1
|
Oh DR, Kim Y, Mun BY, Ko H, Kim Y, Kim MJ, Oh KN, Hong JA, Bae D. Rumex acetosa L. enhance learning and cognitive function by modulating NMDA receptor and BDNF pathways in vitro and in vivo. Metab Brain Dis 2025; 40:185. [PMID: 40244509 DOI: 10.1007/s11011-025-01608-8] [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: 09/12/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Rumex acetosa L. (RA), a member of the Polygonaceae family, is called sorrel and has been used as a vegetable and traditional medicine. RA has various bioactive functions; however, its effects on cognitive function remain unclear. Herein, we investigated the learning- and memory-enhancing effects of RA in rats using the Morris Water Maze (MWM) test. In addition, we evaluated the effect of RA on cognitive function in the hippocampus, both in vitro and in vivo, to identify the underlying molecular mechanisms. RA showed cognitive enhancing effects by activating cyclic AMP-responsive element-binding protein (CREB) via N-methyl-d-aspartate (NMDA) receptor 2 subunits (NR2A and NR2B), postsynaptic density protein-95 (PSD-95) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit (GluA1) signaling in primary cultured rat hippocampal neurons. For the in vivo experiments, RA was orally administered to SD rats for 26 days. RA showed significantly decreased escape latency on the first (acquisition) and last (retention) trials on the 2nd and 3rd training days and increased entry into the platform quadrant of time and distance on the probe trial. Furthermore, RA significantly increased NR2A, NR2B, PSD-95, and GluA1 the following downstream signaling extracellular signal-regulated kinase 1/2 (ERK1/2), calcium/calmodulin-dependent protein kinase II (CaMKII) phosphorylation, and brain-derived neurotrophic factor (BDNF) in the hippocampus. These results demonstrate that RA improves cognitive function, including synaptic plasticity, by regulating the NMDA receptor-dependent and BDNF/CREB signaling pathways.
Collapse
Affiliation(s)
- Dool-Ri Oh
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Yujin Kim
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Bo Yeong Mun
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Haeju Ko
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Yonguk Kim
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Moon Jong Kim
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Kyo-Nyeo Oh
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Ji-Ae Hong
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea
| | - Donghyuck Bae
- Jeonnam Bio Foundation, Jeonnam Institute of Natural Resources Research (JINR), 288, Woodland-gil, Jangheung-gun, 59338, Jeollanamdo, Republic of Korea.
| |
Collapse
|
2
|
Caccialupi Da Prato L, Rezzag Lebza A, Consumi A, Tessier M, Srinivasan A, Rivera C, Laurin J, Pellegrino C. Ectopic expression of the cation-chloride cotransporter KCC2 in blood exosomes as a biomarker for functional rehabilitation. Front Mol Neurosci 2025; 18:1522571. [PMID: 39974187 PMCID: PMC11835807 DOI: 10.3389/fnmol.2025.1522571] [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: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of disabilities in industrialized countries. Cognitive decline typically occurs in the chronic phase of the condition, following cellular and molecular processes. In this study, we described the use of KCC2, a neuronal-specific potassium-chloride cotransporter, as a potent biomarker to predict cognitive dysfunction after TBI. Methods Using neuronal and total exosome collections from the blood serum of the controls and patients with TBI, we were able to anticipate the decline in cognitive performance. Results After TBI, we observed a significant and persistent loss of KCC2 expression in the blood exosomes, which was correlated with the changes in the network activity and cellular processes such as secondary neurogenesis. Furthermore, we established a correlation between this decrease in KCC2 expression and the long-term consequences of brain trauma and identified a link between the loss of KCC2 expression and the emergence of depressive-like behavior observed in the mice. Conclusion We successfully validated our previous findings, supporting the potential therapeutic benefits of bumetanide in mitigating post-traumatic depression (PTD) following TBI. This effect was correlated with the recovery of KCC2 expression in the blood exosomes, the prevention of extensive neuronal loss among the interneurons, and changes in secondary neurogenesis.
Collapse
Affiliation(s)
| | | | - A. Consumi
- Inmed, INSERM, Aix-Marseille University, Marseille, France
| | - M. Tessier
- Inmed, INSERM, Aix-Marseille University, Marseille, France
| | - A. Srinivasan
- Division of Nanoscience and Technology, School of Life Sciences, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Academy of Higher Education and Research, Mysore, India
| | - C. Rivera
- Inmed, INSERM, Aix-Marseille University, Marseille, France
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - J. Laurin
- Inmed, INSERM, Aix-Marseille University, Marseille, France
| | - C. Pellegrino
- Inmed, INSERM, Aix-Marseille University, Marseille, France
| |
Collapse
|
3
|
Talty CE, Murphy S, VandeVord P. Mild traumatic brain injury gives rise to chronic depression-like behavior and associated alterations in glutamatergic protein expression. Neuroscience 2024; 560:198-210. [PMID: 39357641 DOI: 10.1016/j.neuroscience.2024.09.039] [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: 04/23/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
Mild traumatic brain injury (mTBI) is known to result in chronic somatic, cognitive, and emotional symptoms. Depression is commonly reported among individuals suffering from persistent concussion symptoms; however, the underlying mechanisms are not understood. The glutamatergic system has recently been linked with mTBI and depression due to reports of similar changes in expression of glutamatergic proteins. Using a closed-head controlled cortical impact (cCCI) model in adult male rats (n = 8/group), this study investigated the emergence of self-care deficits and changes in social interaction behaviors at four, eight and twelve weeks post-injury. Western blotting was used to assess associated changes in expression of glutamate transporters and N-methyl-D-aspartate (NMDA) receptor subunits at twelve weeks. Splash test results revealed deficits in self-care behaviors beginning at eight weeks, which continued through twelve weeks in the injury group. Injured animals also exhibited decreased preference for social novelty at four weeks and loss of desire for social interaction as a whole by twelve weeks. GluN1 was increased in injured animals compared to shams in the frontal cortex and amygdala, while decreased GLT-1 was observed in the hippocampus. Linear regression was performed to evaluate relationships between behavioral and molecular variables; the results suggested that injury affects these relationships in a region-dependent manner. Together, these results suggest that the development of chronic depression-like behavior was associated with changes in glutamatergic protein expression. Deeper investigations into how injury influences glutamatergic synaptic protein expression are needed, as this has the potential to affect circuit-level neurotransmission that drives depression-like behavior following mTBI.
Collapse
Affiliation(s)
- Caiti-Erin Talty
- Graduate Program in Translational Biology, Medicine & Health, Virginia Tech, 325 Stanger St, Blacksburg, VA 24061, USA
| | - Susan Murphy
- Department of Biomedical Engineering & Mechanics, Virginia Tech, 325 Stanger St, Blacksburg, VA 24061, USA
| | - Pamela VandeVord
- Department of Biomedical Engineering & Mechanics, Virginia Tech, 325 Stanger St, Blacksburg, VA 24061, USA; Veterans Affairs Medical Center, 1970 Roanoke Blvd, Salem, VA 24153, USA.
| |
Collapse
|
4
|
Bocchio M, Vorobyev A, Sadeh S, Brustlein S, Dard R, Reichinnek S, Emiliani V, Baude A, Clopath C, Cossart R. Functional networks of inhibitory neurons orchestrate synchrony in the hippocampus. PLoS Biol 2024; 22:e3002837. [PMID: 39401246 PMCID: PMC11501041 DOI: 10.1371/journal.pbio.3002837] [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: 05/16/2024] [Revised: 10/24/2024] [Accepted: 09/06/2024] [Indexed: 10/26/2024] Open
Abstract
Inhibitory interneurons are pivotal components of cortical circuits. Beyond providing inhibition, they have been proposed to coordinate the firing of excitatory neurons within cell assemblies. While the roles of specific interneuron subtypes have been extensively studied, their influence on pyramidal cell synchrony in vivo remains elusive. Employing an all-optical approach in mice, we simultaneously recorded hippocampal interneurons and pyramidal cells and probed the network influence of individual interneurons using optogenetics. We demonstrate that CA1 interneurons form a functionally interconnected network that promotes synchrony through disinhibition during awake immobility, while preserving endogenous cell assemblies. Our network model underscores the importance of both cell assemblies and dense, unspecific interneuron connectivity in explaining our experimental findings, suggesting that interneurons may operate not only via division of labor but also through concerted activity.
Collapse
Affiliation(s)
- Marco Bocchio
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
- Department of Psychology, Durham University, Durham, United Kingdom
| | - Artem Vorobyev
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Sadra Sadeh
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Sophie Brustlein
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Robin Dard
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Susanne Reichinnek
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Valentina Emiliani
- Wavefront-Engineering Microscopy Group, Photonics Department, Vision Institute, Sorbonne University, INSERM, CNRS, Paris, France
| | - Agnes Baude
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Claudia Clopath
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Rosa Cossart
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| |
Collapse
|
5
|
Lu W, Wen J. Neuroinflammation and Post-Stroke Depression: Focus on the Microglia and Astrocytes. Aging Dis 2024; 16:AD.2024.0214-1. [PMID: 38421829 PMCID: PMC11745440 DOI: 10.14336/ad.2024.0214-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
Post-stroke depression (PSD), a frequent and disabling complication of stroke, has a strong impact on almost thirty percent of stroke survivors. The pathogenesis of PSD is not completely clear so far. Neuroinflammation following stroke is one of underlying mechanisms that involves in the pathophysiology of PSD and plays an important function in the development of depression and is regarded as a sign of depression. During the neuroinflammation after ischemic stroke onset, both astrocytes and microglia undergo a series of morphological and functional changes and play pro-inflammatory or anti-inflammatory effect in the pathological process of stroke. Importantly, astrocytes and microglia exert dual roles in the pathological process of PSD due to the phenotypic transformation. We summarize the latest evidence of neuroinflammation involving in PSD in this review, focus on the phenotypic transformation of microglia and astrocytes following ischemic stroke and reveal the dual roles of both microglia and astrocytes in the PSD via modulating the neuroinflammation.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Machado DN, Durán-Carabali LE, Odorcyk FK, Carvalho AVS, Martini APR, Schlemmer LM, de Mattos MDM, Bernd GP, Dalmaz C, Netto CA. Bumetanide Attenuates Cognitive Deficits and Brain Damage in Rats Subjected to Hypoxia-Ischemia at Two Time Points of the Early Postnatal Period. Neurotox Res 2023; 41:526-545. [PMID: 37378827 DOI: 10.1007/s12640-023-00654-3] [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: 11/09/2022] [Revised: 05/24/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Neonatal hypoxia-ischemia (HI) is one of the main causes of tissue damage, cell death, and imbalance between neuronal excitation and inhibition and synaptic loss in newborns. GABA, the major inhibitory neurotransmitter of the central nervous system (CNS) in adults, is excitatory at the onset of neurodevelopment and its action depends on the chloride (Cl-) cotransporters NKCC1 (imports Cl-) and KCC2 (exports Cl-) expression. Under basal conditions, the NKCC1/KCC2 ratio decreases over neurodevelopment. Thus, changes in this ratio caused by HI may be related to neurological disorders. The present study evaluated the effects of bumetanide (NKCC cotransporters inhibitor) on HI impairments in two neurodevelopmental periods. Male Wistar rat pups, 3 (PND3) and 11 (PND11) days old, were submitted to the Rice-Vannucci model. Animals were divided into 3 groups: SHAM, HI-SAL, and HI-BUM, considering each age. Bumetanide was administered intraperitoneally at 1, 24, 48, and 72 h after HI. NKCC1, KCC2, PSD-95, and synaptophysin proteins were analyzed after the last injection by western blot. Negative geotaxis, righting reflex, open field, object recognition test, and Morris water maze task were performed to assess neurological reflexes, locomotion, and memory function. Tissue atrophy and cell death were evaluated by histology. Bumetanide prevented neurodevelopmental delay, hyperactivity, and declarative and spatial memory deficits. Furthermore, bumetanide reversed HI-induced brain tissue damage, reduced neuronal death and controlled GABAergic tone, maintained the NKCC1/KCC2 ratio, and synaptogenesis close to normality. Thereby, bumetanide appears to play an important therapeutic role in the CNS, protecting the animals against HI damage and improving functional performance.
Collapse
Affiliation(s)
- Diorlon Nunes Machado
- Graduate Program in Biological Sciences: Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre, RS, CEP: 90035-003, Brazil.
| | - Luz Elena Durán-Carabali
- Graduate Program in Biological Sciences: Physiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Felipe Kawa Odorcyk
- Graduate Program in Biological Sciences: Physiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Andrey Vinicios Soares Carvalho
- Graduate Program in Biological Sciences: Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre, RS, CEP: 90035-003, Brazil
| | - Ana Paula Rodrigues Martini
- Graduate Program in Biological Sciences: Neuroscience, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Livia Machado Schlemmer
- Graduate Program in Biological Sciences: Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre, RS, CEP: 90035-003, Brazil
| | - Marcel de Medeiros de Mattos
- Graduate Program in Biological Sciences: Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre, RS, CEP: 90035-003, Brazil
| | - Gabriel Pereira Bernd
- Graduate Program in Biological Sciences: Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre, RS, CEP: 90035-003, Brazil
| | - Carla Dalmaz
- Departament of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carlos Alexandre Netto
- Departament of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Departament of Physiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
7
|
Tessier M, Garcia MS, Goubert E, Blasco E, Consumi A, Dehapiot B, Tian L, Molinari F, Laurin J, Guillemot F, Hübner CA, Pellegrino C, Rivera C. Bumetanide induces post-traumatic microglia-interneuron contact to promote neurogenesis and recovery. Brain 2023; 146:4247-4261. [PMID: 37082944 PMCID: PMC10545516 DOI: 10.1093/brain/awad132] [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: 03/03/2022] [Revised: 02/20/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
Although the Na-K-Cl cotransporter (NKCC1) inhibitor bumetanide has prominent positive effects on the pathophysiology of many neurological disorders, the mechanism of action is obscure. Attention paid to elucidating the role of Nkcc1 has mainly been focused on neurons, but recent single cell mRNA sequencing analysis has demonstrated that the major cellular populations expressing NKCC1 in the cortex are non-neuronal. We used a combination of conditional transgenic animals, in vivo electrophysiology, two-photon imaging, cognitive behavioural tests and flow cytometry to investigate the role of Nkcc1 inhibition by bumetanide in a mouse model of controlled cortical impact (CCI). Here, we found that bumetanide rescues parvalbumin-positive interneurons by increasing interneuron-microglia contacts shortly after injury. The longitudinal phenotypic changes in microglia were significantly modified by bumetanide, including an increase in the expression of microglial-derived BDNF. These effects were accompanied by the prevention of CCI-induced decrease in hippocampal neurogenesis. Treatment with bumetanide during the first week post-CCI resulted in significant recovery of working and episodic memory as well as changes in theta band oscillations 1 month later. These results disclose a novel mechanism for the neuroprotective action of bumetanide mediated by an acceleration of microglial activation dynamics that leads to an increase in parvalbumin interneuron survival following CCI, possibly resulting from increased microglial BDNF expression and contact with interneurons. Salvage of interneurons may normalize ambient GABA, resulting in the preservation of adult neurogenesis processes as well as contributing to bumetanide-mediated improvement of cognitive performance.
Collapse
Affiliation(s)
- Marine Tessier
- Aix Marseille Univ, INSERM, INMED, 13273 Marseille, France
| | - Marta Saez Garcia
- Neuroscience Center, 00014 University of Helsinki, Helsinki, Finland
| | | | - Edith Blasco
- Aix Marseille Univ, INSERM, INMED, 13273 Marseille, France
| | | | - Benoit Dehapiot
- Aix Marseille Univ, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13288 Marseille, France
| | - Li Tian
- Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | | | - Jerome Laurin
- Aix Marseille Univ, INSERM, INMED, 13273 Marseille, France
| | | | - Christian A Hübner
- Institut für Humangenetik, Universitätsklinikum Jena, 07747 Jena, Germany
| | | | - Claudio Rivera
- Aix Marseille Univ, INSERM, INMED, 13273 Marseille, France
- Neuroscience Center, 00014 University of Helsinki, Helsinki, Finland
| |
Collapse
|
8
|
Woodman R, Miller C, Student J, Freeman K, Perl D, Lockette W. Alpha-methyltyrosine reduces the acute cardiovascular and behavioral sequelae in a murine model of traumatic brain injury. J Trauma Acute Care Surg 2023; 95:542-548. [PMID: 37165479 PMCID: PMC10545058 DOI: 10.1097/ta.0000000000004023] [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: 01/10/2023] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND Increased catecholamines contribute to heightened cardiovascular reactivity and behavioral deficits after traumatic brain injury (TBI); adrenergic receptor blockade has limited success in reducing adverse sequelae of TBI. Injury-induced increases in the synthesis of catecholamines could contribute to adverse outcomes in TBI. Inhibition of catecholamine synthesis with alpha-methyltyrosine (αMT) could offer a benefit after TBI. METHODS Original research trial in mice randomized to αMT (50 mg·kg -1 ·d -1 ) or vehicle for 1 week after TBI induced by controlled cortical impact. Primary outcomes of cardiovascular reactivity and behavioral deficits were assessed after 1 week. Secondary outcomes included blood brain barrier permeability and quantification of gene transcription whose products determine intraneuronal chloride concentrations, the release of catecholamines, and activation of the sympathetic nervous system. These genes were the alpha-2 adrenergic receptor ("Adra2c"), the sodium-potassium-chloride cotransporter ("Nkcc1"), and the potassium chloride cotransporter ("Kcc2"). We also assessed the effect of TBI and αMT on the neuronal chloride/bicarbonate exchanger ("Ae3"). RESULTS Traumatic brain injury-induced increases in blood pressure and cardiac reactivity were blocked by αMT. Inhibition of catecholamine synthesis decreased blood brain barrier leakage and improved behavioral outcomes after TBI. Traumatic brain injury diminished the transcription of Adra2c and enhanced expression of Nkcc1 while reducing Kcc2 transcription; αMT prevented the induction of the Nkcc1 by TBI without reversing the effects of TBI on Kcc2 expression; αMT also diminished Ae3 transcription. CONCLUSION Traumatic brain injury acutely increases cardiovascular reactivity and induces behavioral deficits in an αMT-sensitive manner, most likely by inducing Nkcc1 gene transcription. Alpha-methyltyrosine may prove salutary in the treatment of TBI by attenuating the enhanced expression of Nkcc1, minimizing blood brain barrier leakage, and diminishing central catecholamine and sympathetic output. We also found an unreported relationship between Kcc2 and the chloride/bicarbonate exchanger, which should be considered in the design of trials planned to manipulate central intraneuronal chloride concentrations following acute brain injury.
Collapse
|
9
|
Yang S, Zhang B, Wang D, Hu S, Wang W, Liu C, Wu Z, Yang C. Role of GABAergic system in the comorbidity of pain and depression. Brain Res Bull 2023:110691. [PMID: 37331640 DOI: 10.1016/j.brainresbull.2023.110691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/23/2023] [Accepted: 06/16/2023] [Indexed: 06/20/2023]
Abstract
Patients with chronic pain often suffer with depressive symptoms, and these two conditions can be aggravated by each other over time, leading to an increase in symptom intensity and duration. The comorbidity of pain and depression poses a significant challenge to human health and quality of life, as it is often difficult to diagnose early and treat effectively. Therefore, exploring the molecular mechanisms underlying the comorbidity of chronic pain and depression is crucial to identifying new therapeutic targets for treatment. However, understanding the pathogenesis of comorbidity requires examining interactions among multiple factors, which calls for an integrative perspective. While several studies have explored the role of the GABAergic system in pain and depression, fewer have examined its interactions with other systems involved in their comorbidity. Here, we review the evidence that the role of GABAergic system in the comorbidity of chronic pain and depression, as well as the interactions between the GABAergic system and other secondary systems involved in pain and depression comorbidity, providing a comprehensive understanding of their intricate interplay.
Collapse
Affiliation(s)
- Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Bingyuan Zhang
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, No. 399 Hailing South Road, Taizhou City, 225300, Jiangsu Province, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Wenli Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China.
| |
Collapse
|
10
|
Gharaylou Z, Shafaghi L, Pestehei SK, Hadjighassem M. Long-term bumetanide administration altered behavioral pattern in mosaic Down's Syndrome: A case report. APPLIED NEUROPSYCHOLOGY. CHILD 2023; 12:88-95. [PMID: 34860628 DOI: 10.1080/21622965.2021.2007481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The behavioral phenotypes emerge from cognitive architecture comprising attention, executive functions, and primary communication skills that all have shown remarkable deficits in Down's Syndrome (DS). These states arise from the proper functional interactions of the contributing neurotransmission and neuromodulation systems and other coding platforms. Gamma-aminobutyric acid (GABA) is an integral part of the neural interaction and regulation networks that its reverse action leads to broad detrimental consequences. This inhibitory substance needs an appropriate balance of co-transporters that largely shape the ionic milieu. Bumetanide, a specific NKCC1 inhibitor used for an eighteen-month interval, showed promising effects in restoring some behavior deficits in a fourteen-year-old boy diagnosed with genetically confirmed mosaic Down's Syndrome.
Collapse
Affiliation(s)
- Zeinab Gharaylou
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Shefa Neuroscience Research Center, Tehran, Iran
| | - Lida Shafaghi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahmoudreza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Chen Y, Hao C, Chen W, Cheng W, Li P, Shen J, Tong T, Yan S, Huang S, He T, Huang Z, Meng X. Anti-depressant effects of acupuncture: The insights from NLRP3 mediated pyroptosis and inflammation. Neurosci Lett 2022; 785:136787. [PMID: 35820551 DOI: 10.1016/j.neulet.2022.136787] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/13/2022] [Accepted: 07/07/2022] [Indexed: 11/19/2022]
Abstract
The NLRP3-mediated pyroptosis, which could affect inflammation response, plays a key role in the development of depression. Acupuncture has been shown to be an effective treatment for depression. In this study, we aimed to determine whether acupuncture could confer antidepressant activity via decreasing NLRP3-mediated pyroptosis by reducing inflammation. Here, depression model of rats was induced by chronic unpredictable mild stress (CUMS) for 4 weeks. Acupuncture group was subjected to acupuncture at the Shangxing (GV23) and Fengfu (GV16) acupoints for 20 min every other day (a total of 14 times). Fluoxetine group was administered with fluoxetine (2.1 mg/kg with the concentration of 0.21 mg/mL) by oral gavage (1 mL/100 g) once a day for 28 days. Rats' depression-like phenotypes were reflected with behavioral tests and biological detection methods. Results showed that acupuncture significantly improved the depression-like behavior of CUMS rat, suppressed the expressions of NLRP3, ASC, caspase-1, GSDMD, IL-1β, IL-18, HMGB1, IFN-γ, IL-6 and TNF-α in the serum and hippocampus, restored the %area of microglia, astrocytes and neuronal cells in the hippocampus. These indicate that acupuncture can prevent CUMS-induced depression-like behaviors by reducing NLRP3-mediated pyroptosis and inflammation.
Collapse
Affiliation(s)
- Yiping Chen
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, China; Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chongyao Hao
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Wenjie Chen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenjing Cheng
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Peng Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junliang Shen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Tao Tong
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Simin Yan
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuqiong Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Tianwei He
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhiyong Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xianjun Meng
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, China; Shenzhen Research Institute of Xiamen University, Shenzhen, China.
| |
Collapse
|
12
|
Du Y, Shi Y, Wang X, Song H, Wang X, Hao Y, Zhao Y, Guo X, Shi M, Gong M, Song L, Wang S, Gao Y, Shi H. Hippocampal Semaphorin 3B Improves Depression-like Behaviours in Mice by Upregulating Synaptic Plasticity and Inhibiting Neuronal Apoptosis. J Neurochem 2022; 163:133-148. [PMID: 35892177 DOI: 10.1111/jnc.15680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 07/03/2022] [Accepted: 07/20/2022] [Indexed: 12/01/2022]
Abstract
Depression is a global health problem, and there is a pressing need for a better understanding of its pathogenesis. Semaphorin 3B (Sema 3B) is an important axon guidance molecule that is primarily expressed in neurons and contributes to synaptic plasticity. Our previous studies using a high-throughput microarray assay suggested that Sema 3B expression was tremendously decreased during the development of depression, but the specific role and mechanisms of Sema 3B in depression are still unknown. Herein, we report that levels of Sema 3B protein are decreased in the hippocampus and serum of chronic mild stress (CMS)-treated mice. Increasing the levels of Sema 3B, either by injecting AAV-Sema 3B into the hippocampus or by injecting recombinant Sema 3B protein into the lateral ventricles, alleviated CMS-induced depression-like behaviours and enhanced the resistance to acute stress by increasing dendritic spine density in hippocampal neurons. In contrast, interfering the function of Sema 3B by injecting anti-Sema 3B antibody into the lateral ventricles decreased the resistance to acute stress. In vitro, corticosterone (CORT) treatment decreased survival rate and protein levels of Sema 3B and synapse-associated proteins in HT22 cells. Overexpression of Sema 3B improved the decreased survival rate caused by CORT by inhibiting apoptosis and increasing levels of synaptic-associated proteins. And knockdown of Sema 3B reduces the cellular resistance to CORT and the levels of synapse-associated proteins. These findings represent the first evidence for the neuroprotective mechanism of Sema 3B against stresses, suggesting that Sema 3B could be a promising novel target for the prevention and treatment of depression.
Collapse
Affiliation(s)
- Yuru Du
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Yun Shi
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China.,Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of, China, Shijiazhuang, Hebei
| | - Xi Wang
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Han Song
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Xinhao Wang
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Ying Hao
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Ye Zhao
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Xiangfei Guo
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Mengxu Shi
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Miao Gong
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Li Song
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Sheng Wang
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Yuan Gao
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Haishui Shi
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.,Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
13
|
Savardi A, Borgogno M, De Vivo M, Cancedda L. Pharmacological tools to target NKCC1 in brain disorders. Trends Pharmacol Sci 2021; 42:1009-1034. [PMID: 34620512 DOI: 10.1016/j.tips.2021.09.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/27/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023]
Abstract
The chloride importer NKCC1 and the chloride exporter KCC2 are key regulators of neuronal chloride concentration. A defective NKCC1/KCC2 expression ratio is associated with several brain disorders. Preclinical/clinical studies have shown that NKCC1 inhibition by the United States FDA-approved diuretic bumetanide is a potential therapeutic strategy in preclinical/clinical studies of multiple neurological conditions. However, bumetanide has poor brain penetration and causes unwanted diuresis by inhibiting NKCC2 in the kidney. To overcome these issues, a growing number of studies have reported more brain-penetrating and/or selective bumetanide prodrugs, analogs, and new molecular entities. Here, we review the evidence for NKCC1 pharmacological inhibition as an effective strategy to manage neurological disorders. We also discuss the advantages and limitations of bumetanide repurposing and the benefits and risks of new NKCC1 inhibitors as therapeutic agents for brain disorders.
Collapse
Affiliation(s)
- Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy; Dulbecco Telethon Institute, 00185 Rome, Italy; Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Marco Borgogno
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Marco De Vivo
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy; Dulbecco Telethon Institute, 00185 Rome, Italy.
| |
Collapse
|
14
|
Taubes A, Nova P, Zalocusky KA, Kosti I, Bicak M, Zilberter MY, Hao Y, Yoon SY, Oskotsky T, Pineda S, Chen B, Jones EAA, Choudhary K, Grone B, Balestra ME, Chaudhry F, Paranjpe I, De Freitas J, Koutsodendris N, Chen N, Wang C, Chang W, An A, Glicksberg BS, Sirota M, Huang Y. Experimental and real-world evidence supporting the computational repurposing of bumetanide for APOE4-related Alzheimer's disease. NATURE AGING 2021; 1:932-947. [PMID: 36172600 PMCID: PMC9514594 DOI: 10.1038/s43587-021-00122-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
The evident genetic, pathological, and clinical heterogeneity of Alzheimer's disease (AD) poses challenges for traditional drug development. We conducted a computational drug repurposing screen for drugs to treat apolipoprotein (apo) E4-related AD. We first established apoE-genotype-dependent transcriptomic signatures of AD by analyzing publicly-available human brain database. We then queried these signatures against the Connectivity Map database containing transcriptomic perturbations of >1300 drugs to identify those that best reverse apoE-genotype-specific AD signatures. Bumetanide was identified as a top drug for apoE4 AD. Bumetanide treatment of apoE4 mice without or with Aβ accumulation rescued electrophysiological, pathological, or cognitive deficits. Single-nucleus RNA-sequencing revealed transcriptomic reversal of AD signatures in specific cell types in these mice, a finding confirmed in apoE4-iPSC-derived neurons. In humans, bumetanide exposure was associated with a significantly lower AD prevalence in individuals over the age of 65 in two electronic health record databases, suggesting effectiveness of bumetanide in preventing AD.
Collapse
Affiliation(s)
- Alice Taubes
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA
| | - Phil Nova
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA
| | - Kelly A. Zalocusky
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Idit Kosti
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA, USA
| | - Mesude Bicak
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Misha Y. Zilberter
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Tomiko Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA, USA
| | - Silvia Pineda
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Bin Chen
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA
| | - Emily A. Aery Jones
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA
| | - Krishna Choudhary
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Maureen E. Balestra
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Fayzan Chaudhry
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Ishan Paranjpe
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Jessica De Freitas
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Development and Stem Cell Biology Graduate Program, University of California, San Francisco, CA 94143, USA
| | - Nuo Chen
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Celine Wang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - William Chang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Alice An
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Benjamin S. Glicksberg
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10065, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA, USA
- Correspondence: Yadong Huang () or Marina Sirota ()
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA 94143, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, CA 94143, USA
- Department of Pathology, University of California, San Francisco, CA 94143, USA
- Correspondence: Yadong Huang () or Marina Sirota ()
| |
Collapse
|
15
|
Matsumoto D, Ushio S, Wada Y, Noda Y, Esumi S, Izushi Y, Kitamura Y, Sendo T. Bumetanide prevents diazepam-modified anxiety-like behavior in lipopolysaccharide-treated mice. Eur J Pharmacol 2021; 904:174195. [PMID: 34004209 DOI: 10.1016/j.ejphar.2021.174195] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/27/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
Benzodiazepine receptor agonists are widely prescribed therapeutic agents that alter gamma-aminobutyric acid (GABA)A receptor activity and have anxiolytic effects. Post-operative use of benzodiazepines is a risk factor of delirium. Inflammatory conditions alter the anxiolytic effects of benzodiazepine. We investigated the effect of diazepam, a typical benzodiazepine anxiolytic, on changes in the emotional behavior of mice in a hole-board test after lipopolysaccharide (LPS) treatment. Diazepam dose-dependently increased the number of head-dips at doses that did not alter locomotor activity; however, diazepam dose-dependently significantly decreased the number of head-dips at doses that did not alter locomotor activity in LPS-treated mice. Flumazenil, a benzodiazepine receptor antagonist, normalized the decrease in head-dipping behavior caused by diazepam treatment in normal and LPS-treated mice. The decrease of the head-dipping effect caused by diazepam was attenuated by minocycline in LPS-treated mice. We further found that the decrease in head-dipping behavior caused by diazepam was blocked by bumetanide, a Na+-K+-2Cl- cotransporter isoform 1 (NKCC1) antagonist, in LPS-treated mice. These findings suggest that diazepam induces the anxiety-like behavior under inflammation conditions, and may cause the GABAA receptor dysfunction associated with the chloride plasticity mediated by NKCC1, which contributes to benzodiazepine-induced delirium after surgery.
Collapse
Affiliation(s)
- Daiki Matsumoto
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Soichiro Ushio
- Department of Pharmacy, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yudai Wada
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Yukiko Noda
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Satoru Esumi
- Department of Pharmacy, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yasuhisa Izushi
- Department of Pharmacotherapy, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama, 703-8516, Japan
| | - Yoshihisa Kitamura
- Department of Pharmacy, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan; Department of Pharmacotherapy, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama, 703-8516, Japan.
| | - Toshiaki Sendo
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan; Department of Pharmacy, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
16
|
Santana-Gomez CE, Medel-Matus JS, Rundle BK. Animal models of post-traumatic epilepsy and their neurobehavioral comorbidities. Seizure 2021; 90:9-16. [DOI: 10.1016/j.seizure.2021.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 12/30/2022] Open
|
17
|
Hugues N, Pellegrino C, Rivera C, Berton E, Pin-Barre C, Laurin J. Is High-Intensity Interval Training Suitable to Promote Neuroplasticity and Cognitive Functions after Stroke? Int J Mol Sci 2021; 22:3003. [PMID: 33809413 PMCID: PMC7998434 DOI: 10.3390/ijms22063003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke-induced cognitive impairments affect the long-term quality of life. High-intensity interval training (HIIT) is now considered a promising strategy to enhance cognitive functions. This review is designed to examine the role of HIIT in promoting neuroplasticity processes and/or cognitive functions after stroke. The various methodological limitations related to the clinical relevance of studies on the exercise recommendations in individuals with stroke are first discussed. Then, the relevance of HIIT in improving neurotrophic factors expression, neurogenesis and synaptic plasticity is debated in both stroke and healthy individuals (humans and rodents). Moreover, HIIT may have a preventive role on stroke severity, as found in rodents. The potential role of HIIT in stroke rehabilitation is reinforced by findings showing its powerful neurogenic effect that might potentiate cognitive benefits induced by cognitive tasks. In addition, the clinical role of neuroplasticity observed in each hemisphere needs to be clarified by coupling more frequently to cellular/molecular measurements and behavioral testing.
Collapse
Affiliation(s)
- Nicolas Hugues
- INMED, INSERM, Aix-Marseille University, 13007 Marseille, France; (N.H.); (C.P.); (C.R.)
- CNRS, ISM, Aix-Marseille University, 13007 Marseille, France; (E.B.); (C.P.-B.)
| | - Christophe Pellegrino
- INMED, INSERM, Aix-Marseille University, 13007 Marseille, France; (N.H.); (C.P.); (C.R.)
| | - Claudio Rivera
- INMED, INSERM, Aix-Marseille University, 13007 Marseille, France; (N.H.); (C.P.); (C.R.)
| | - Eric Berton
- CNRS, ISM, Aix-Marseille University, 13007 Marseille, France; (E.B.); (C.P.-B.)
| | - Caroline Pin-Barre
- CNRS, ISM, Aix-Marseille University, 13007 Marseille, France; (E.B.); (C.P.-B.)
| | - Jérôme Laurin
- INMED, INSERM, Aix-Marseille University, 13007 Marseille, France; (N.H.); (C.P.); (C.R.)
| |
Collapse
|
18
|
Khirug S, Soni S, Saez Garcia M, Tessier M, Zhou L, Kulesskaya N, Rauvala H, Lindholm D, Ludwig A, Molinari F, Rivera C. Protective Role of Low Ethanol Administration Following Ischemic Stroke via Recovery of KCC2 and p75 NTR Expression. Mol Neurobiol 2021; 58:1145-1161. [PMID: 33099743 PMCID: PMC7878264 DOI: 10.1007/s12035-020-02176-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/14/2020] [Indexed: 01/31/2023]
Abstract
A striking result from epidemiological studies show a correlation between low alcohol intake and lower incidence for ischemic stroke and severity of derived brain injury. Although reduced apoptosis and inflammation has been suggested to be involved, little is known about the mechanism mediating this effect in vivo. Increase in intracellular chloride concentration and derived depolarizing GABAAR-mediated transmission are common consequences following various brain injuries and are caused by the abnormal expression levels of the chloride cotransporters NKCC1 and KCC2. Downstream pro-apoptotic signaling through p75NTR may link GABAA depolarization with post-injury neuronal apoptosis. Here, we show that changes in GABAergic signaling, Cl- homeostasis, and expression of chloride cotransporters in the post-traumatic mouse brain can be significantly reduced by administration of 3% ethanol to the drinking water. Ethanol-induced upregulation of KCC2 has a positive impact on neuronal survival, preserving a large part of the cortical peri-infarct zone, as well as preventing the massive post-ischemic upregulation of the pro-apoptotic protein p75NTR. Importantly, intracortical multisite in vivo recordings showed that ethanol treatment could significantly ameliorate stroke-induced reduction in cortical activity. This surprising finding discloses a pathway triggered by low concentration of ethanol as a novel therapeutically relevant target.
Collapse
Affiliation(s)
- Stanislav Khirug
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland.
| | - Shetal Soni
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Marta Saez Garcia
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Marine Tessier
- INMED (INSERM U1249), Aix-Marseille Université, Marseille, France
| | - Liang Zhou
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Natalia Kulesskaya
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Heikki Rauvala
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Dan Lindholm
- Medicum, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Helsinki, Finland
| | - Anastasia Ludwig
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | | | - Claudio Rivera
- Neuroscience Center-HiLIFE, University of Helsinki, 00014, Helsinki, Finland.
- INMED (INSERM U1249), Aix-Marseille Université, Marseille, France.
| |
Collapse
|
19
|
Lowjaga KAAT, Kirstgen M, Müller SF, Goldmann N, Lehmann F, Glebe D, Geyer J. Long-term trans-inhibition of the hepatitis B and D virus receptor NTCP by taurolithocholic acid. Am J Physiol Gastrointest Liver Physiol 2021; 320:G66-G80. [PMID: 33174454 DOI: 10.1152/ajpgi.00263.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human hepatic bile acid transporter Na+/taurocholate cotransporting polypeptide (NTCP) represents the liver-specific entry receptor for the hepatitis B and D viruses (HBV/HDV). Chronic hepatitis B and D affect several million people worldwide, but treatment options are limited. Recently, HBV/HDV entry inhibitors targeting NTCP have emerged as promising novel drug candidates. Nevertheless, the exact molecular mechanism that NTCP uses to mediate virus binding and entry into hepatocytes is still not completely understood. It is already known that human NTCP mRNA expression is downregulated under cholestasis. Furthermore, incubation of rat hepatocytes with the secondary bile acid taurolithocholic acid (TLC) triggers internalization of the rat Ntcp protein from the plasma membrane. In the present study, the long-term inhibitory effect of TLC on transport function, HBV/HDV receptor function, and membrane expression of human NTCP were analyzed in HepG2 and human embryonic kidney (HEK293) cells stably overexpressing NTCP. Even after short-pulse preincubation, TLC had a significant long-lasting inhibitory effect on the transport function of NTCP, but the NTCP protein was still present at the plasma membrane. Furthermore, binding of the HBV/HDV myr-preS1 peptide and susceptibility for in vitro HDV infection were significantly reduced by TLC preincubation. We hypothesize that TLC rapidly accumulates in hepatocytes and mediates long-lasting trans-inhibition of the transport and receptor function of NTCP via a particular TLC-binding site at an intracellularly accessible domain of NTCP. Physiologically, this trans-inhibition might protect hepatocytes from toxic overload of bile acids. Pharmacologically, it provides an interesting novel NTCP target site for potential long-acting HBV/HDV entry inhibitors.NEW & NOTEWORTHY The hepatic bile acid transporter NTCP is a high-affinity receptor for hepatitis B and D viruses. This study shows that TLC rapidly accumulates in NTCP-expressing hepatoma cells and mediates long-lasting trans-inhibition of NTCP's transporter and receptor function via an intracellularly accessible domain, without substantially affecting its membrane expression. This domain is a promising novel NTCP target site for pharmacological long-acting HBV/HDV entry inhibitors.
Collapse
Affiliation(s)
- Kira A A T Lowjaga
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Michael Kirstgen
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Simon F Müller
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Nora Goldmann
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Felix Lehmann
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Dieter Glebe
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Joachim Geyer
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
20
|
Rahman S, Alzarea S. Glial mechanisms underlying major depressive disorder: Potential therapeutic opportunities. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 167:159-178. [DOI: 10.1016/bs.pmbts.2019.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|