACC glutamatergic metabolite were assessed. We found overall increased glutamatergic metabolites in AA carriers in BD. Specifically, higher Glx/Cr was observed in subjects with the AA genotype compared to both AG and GG in the overall sample (BD + HC). Also, female individuals in the BD group with AA genotype were found to have higher Glx/Cr compared to those with other genotypes. CACNA1C AA carriers in use of anticonvulsant medication had higher estimated Glutamine (Glx-Glu) than the other genotypes. Thus, this study suggest an association between calcium channel genetics and increased glutamatergic metabolites in BD, possibly playing a synergic role in intracellular Ca2+ overload and excitotoxicity.
Collapse
Affiliation(s)
- Estêvão Scotti-Muzzi
- Department of Psychiatry, University of São Paulo (FMUSP), Institute of Psychiatry, CEAPESQ, PROGRUDA, School of Medicine, Dr. Ovidio Pires de Campos s / n. Clinic Hospital, São Paulo 05403-010, Brazil.
| | - Thais Chile
- Genetics and Pharmacogenetics Unit (PROGENE), Institute of Psychiatry, School of Medicine, University of São Paulo (IPq-FMUSP), Brazil
| | - Homero Vallada
- Genetics and Pharmacogenetics Unit (PROGENE), Institute of Psychiatry, School of Medicine, University of São Paulo (IPq-FMUSP), Brazil
| | - Maria Concepción Garcia Otaduy
- Laboratory of Magnetic Resonance LIM44, Department and Institute of Radiology, University of São Paulo (InRad-FMUSP), Brazil
| | - Márcio Gerhardt Soeiro-de-Souza
- Department of Psychiatry, University of São Paulo (FMUSP), Institute of Psychiatry, CEAPESQ, PROGRUDA, School of Medicine, Dr. Ovidio Pires de Campos s / n. Clinic Hospital, São Paulo 05403-010, Brazil
| |
Collapse
|
48
|
Benedetti B, Weidenhammer A, Reisinger M, Couillard-Despres S. Spinal Cord Injury and Loss of Cortical Inhibition. Int J Mol Sci 2022; 23:5622. [PMID: 35628434 PMCID: PMC9144195 DOI: 10.3390/ijms23105622] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
After spinal cord injury (SCI), the destruction of spinal parenchyma causes permanent deficits in motor functions, which correlates with the severity and location of the lesion. Despite being disconnected from their targets, most cortical motor neurons survive the acute phase of SCI, and these neurons can therefore be a resource for functional recovery, provided that they are properly reconnected and retuned to a physiological state. However, inappropriate re-integration of cortical neurons or aberrant activity of corticospinal networks may worsen the long-term outcomes of SCI. In this review, we revisit recent studies addressing the relation between cortical disinhibition and functional recovery after SCI. Evidence suggests that cortical disinhibition can be either beneficial or detrimental in a context-dependent manner. A careful examination of clinical data helps to resolve apparent paradoxes and explain the heterogeneity of treatment outcomes. Additionally, evidence gained from SCI animal models indicates probable mechanisms mediating cortical disinhibition. Understanding the mechanisms and dynamics of cortical disinhibition is a prerequisite to improve current interventions through targeted pharmacological and/or rehabilitative interventions following SCI.
Collapse
Affiliation(s)
- Bruno Benedetti
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria; (B.B.); (A.W.); (M.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Annika Weidenhammer
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria; (B.B.); (A.W.); (M.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), 5020 Salzburg, Austria
| | - Maximilian Reisinger
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria; (B.B.); (A.W.); (M.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), 5020 Salzburg, Austria
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, 5020 Salzburg, Austria; (B.B.); (A.W.); (M.R.)
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), 5020 Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
49
|
Continuous Ingestion of Lacticaseibacillus rhamnosus JB-1 during Chronic Stress Ensures Neurometabolic and Behavioural Stability in Rats. Int J Mol Sci 2022; 23:ijms23095173. [PMID: 35563564 PMCID: PMC9106030 DOI: 10.3390/ijms23095173] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022] Open
Abstract
The intestinal microbiome composition and dietary supplementation with psychobiotics can result in neurochemical alterations in the brain, which are possible due to the presence of the brain–gut–microbiome axis. In the present study, magnetic resonance spectroscopy (MRS) and behavioural testing were used to evaluate whether treatment with Lacticaseibacillus rhamnosus JB-1 (JB-1) bacteria alters brain metabolites’ levels and behaviour during continuous exposure to chronic stress. Twenty Wistar rats were subjected to eight weeks of a chronic unpredictable mild stress protocol. Simultaneously, half of them were fed with JB-1 bacteria, and the second half was given a daily placebo. Animals were examined at three-time points: before starting the stress protocol and after five and eight weeks of stress onset. In the elevated plus maze behavioural test the placebo group displayed increased anxiety expressed by almost complete avoidance of exploration, while the JB-1 dietary supplementation mitigated anxiety which resulted in a longer exploration time. Hippocampal MRS measurements demonstrated a significant decrease in glutamine + glutathione concentration in the placebo group compared to the JB-1 bacteria-supplemented group after five weeks of stress. With the progression of stress, the decrease of glutamate, glutathione, taurine, and macromolecular concentrations were observed in the placebo group as compared to baseline. The level of brain metabolites in the JB-1-supplemented rats were stable throughout the experiment, with only the taurine level decreasing between weeks five and eight of stress. These data indicated that the JB-1 bacteria diet might stabilize levels of stress-related neurometabolites in rat brain and could prevent the development of anxiety/depressive-like behaviour.
Collapse
|
50
|
Kamel AY, Emtiazjoo AM, Adkins L, Shahmohammadi A, Alnuaimat H, Pelaez A, Machuca T, Pipkin M, Lee HW, Weiner ID, Chandrashekaran S. Hyperammonemia After Lung Transplantation: Systematic Review and a Mini Case Series. Transpl Int 2022; 35:10433. [PMID: 35620675 PMCID: PMC9128545 DOI: 10.3389/ti.2022.10433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/18/2022] [Indexed: 11/21/2022]
Abstract
Background: Hyperammonemia after lung transplantation (HALT) is a rare but serious complication with high mortality. This systematic review delineates possible etiologies of HALT and highlights successful strategies used to manage this fatal complication. Methods: Seven biomedical databases and grey literature sources were searched using keywords relevant to hyperammonemia and lung transplantation for publications between 1995 and 2020. Additionally, we retrospectively analyzed HALT cases managed at our institution between January 2016 and August 2018. Results: The systematic review resulted in 18 studies with 40 individual cases. The mean peak ammonia level was 769 μmol/L at a mean of 14.1 days post-transplant. The mortality due to HALT was 57.5%. In our cohort of 120 lung transplants performed, four cases of HALT were identified. The mean peak ammonia level was 180.5 μmol/L at a mean of 11 days after transplantation. HALT in all four patients was successfully treated using a multimodal approach with an overall mortality of 25%. Conclusion: The incidence of HALT (3.3%) in our institution is comparable to prior reports. Nonetheless, ammonia levels in our cohort were not as high as previously reported and peaked earlier. We attributed these significant differences to early recognition and prompt institution of multimodal treatment approach.
Collapse
Affiliation(s)
- Amir Y. Kamel
- Department of Pharmacy, UF Health Shands Hospital, College of Pharmacy, University of Florida, Gainesville, FL, United States
- *Correspondence: Amir Y. Kamel,
| | - Amir M. Emtiazjoo
- Division of Pulmonary, Critical Care and Sleep Medicine, UF Lung Transplant Program, College of Medicine, University of Florida Health Hospital, Gainesville, FL, United States
| | - Lauren Adkins
- College of Pharmacy Liaison Librarian, Health Science Center Libraries, Gainesville, FL, United States
| | - Abbas Shahmohammadi
- Division of Pulmonary, Critical Care and Sleep Medicine, UF Lung Transplant Program, College of Medicine, University of Florida Health Hospital, Gainesville, FL, United States
| | - Hassan Alnuaimat
- Division of Pulmonary, Critical Care and Sleep Medicine, UF Lung Transplant Program, College of Medicine, University of Florida Health Hospital, Gainesville, FL, United States
| | - Andres Pelaez
- Division of Pulmonary, Critical Care and Sleep Medicine, UF Lung Transplant Program, College of Medicine, University of Florida Health Hospital, Gainesville, FL, United States
| | - Tiago Machuca
- Division of Cardiothoracic Surgery, UF Lung Transplant Program, University of Florida Health Hospital, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Mauricio Pipkin
- Division of Cardiothoracic Surgery, UF Lung Transplant Program, University of Florida Health Hospital, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Hyun-wook Lee
- Division of Nephrology, Hypertension and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL, United States
| | - I. David Weiner
- Division of Nephrology, Hypertension and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL, United States
- North Florida/South Georgia Veterans Health System, Gainesville, FL, United States
| | - Satish Chandrashekaran
- Division of Pulmonary, Critical Care and Sleep Medicine, UF Lung Transplant Program, College of Medicine, University of Florida Health Hospital, Gainesville, FL, United States
| |
Collapse
|