1
|
Michetti C, Ferrante D, Parisi B, Ciano L, Prestigio C, Casagrande S, Martinoia S, Terranova F, Millo E, Valente P, Giovedi' S, Benfenati F, Baldelli P. Low glycemic index diet restrains epileptogenesis in a gender-specific fashion. Cell Mol Life Sci 2023; 80:356. [PMID: 37947886 PMCID: PMC10638170 DOI: 10.1007/s00018-023-04988-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 11/12/2023]
Abstract
Dietary restriction, such as low glycemic index diet (LGID), have been successfully used to treat drug-resistant epilepsy. However, if such diet could also counteract antiepileptogenesis is still unclear. Here, we investigated whether the administration of LGID during the latent pre-epileptic period, prevents or delays the appearance of the overt epileptic phenotype. To this aim, we used the Synapsin II knockout (SynIIKO) mouse, a model of temporal lobe epilepsy in which seizures manifest 2-3 months after birth, offering a temporal window in which LGID may affect epileptogenesis. Pregnant SynIIKO mice were fed with either LGID or standard diet during gestation and lactation. Both diets were maintained in weaned mice up to 5 months of age. LGID delayed the seizure onset and induced a reduction of seizures severity only in female SynIIKO mice. In parallel with the epileptic phenotype, high-density multielectrode array recordings revealed a reduction of frequency, amplitude, duration, velocity of propagation and spread of interictal events by LGID in the hippocampus of SynIIKO females, but not mutant males, confirming the gender-specific effect. ELISA-based analysis revealed that LGID increased cortico-hippocampal allopregnanolone (ALLO) levels only in females, while it was unable to affect ALLO plasma concentrations in either sex. The results indicate that the gender-specific interference of LGID with the epileptogenic process can be ascribed to a gender-specific increase in cortical ALLO, a neurosteroid known to strengthen GABAergic transmission. The study highlights the possibility of developing a personalized gender-based therapy for temporal lobe epilepsy.
Collapse
Affiliation(s)
- Caterina Michetti
- Department of Experimental Medicine, University of Genova, Genoa, Italy.
- Center for Synaptic Neuroscience and Technology, Italian Institute of Technology, Genoa, Italy.
| | - Daniele Ferrante
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Barbara Parisi
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Lorenzo Ciano
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- Center for Synaptic Neuroscience and Technology, Italian Institute of Technology, Genoa, Italy
| | - Cosimo Prestigio
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Silvia Casagrande
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Sergio Martinoia
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genova, Genoa, Italy
| | - Fabio Terranova
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genova, Genoa, Italy
| | - Enrico Millo
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Pierluigi Valente
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- IRCCS, Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Giovedi'
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- IRCCS, Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Italian Institute of Technology, Genoa, Italy
- IRCCS, Ospedale Policlinico San Martino, Genoa, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genova, Genoa, Italy.
- IRCCS, Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
2
|
Delgado-Zabalza L, Mallet NP, Glangetas C, Dabee G, Garret M, Miguelez C, Baufreton J. Targeting parvalbumin-expressing neurons in the substantia nigra pars reticulata restores motor function in parkinsonian mice. Cell Rep 2023; 42:113287. [PMID: 37843977 DOI: 10.1016/j.celrep.2023.113287] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 10/18/2023] Open
Abstract
The activity of substantia nigra pars reticulata (SNr) neurons, the main output structure of basal ganglia, is altered in Parkinson's disease (PD). However, neither the underlying mechanisms nor the type of neurons responsible for PD-related motor dysfunctions have been elucidated yet. Here, we show that parvalbumin-expressing SNr neurons (SNr-PV+) occupy dorsolateral parts and possess specific electrophysiological properties compared with other SNr cells. We also report that only SNr-PV+ neurons' intrinsic excitability is reduced by downregulation of sodium leak channels in a PD mouse model. Interestingly, in anesthetized parkinsonian mice in vivo, SNr-PV+ neurons display a bursty pattern of activity dependent on glutamatergic tone. Finally, we demonstrate that chemogenetic inhibition of SNr-PV+ neurons is sufficient to alleviate motor impairments in parkinsonian mice. Overall, our findings establish cell-type-specific dysfunction in experimental parkinsonism in the SNr and provide a potential cellular therapeutic target to alleviate motor symptoms in PD.
Collapse
Affiliation(s)
- Lorena Delgado-Zabalza
- University Bordeaux, CNRS, IMN, UMR 5293, 33000 Bordeaux, France; Department of Pharmacology. University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Nicolas P Mallet
- University Bordeaux, CNRS, IMN, UMR 5293, 33000 Bordeaux, France
| | | | - Guillaume Dabee
- University Bordeaux, CNRS, IMN, UMR 5293, 33000 Bordeaux, France
| | - Maurice Garret
- University Bordeaux, CNRS, INCIA, UMR 5287, 33000 Bordeaux, France
| | - Cristina Miguelez
- Department of Pharmacology. University of the Basque Country (UPV/EHU), 48940 Leioa, Spain; Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Jérôme Baufreton
- University Bordeaux, CNRS, IMN, UMR 5293, 33000 Bordeaux, France.
| |
Collapse
|
3
|
Dhoundiyal A, Goeschl V, Boehm S, Kubista H, Hotka M. Glycerol-3-Phosphate Shuttle Is a Backup System Securing Metabolic Flexibility in Neurons. J Neurosci 2022; 42:7339-7354. [PMID: 35999055 PMCID: PMC9525167 DOI: 10.1523/jneurosci.0193-22.2022] [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: 01/17/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Electrical activity in neurons is highly energy demanding and accompanied by rises in cytosolic Ca2+ Cytosolic Ca2+, in turn, secures energy supply by pushing mitochondrial metabolism either through augmented NADH (nicotinamide adenine dinucleotide) transfer into mitochondria via the malate-aspartate shuttle (MAS) or via direct activation of dehydrogenases of the TCA cycle after passing into the matrix through the mitochondrial Ca2+ uniporter (MCU). Another Ca2+-sensitive booster of mitochondrial ATP synthesis is the glycerol-3-phosphate shuttle (G3PS), whose role in neuronal energy supply has remained elusive. Essential components of G3PS are expressed in hippocampal neurons. Single neuron metabolic measurements in primary hippocampal cultures derived from rat pups of either sex reveal only moderate, if any, constitutive activity of G3PS. However, during electrical activity neurons fully rely on G3PS when MAS and MCU are unavailable. Under these conditions, G3PS is required for appropriate action potential firing. Accordingly, G3PS safeguards metabolic flexibility of neurons to cope with energy demands of electrical signaling.SIGNIFICANCE STATEMENT Ca2+ ions are known to provide a link between the energy-demanding electrical activity and an adequate ATP supply in neurons. To do so, Ca2+ acts both from outside and inside of the mitochondrial inner membrane. Neuronal function critically depends on this regulation, and its defects are often found in various neurologic disorders. Although interest in neuronal metabolism has increased, many aspects thereof have remained unresolved. In particular, a Ca2+-sensitive NADH (nicotinamide adenine dinucleotide) shuttling system, the glycerol-3-phosphate shuttle, has been largely ignored with respect to its function in neurons. Our results demonstrate that this shuttle is functional in hippocampal neurons and safeguards ATP supply and appropriate action potential firing when malate aspartate shuttle and mitochondrial Ca2+ uniporter are unavailable, thereby ensuring neuronal metabolic flexibility.
Collapse
Affiliation(s)
- Ankit Dhoundiyal
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Vanessa Goeschl
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Boehm
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Helmut Kubista
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Matej Hotka
- Center of Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
4
|
Qi J, Gan L, Fang J, Zhang J, Yu X, Guo H, Cai D, Cui H, Gou L, Deng J, Wang Z, Zuo Z. Beta-Hydroxybutyrate: A Dual Function Molecular and Immunological Barrier Function Regulator. Front Immunol 2022; 13:805881. [PMID: 35784364 PMCID: PMC9243231 DOI: 10.3389/fimmu.2022.805881] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 05/09/2022] [Indexed: 12/27/2022] Open
Abstract
Ketone bodies are crucial intermediate metabolites widely associated with treating metabolic diseases. Accumulating evidence suggests that ketone bodies may act as immunoregulators in humans and animals to attenuate pathological inflammation through multiple strategies. Although the clues are scattered and untrimmed, the elevation of these ketone bodies in the circulation system and tissues induced by ketogenic diets was reported to affect the immunological barriers, an important part of innate immunity. Therefore, beta-hydroxybutyrate, a key ketone body, might also play a vital role in regulating the barrier immune systems. In this review, we retrospected the endogenous ketogenesis in animals and the dual roles of ketone bodies as energy carriers and signal molecules focusing on beta-hydroxybutyrate. In addition, the research regarding the effects of beta-hydroxybutyrate on the function of the immunological barrier, mainly on the microbiota, chemical, and physical barriers of the mucosa, were outlined and discussed. As an inducible endogenous metabolic small molecule, beta-hydroxybutyrate deserves delicate investigations focusing on its immunometabolic efficacy. Comprehending the connection between ketone bodies and the barrier immunological function and its underlining mechanisms may help exploit individualised approaches to treat various mucosa or skin-related diseases.
Collapse
Affiliation(s)
- Jiancheng Qi
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Linli Gan
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Fang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jizong Zhang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xin Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hongrui Guo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dongjie Cai
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hengmin Cui
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liping Gou
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhisheng Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Zhicai Zuo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Zhicai Zuo,
| |
Collapse
|
5
|
Pietrzak D, Kasperek K, Rękawek P, Piątkowska-Chmiel I. The Therapeutic Role of Ketogenic Diet in Neurological Disorders. Nutrients 2022; 14:1952. [PMID: 35565918 PMCID: PMC9102882 DOI: 10.3390/nu14091952] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/30/2022] [Accepted: 05/04/2022] [Indexed: 02/01/2023] Open
Abstract
The ketogenic diet (KD) is a high-fat, low-carbohydrate and adequate-protein diet that has gained popularity in recent years in the context of neurological diseases (NDs). The complexity of the pathogenesis of these diseases means that effective forms of treatment are still lacking. Conventional therapy is often associated with increasing tolerance and/or drug resistance. Consequently, more effective therapeutic strategies are being sought to increase the effectiveness of available forms of therapy and improve the quality of life of patients. For the moment, it seems that KD can provide therapeutic benefits in patients with neurological problems by effectively controlling the balance between pro- and antioxidant processes and pro-excitatory and inhibitory neurotransmitters, and modulating inflammation or changing the composition of the gut microbiome. In this review we evaluated the potential therapeutic efficacy of KD in epilepsy, depression, migraine, Alzheimer's disease and Parkinson's disease. In our opinion, KD should be considered as an adjuvant therapeutic option for some neurological diseases.
Collapse
Affiliation(s)
- Diana Pietrzak
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland; (K.K.); (P.R.)
| | | | | | - Iwona Piątkowska-Chmiel
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland; (K.K.); (P.R.)
| |
Collapse
|
6
|
Grieb B, Uppala S, Sapir G, Shaul D, Gomori JM, Katz-Brull R. Curbing action potential generation or ATP-synthase leads to a decrease in in-cell pyruvate dehydrogenase activity in rat cerebrum slices. Sci Rep 2021; 11:10211. [PMID: 33986346 PMCID: PMC8119472 DOI: 10.1038/s41598-021-89534-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/19/2021] [Indexed: 11/24/2022] Open
Abstract
Direct and real-time monitoring of cerebral metabolism exploiting the drastic increase in sensitivity of hyperpolarized 13C-labeled metabolites holds the potential to report on neural activity via in-cell metabolic indicators. Here, we followed the metabolic consequences of curbing action potential generation and ATP-synthase in rat cerebrum slices, induced by tetrodotoxin and oligomycin, respectively. The results suggest that pyruvate dehydrogenase (PDH) activity in the cerebrum is 4.4-fold higher when neuronal firing is unperturbed. The PDH activity was 7.4-fold reduced in the presence of oligomycin, and served as a pharmacological control for testing the ability to determine changes to PDH activity in viable cerebrum slices. These findings may open a path towards utilization of PDH activity, observed by magnetic resonance of hyperpolarized 13C-labeled pyruvate, as a reporter of neural activity.
Collapse
Affiliation(s)
- Benjamin Grieb
- Department of Radiology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, 9112001, Jerusalem, Israel. .,Department of Psychiatry and Psychotherapie I (Weissenau), Ulm University, ZfP Suedwuerttemberg, Ravensburg, Germany.
| | - Sivaranjan Uppala
- Department of Radiology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, 9112001, Jerusalem, Israel
| | - Gal Sapir
- Department of Radiology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, 9112001, Jerusalem, Israel
| | - David Shaul
- Department of Radiology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, 9112001, Jerusalem, Israel
| | - J Moshe Gomori
- Department of Radiology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, 9112001, Jerusalem, Israel
| | - Rachel Katz-Brull
- Department of Radiology, Hadassah Medical Center, The Faculty of Medicine, Hebrew University of Jerusalem, 9112001, Jerusalem, Israel. .,The Wohl Institute for Translational Medicine, Jerusalem, Israel.
| |
Collapse
|
7
|
Yang W, Pang D, Chen M, Du C, Jia L, Wang L, He Y, Jiang W, Luo L, Yu Z, Mao M, Yuan Q, Tang P, Xia X, Cui Y, Jing B, Platero A, Liu Y, Wei Y, Worley PF, Xiao B. Rheb mediates neuronal-activity-induced mitochondrial energetics through mTORC1-independent PDH activation. Dev Cell 2021; 56:811-825.e6. [PMID: 33725483 PMCID: PMC9096910 DOI: 10.1016/j.devcel.2021.02.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/29/2020] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
Neuronal activity increases energy consumption and requires balanced production to maintain neuronal function. How activity is coupled to energy production remains incompletely understood. Here, we report that Rheb regulates mitochondrial tricarboxylic acid cycle flux of acetyl-CoA by activating pyruvate dehydrogenase (PDH) to increase ATP production. Rheb is induced by synaptic activity and lactate and dynamically trafficked to the mitochondrial matrix through its interaction with Tom20. Mitochondria-localized Rheb protein is required for activity-induced PDH activation and ATP production. Cell-type-specific gain- and loss-of-function genetic models for Rheb reveal reciprocal changes in PDH phosphorylation/activity, acetyl-CoA, and ATP that are not evident with genetic or pharmacological manipulations of mTORC1. Mechanistically, Rheb physically associates with PDH phosphatase (PDP), enhancing its activity and association with the catalytic E1α-subunit of PDH to reduce PDH phosphorylation and increase its activity. Findings identify Rheb as a nodal point that balances neuronal activity and neuroenergetics via Rheb-PDH axis.
Collapse
Affiliation(s)
- Wanchun Yang
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Dejiang Pang
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Mina Chen
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Chongyangzi Du
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Lanlan Jia
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Luoling Wang
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Yunling He
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Wanxiang Jiang
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Liping Luo
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zongyan Yu
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Mengqian Mao
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qiuyun Yuan
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ping Tang
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaoqiang Xia
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yiyuan Cui
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Bo Jing
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China
| | - Alexander Platero
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yanhui Liu
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yuquan Wei
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Paul F Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Bo Xiao
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Shenzhen 518055, People's Republic of China.
| |
Collapse
|
8
|
Adams S, Zubov T, Bueschke N, Santin JM. Neuromodulation or energy failure? Metabolic limitations silence network output in the hypoxic amphibian brainstem. Am J Physiol Regul Integr Comp Physiol 2021; 320:R105-R116. [PMID: 33175586 PMCID: PMC7948128 DOI: 10.1152/ajpregu.00209.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/10/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
Hypoxia tolerance in the vertebrate brain often involves chemical modulators that arrest neuronal activity to conserve energy. However, in intact networks, it can be difficult to determine whether hypoxia triggers modulators to stop activity in a protective manner or whether activity stops because rates of ATP synthesis are insufficient to support network function. Here, we assessed the extent to which neuromodulation or metabolic limitations arrest activity in the respiratory network of bullfrogs-a circuit that survives moderate periods of oxygen deprivation, presumably, by activating an inhibitory noradrenergic pathway. We confirmed that hypoxia and norepinephrine (NE) reduce network output, consistent with the view that hypoxia may cause the release of NE to inhibit activity. However, these responses differed qualitatively; hypoxia, but not NE, elicited a large motor burst and silenced the network. The stereotyped response to hypoxia persisted in the presence of both NE and an adrenergic receptor blocker that eliminates sensitivity to NE, indicating that noradrenergic signaling does not cause the arrest. Pharmacological inhibition of glycolysis and mitochondrial respiration recapitulated all features of hypoxia on network activity, implying that reduced ATP synthesis underlies the effects of hypoxia. Finally, activating modulatory mechanisms that dampen neuronal excitability when ATP levels fall, KATP channels and AMP-dependent protein kinase, did not resemble the hypoxic response. These results suggest that energy failure-rather than inhibitory modulation-silences the respiratory network during hypoxia and emphasize the need to account for metabolic limitations before concluding that modulators arrest activity as an adaptation for energy conservation in the nervous system.
Collapse
Affiliation(s)
- Sasha Adams
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Tanya Zubov
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Nikolaus Bueschke
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| | - Joseph M Santin
- Department of Biology, The University of North Carolina at Greensboro, Greensboro, North Carolina
| |
Collapse
|
9
|
Pospelov AS, Puskarjov M, Kaila K, Voipio J. Endogenous brain-sparing responses in brain pH and PO 2 in a rodent model of birth asphyxia. Acta Physiol (Oxf) 2020; 229:e13467. [PMID: 32174009 DOI: 10.1111/apha.13467] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
AIM To study brain-sparing physiological responses in a rodent model of birth asphyxia which reproduces the asphyxia-defining systemic hypoxia and hypercapnia. METHODS Steady or intermittent asphyxia was induced for 15-45 minutes in anaesthetized 6- and 11-days old rats and neonatal guinea pigs using gases containing 5% or 9% O2 plus 20% CO2 (in N2 ). Hypoxia and hypercapnia were induced with low O2 and high CO2 respectively. Oxygen partial pressure (PO2 ) and pH were measured with microsensors within the brain and subcutaneous ("body") tissue. Blood lactate was measured after asphyxia. RESULTS Brain and body PO2 fell to apparent zero with little recovery during 5% O2 asphyxia and 5% or 9% O2 hypoxia, and increased more than twofold during 20% CO2 hypercapnia. Unlike body PO2 , brain PO2 recovered rapidly to control after a transient fall (rat), or was slightly higher than control (guinea pig) during 9% O2 asphyxia. Asphyxia (5% O2 ) induced a respiratory acidosis paralleled by a progressive metabolic (lact)acidosis that was much smaller within than outside the brain. Hypoxia (5% O2 ) produced a brain-confined alkalosis. Hypercapnia outlasting asphyxia suppressed pH recovery and prolonged the post-asphyxia PO2 overshoot. All pH changes were accompanied by consistent shifts in the blood-brain barrier potential. CONCLUSION Regardless of brain maturation stage, hypercapnia can restore brain PO2 and protect the brain against metabolic acidosis despite compromised oxygen availability during asphyxia. This effect extends to the recovery phase if normocapnia is restored slowly, and it is absent during hypoxia, demonstrating that exposure to hypoxia does not mimic asphyxia.
Collapse
Affiliation(s)
- Alexey S. Pospelov
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| | - Martin Puskarjov
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| | - Kai Kaila
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
- Neuroscience Center (HiLIFE) University of Helsinki Helsinki Finland
| | - Juha Voipio
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences University of Helsinki Helsinki Finland
| |
Collapse
|
10
|
Timper K, Del Río-Martín A, Cremer AL, Bremser S, Alber J, Giavalisco P, Varela L, Heilinger C, Nolte H, Trifunovic A, Horvath TL, Kloppenburg P, Backes H, Brüning JC. GLP-1 Receptor Signaling in Astrocytes Regulates Fatty Acid Oxidation, Mitochondrial Integrity, and Function. Cell Metab 2020; 31:1189-1205.e13. [PMID: 32433922 PMCID: PMC7272126 DOI: 10.1016/j.cmet.2020.05.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 12/09/2019] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
Astrocytes represent central regulators of brain glucose metabolism and neuronal function. They have recently been shown to adapt their function in response to alterations in nutritional state through responding to the energy state-sensing hormones leptin and insulin. Here, we demonstrate that glucagon-like peptide (GLP)-1 inhibits glucose uptake and promotes β-oxidation in cultured astrocytes. Conversely, postnatal GLP-1 receptor (GLP-1R) deletion in glial fibrillary acidic protein (GFAP)-expressing astrocytes impairs astrocyte mitochondrial integrity and activates an integrated stress response with enhanced fibroblast growth factor (FGF)21 production and increased brain glucose uptake. Accordingly, central neutralization of FGF21 or astrocyte-specific FGF21 inactivation abrogates the improvements in glucose tolerance and learning in mice lacking GLP-1R expression in astrocytes. Collectively, these experiments reveal a role for astrocyte GLP-1R signaling in maintaining mitochondrial integrity, and lack of GLP-1R signaling mounts an adaptive stress response resulting in an improvement of systemic glucose homeostasis and memory formation.
Collapse
Affiliation(s)
- Katharina Timper
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Almudena Del Río-Martín
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Anna Lena Cremer
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany
| | - Stephan Bremser
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Institute for Zoology, Biocenter, University of Cologne, Zuelpicher Str. 47B, 50674 Cologne, Germany
| | - Jens Alber
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Patrick Giavalisco
- Max Planck Institute for Biology of Aging, Joseph-Stelzmann-Str. 9b, 50931 Cologne, Germany
| | - Luis Varela
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Christian Heilinger
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Hendrik Nolte
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Aleksandra Trifunovic
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Tamas L Horvath
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anatomy and Histology, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Institute for Zoology, Biocenter, University of Cologne, Zuelpicher Str. 47B, 50674 Cologne, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
11
|
Morris G, Maes M, Berk M, Carvalho AF, Puri BK. Nutritional ketosis as an intervention to relieve astrogliosis: Possible therapeutic applications in the treatment of neurodegenerative and neuroprogressive disorders. Eur Psychiatry 2020; 63:e8. [PMID: 32093791 PMCID: PMC8057392 DOI: 10.1192/j.eurpsy.2019.13] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Nutritional ketosis, induced via either the classical ketogenic diet or the use of emulsified medium-chain triglycerides, is an established treatment for pharmaceutical resistant epilepsy in children and more recently in adults. In addition, the use of oral ketogenic compounds, fractionated coconut oil, very low carbohydrate intake, or ketone monoester supplementation has been reported to be potentially helpful in mild cognitive impairment, Parkinson’s disease, schizophrenia, bipolar disorder, and autistic spectrum disorder. In these and other neurodegenerative and neuroprogressive disorders, there are detrimental effects of oxidative stress, mitochondrial dysfunction, and neuroinflammation on neuronal function. However, they also adversely impact on neurone–glia interactions, disrupting the role of microglia and astrocytes in central nervous system (CNS) homeostasis. Astrocytes are the main site of CNS fatty acid oxidation; the resulting ketone bodies constitute an important source of oxidative fuel for neurones in an environment of glucose restriction. Importantly, the lactate shuttle between astrocytes and neurones is dependent on glycogenolysis and glycolysis, resulting from the fact that the astrocytic filopodia responsible for lactate release are too narrow to accommodate mitochondria. The entry into the CNS of ketone bodies and fatty acids, as a result of nutritional ketosis, has effects on the astrocytic glutamate–glutamine cycle, glutamate synthase activity, and on the function of vesicular glutamate transporters, EAAT, Na+, K+-ATPase, Kir4.1, aquaporin-4, Cx34 and KATP channels, as well as on astrogliosis. These mechanisms are detailed and it is suggested that they would tend to mitigate the changes seen in many neurodegenerative and neuroprogressive disorders. Hence, it is hypothesized that nutritional ketosis may have therapeutic applications in such disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | | |
Collapse
|
12
|
Santin JM. Motor inactivity in hibernating frogs: Linking plasticity that stabilizes neuronal function to behavior in the natural environment. Dev Neurobiol 2019; 79:880-891. [DOI: 10.1002/dneu.22721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/07/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Joseph M. Santin
- Department of BiologyUniversity of North Carolina at Greensboro Greensboro North Carolina
| |
Collapse
|
13
|
Cellular and Synaptic Dysfunctions in Parkinson's Disease: Stepping out of the Striatum. Cells 2019; 8:cells8091005. [PMID: 31470672 PMCID: PMC6769933 DOI: 10.3390/cells8091005] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/30/2022] Open
Abstract
The basal ganglia (BG) are a collection of interconnected subcortical nuclei that participate in a great variety of functions, ranging from motor programming and execution to procedural learning, cognition, and emotions. This network is also the region primarily affected by the degeneration of midbrain dopaminergic neurons localized in the substantia nigra pars compacta (SNc). This degeneration causes cellular and synaptic dysfunctions in the BG network, which are responsible for the appearance of the motor symptoms of Parkinson’s disease. Dopamine (DA) modulation and the consequences of its loss on the striatal microcircuit have been extensively studied, and because of the discrete nature of DA innervation of other BG nuclei, its action outside the striatum has been considered negligible. However, there is a growing body of evidence supporting functional extrastriatal DA modulation of both cellular excitability and synaptic transmission. In this review, the functional relevance of DA modulation outside the striatum in both normal and pathological conditions will be discussed.
Collapse
|
14
|
Díaz-García CM, Lahmann C, Martínez-François JR, Li B, Koveal D, Nathwani N, Rahman M, Keller JP, Marvin JS, Looger LL, Yellen G. Quantitative in vivo imaging of neuronal glucose concentrations with a genetically encoded fluorescence lifetime sensor. J Neurosci Res 2019; 97:946-960. [PMID: 31106909 PMCID: PMC6565483 DOI: 10.1002/jnr.24433] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 01/07/2023]
Abstract
Glucose is an essential source of energy for the brain. Recently, the development of genetically encoded fluorescent biosensors has allowed real time visualization of glucose dynamics from individual neurons and astrocytes. A major difficulty for this approach, even for ratiometric sensors, is the lack of a practical method to convert such measurements into actual concentrations in ex vivo brain tissue or in vivo. Fluorescence lifetime imaging provides a strategy to overcome this. In a previous study, we reported the lifetime glucose sensor iGlucoSnFR-TS (then called SweetieTS) for monitoring changes in neuronal glucose levels in response to stimulation. This genetically encoded sensor was generated by combining the Thermus thermophilus glucose-binding protein with a circularly permuted variant of the monomeric fluorescent protein T-Sapphire. Here, we provide more details on iGlucoSnFR-TS design and characterization, as well as pH and temperature sensitivities. For accurate estimation of glucose concentrations, the sensor must be calibrated at the same temperature as the experiments. We find that when the extracellular glucose concentration is in the range 2-10 mM, the intracellular glucose concentration in hippocampal neurons from acute brain slices is ~20% of the nominal external glucose concentration (~0.4-2 mM). We also measured the cytosolic neuronal glucose concentration in vivo, finding a range of ~0.7-2.5 mM in cortical neurons from awake mice.
Collapse
Affiliation(s)
| | - Carolina Lahmann
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | | - Binsen Li
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Dorothy Koveal
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Nidhi Nathwani
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Mahia Rahman
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Jacob P. Keller
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jonathan S. Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Loren L. Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Koenig JB, Cantu D, Low C, Sommer M, Noubary F, Croker D, Whalen M, Kong D, Dulla CG. Glycolytic inhibitor 2-deoxyglucose prevents cortical hyperexcitability after traumatic brain injury. JCI Insight 2019; 5:126506. [PMID: 31038473 DOI: 10.1172/jci.insight.126506] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) causes cortical dysfunction and can lead to post-traumatic epilepsy. Multiple studies demonstrate that GABAergic inhibitory network function is compromised following TBI, which may contribute to hyperexcitability and motor, behavioral, and cognitive deficits. Preserving the function of GABAergic interneurons, therefore, is a rational therapeutic strategy to preserve cortical function after TBI and prevent long-term clinical complications. Here, we explored an approach based on the ketogenic diet, a neuroprotective and anticonvulsant dietary therapy which results in reduced glycolysis and increased ketosis. Utilizing a pharmacologic inhibitor of glycolysis (2-deoxyglucose, or 2-DG), we found that acute in vitro application of 2-DG decreased the excitability of excitatory neurons, but not inhibitory interneurons, in cortical slices from naïve mice. Employing the controlled cortical impact (CCI) model of TBI in mice, we found that in vitro 2-DG treatment rapidly attenuated epileptiform activity seen in acute cortical slices 3 to 5 weeks after TBI. One week of in vivo 2-DG treatment immediately after TBI prevented the development of epileptiform activity, restored excitatory and inhibitory synaptic activity, and attenuated the loss of parvalbumin-expressing inhibitory interneurons. In summary, 2-DG may have therapeutic potential to restore network function following TBI.
Collapse
Affiliation(s)
- Jenny B Koenig
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA.,Neuroscience Program, Tufts University Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts, USA
| | - David Cantu
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Cho Low
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA.,Cellular, Molecular, and Developmental Biology Program, Tufts University Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts, USA
| | - Mary Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Farzad Noubary
- Department of Health Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Danielle Croker
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Michael Whalen
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dong Kong
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Photopharmacology and opto-chemogenetics of TRPC channels-some therapeutic visions. Pharmacol Ther 2019; 200:13-26. [PMID: 30974125 DOI: 10.1016/j.pharmthera.2019.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/27/2019] [Indexed: 12/28/2022]
Abstract
Non-selective cation conductances formed by transient receptor potential canonical (TRPC) proteins govern the function and fate of a wide range of human cell types. In the past decade, evidence has accumulated for a pivotal role of these channels in human diseases, raising substantial interest in their therapeutic targeting. As yet, an appreciable number of small molecules for block and modulation of recombinant TRPC conductances have been identified. However, groundbreaking progress in TRPC pharmacology towards therapeutic applications is lagging behind due to incomplete understanding of their molecular pharmacology and their exact role in disease. A major breakthrough that is expected to overcome these hurdles is the recent success in obtaining high-resolution structure information on TRPC channel complexes and the advent of TRP photopharmacology and optogenetics. Here, we summarize current concepts of enhancing the precision of therapeutic interference with TRPC signaling and TRPC-mediated pathological processes.
Collapse
|
17
|
Fogle KJ, Smith AR, Satterfield SL, Gutierrez AC, Hertzler JI, McCardell CS, Shon JH, Barile ZJ, Novak MO, Palladino MJ. Ketogenic and anaplerotic dietary modifications ameliorate seizure activity in Drosophila models of mitochondrial encephalomyopathy and glycolytic enzymopathy. Mol Genet Metab 2019; 126:439-447. [PMID: 30683556 PMCID: PMC6536302 DOI: 10.1016/j.ymgme.2019.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/13/2022]
Abstract
Seizures are a feature not only of the many forms of epilepsy, but also of global metabolic diseases such as mitochondrial encephalomyopathy (ME) and glycolytic enzymopathy (GE). Modern anti-epileptic drugs (AEDs) are successful in many cases, but some patients are refractory to existing AEDs, which has led to a surge in interest in clinically managed dietary therapy such as the ketogenic diet (KD). This high-fat, low-carbohydrate diet causes a cellular switch from glycolysis to fatty acid oxidation and ketone body generation, with a wide array of downstream effects at the genetic, protein, and metabolite level that may mediate seizure protection. We have recently shown that a Drosophila model of human ME (ATP61) responds robustly to the KD; here, we have investigated the mechanistic importance of the major metabolic consequences of the KD in the context of this bioenergetics disease: ketogenesis, reduction of glycolysis, and anaplerosis. We have found that reduction of glycolysis does not confer seizure protection, but that dietary supplementation with ketone bodies or the anaplerotic lipid triheptanoin, which directly replenishes the citric acid cycle, can mimic the success of the ketogenic diet even in the presence of standard carbohydrate levels. We have also shown that the proper functioning of the citric acid cycle is crucial to the success of the KD in the context of ME. Furthermore, our data reveal that multiple seizure models, in addition to ATP61, are treatable with the ketogenic diet. Importantly, one of these mutants is TPIsugarkill, which models human glycolytic enzymopathy, an incurable metabolic disorder with severe neurological consequences. Overall, these studies reveal widespread success of the KD in Drosophila, further cementing its status as an excellent model for studies of KD treatment and mechanism, and reveal key insights into the therapeutic potential of dietary therapy against neuronal hyperexcitability in epilepsy and metabolic disease.
Collapse
Affiliation(s)
- Keri J Fogle
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| | - Amber R Smith
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sidney L Satterfield
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Alejandra C Gutierrez
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - J Ian Hertzler
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Caleb S McCardell
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Joy H Shon
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Zackery J Barile
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Molly O Novak
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Michael J Palladino
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
18
|
Tiapko O, Shrestha N, Lindinger S, Guedes de la Cruz G, Graziani A, Klec C, Butorac C, Graier WF, Kubista H, Freichel M, Birnbaumer L, Romanin C, Glasnov T, Groschner K. Lipid-independent control of endothelial and neuronal TRPC3 channels by light. Chem Sci 2019; 10:2837-2842. [PMID: 30997005 PMCID: PMC6427946 DOI: 10.1039/c8sc05536j] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Lipid-gated TRPC channels are highly expressed in cardiovascular and neuronal tissues. Exerting precise pharmacological control over their activity in native cells is expected to serve as a basis for the development of novel therapies. Here we report on a new photopharmacological tool that enables manipulation of TRPC3 channels by light, in a manner independent of lipid metabolism and with higher temporal precision than lipid photopharmacology. Using the azobenzene photoswitch moiety, we modified GSK1702934A to generate light-controlled TRPC agonists. We obtained one light-sensitive molecule (OptoBI-1) that allows us to exert efficient, light-mediated control over TRPC3 activity and the associated cellular Ca2+ signaling. OptoBI-1 enabled high-precision, temporal control of TRPC3-linked cell functions such as neuronal firing and endothelial Ca2+ transients. With these findings, we introduce a novel photopharmacological strategy to control native TRPC conductances.
Collapse
Affiliation(s)
- Oleksandra Tiapko
- Gottfried Schatz Research Center - Biophysics , Medical University of Graz , Neue Stiftingtalstraße 6/D/04 , 8010 Graz , Austria .
| | - Niroj Shrestha
- Gottfried Schatz Research Center - Biophysics , Medical University of Graz , Neue Stiftingtalstraße 6/D/04 , 8010 Graz , Austria .
| | - Sonja Lindinger
- Institute of Biophysics , University of Linz , Gruberstrasse 40/1 , 4020 Linz , Austria
| | | | - Annarita Graziani
- Gottfried Schatz Research Center - Biophysics , Medical University of Graz , Neue Stiftingtalstraße 6/D/04 , 8010 Graz , Austria .
| | - Christiane Klec
- Gottfried Schatz Research Center - Molecular Biology and Biochemistry , Medical University of Graz , Neue Stiftingtalstraße 6/6 , 8010 Graz , Austria
| | - Carmen Butorac
- Institute of Biophysics , University of Linz , Gruberstrasse 40/1 , 4020 Linz , Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center - Molecular Biology and Biochemistry , Medical University of Graz , Neue Stiftingtalstraße 6/6 , 8010 Graz , Austria
| | - Helmut Kubista
- Institute of Pharmacology , Medical University of Vienna , Währinger Straße 13A , 1090 Vienna , Austria
| | - Marc Freichel
- Pharmakologisches Institut , Universität Heidelberg , Im Neuenheimer Feld 366 , D-69120 Heidelberg , Germany
| | - Lutz Birnbaumer
- Neurobiology Laboratory , National Institute of Environmental Health Sciences , Research Triangle Park , North Carolina 27709 , USA
- Institute of Biomedical Research (BIOMED) , Catholique University of Argentina , Buenos Aires C1107AZZ , Argentina
| | - Christoph Romanin
- Institute of Biophysics , University of Linz , Gruberstrasse 40/1 , 4020 Linz , Austria
| | - Toma Glasnov
- Institute of Chemistry , University of Graz , Heinrichstraße 28/I , 8010 Graz , Austria
| | - Klaus Groschner
- Gottfried Schatz Research Center - Biophysics , Medical University of Graz , Neue Stiftingtalstraße 6/D/04 , 8010 Graz , Austria .
| |
Collapse
|
19
|
Tiroshi L, Goldberg JA. Population dynamics and entrainment of basal ganglia pacemakers are shaped by their dendritic arbors. PLoS Comput Biol 2019; 15:e1006782. [PMID: 30730886 PMCID: PMC6382172 DOI: 10.1371/journal.pcbi.1006782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 02/20/2019] [Accepted: 01/10/2019] [Indexed: 11/30/2022] Open
Abstract
The theory of phase oscillators is an essential tool for understanding population dynamics of pacemaking neurons. GABAergic pacemakers in the substantia nigra pars reticulata (SNr), a main basal ganglia (BG) output nucleus, receive inputs from the direct and indirect pathways at distal and proximal regions of their dendritic arbors, respectively. We combine theory, optogenetic stimulation and electrophysiological experiments in acute brain slices to ask how dendritic properties impact the propensity of the various inputs, arriving at different locations along the dendrite, to recruit or entrain SNr pacemakers. By combining cable theory with sinusoidally-modulated optogenetic activation of either proximal somatodendritic regions or the entire somatodendritic arbor of SNr neurons, we construct an analytical model that accurately fits the empirically measured somatic current response to inputs arising from illuminating the soma and various portions of the dendritic field. We show that the extent of the dendritic tree that is illuminated generates measurable and systematic differences in the pacemaker’s phase response curve (PRC), causing a shift in its peak. Finally, we show that the divergent PRCs correctly predict differences in two major features of the collective dynamics of SNr neurons: the fidelity of population responses to sudden step-like changes in inputs; and the phase latency at which SNr neurons are entrained by rhythmic stimulation, which can occur in the BG under both physiological and pathophysiological conditions. Our novel method generates measurable and physiologically meaningful spatial effects, and provides the first empirical demonstration of how the collective responses of SNr pacemakers are determined by the transmission properties of their dendrites. SNr dendrites may serve to delay distal striatal inputs so that they impinge on the spike initiation zone simultaneously with pallidal and subthalamic inputs in order to guarantee a fair competition between the influence of the monosynaptic direct- and polysynaptic indirect pathways. The substantia nigra pars reticulata (SNr) is a main output nucleus of the basal ganglia (BG), where inputs from the competing direct and indirect pathways converge onto the same neurons. Interestingly, these inputs are differentially distributed with direct and indirect pathway projections arriving at distal and proximal regions of the dendritic arbor, respectively. We employ a novel method combining theory with electrophysiological experiments and optogenetics to study the distinct effects of inputs arriving at different locations along the dendrite. Our approach represents a useful compromise between complexity and reduction in modelling. Our work addresses the question of high fidelity encoding of inputs by networks of neurons in the new context of pacemaking neurons, which are driven to fire by their intrinsic dynamics rather than by a network state. We provide the first empirical demonstration that dendritic delays can introduce latencies in the responses of a population of neurons that are commensurate with synaptic delays, suggesting a new role for SNr dendrites with implications for BG function.
Collapse
Affiliation(s)
- Lior Tiroshi
- Department of Medical Neurobiology, Institute of Medical Research Israel–Canada, The Faculty of Medicine, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joshua A. Goldberg
- Department of Medical Neurobiology, Institute of Medical Research Israel–Canada, The Faculty of Medicine, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
20
|
Abstract
Glucose is the long-established, obligatory fuel for brain that fulfills many critical functions, including ATP production, oxidative stress management, and synthesis of neurotransmitters, neuromodulators, and structural components. Neuronal glucose oxidation exceeds that in astrocytes, but both rates increase in direct proportion to excitatory neurotransmission; signaling and metabolism are closely coupled at the local level. Exact details of neuron-astrocyte glutamate-glutamine cycling remain to be established, and the specific roles of glucose and lactate in the cellular energetics of these processes are debated. Glycolysis is preferentially upregulated during brain activation even though oxygen availability is sufficient (aerobic glycolysis). Three major pathways, glycolysis, pentose phosphate shunt, and glycogen turnover, contribute to utilization of glucose in excess of oxygen, and adrenergic regulation of aerobic glycolysis draws attention to astrocytic metabolism, particularly glycogen turnover, which has a high impact on the oxygen-carbohydrate mismatch. Aerobic glycolysis is proposed to be predominant in young children and specific brain regions, but re-evaluation of data is necessary. Shuttling of glucose- and glycogen-derived lactate from astrocytes to neurons during activation, neurotransmission, and memory consolidation are controversial topics for which alternative mechanisms are proposed. Nutritional therapy and vagus nerve stimulation are translational bridges from metabolism to clinical treatment of diverse brain disorders.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas ; and Department of Cell Biology and Physiology, University of New Mexico , Albuquerque, New Mexico
| |
Collapse
|
21
|
Koenig JB, Dulla CG. Dysregulated Glucose Metabolism as a Therapeutic Target to Reduce Post-traumatic Epilepsy. Front Cell Neurosci 2018; 12:350. [PMID: 30459556 PMCID: PMC6232824 DOI: 10.3389/fncel.2018.00350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a significant cause of disability worldwide and can lead to post-traumatic epilepsy. Multiple molecular, cellular, and network pathologies occur following injury which may contribute to epileptogenesis. Efforts to identify mechanisms of disease progression and biomarkers which predict clinical outcomes have focused heavily on metabolic changes. Advances in imaging approaches, combined with well-established biochemical methodologies, have revealed a complex landscape of metabolic changes that occur acutely after TBI and then evolve in the days to weeks after. Based on this rich clinical and preclinical data, combined with the success of metabolic therapies like the ketogenic diet in treating epilepsy, interest has grown in determining whether manipulating metabolic activity following TBI may have therapeutic value to prevent post-traumatic epileptogenesis. Here, we focus on changes in glucose utilization and glycolytic activity in the brain following TBI and during seizures. We review relevant literature and outline potential paths forward to utilize glycolytic inhibitors as a disease-modifying therapy for post-traumatic epilepsy.
Collapse
Affiliation(s)
- Jenny B Koenig
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
22
|
Cáceres-Chávez VA, Hernández-Martínez R, Pérez-Ortega J, Herrera-Valdez MA, Aceves JJ, Galarraga E, Bargas J. Acute dopamine receptor blockade in substantia nigra pars reticulata: a possible model for drug-induced Parkinsonism. J Neurophysiol 2018; 120:2922-2938. [PMID: 30256736 DOI: 10.1152/jn.00579.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dopamine (DA) depletion modifies the firing pattern of neurons in the substantia nigra pars reticulata (SNr), shifting their mostly tonic firing toward irregularity and bursting, traits of pathological firing underlying rigidity and postural instability in Parkinson's disease (PD) patients and animal models of Parkinsonism (PS). Drug-induced Parkinsonism (DIP) represents 20-40% of clinical cases of PS, becoming a problem for differential diagnosis, and is still not well studied with physiological tools. It may co-occur with tardive dyskinesia. Here we use in vitro slice preparations including the SNr to observe drug-induced pathological firing by using drugs that most likely produce it, DA-receptor antagonists (SCH23390 plus sulpiride), to compare with firing patterns found in DA-depleted tissue. The hypothesis is that SNr firing would be similar under both conditions, a prerequisite to the proposal of a similar preparation to test other DIP-producing drugs. Firing was analyzed with three complementary metrics, showing similarities between DA depletion and acute DA-receptor blockade. Moreover, blockade of either nonselective cationic channels or Cav3 T-type calcium channels hyperpolarized the membrane and abolished bursting and irregular firing, silencing SNr neurons in both conditions. Therefore, currents generating firing in control conditions are in part responsible for pathological firing. Haloperidol, a DIP-producing drug, reproduced DA-receptor antagonist firing modifications. Since acute DA-receptor blockade induces SNr neuron firing similar to that found in the 6-hydroxydopamine model of PS, output basal ganglia neurons may play a role in generating DIP. Therefore, this study opens the way to test other DIP-producing drugs. NEW & NOTEWORTHY Dopamine (DA) depletion enhances substantia nigra pars reticulata (SNr) neuron bursting and irregular firing, hallmarks of Parkinsonism. Several drugs, including antipsychotics, antidepressants, and calcium channel antagonists, among others, produce drug-induced Parkinsonism. Here we show the first comparison between SNr neuron firing after DA depletion vs. firing found after acute blockade of DA receptors. It was found that firing in both conditions is similar, implying that pathological SNr neuron firing is also a physiological correlate of drug-induced Parkinsonism.
Collapse
Affiliation(s)
| | - Ricardo Hernández-Martínez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City, México
| | - Jesús Pérez-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México , Querétaro, México
| | - Marco Arieli Herrera-Valdez
- Departamento de Matemáticas, Facultad de Ciencias, Universidad Nacional Autónoma de México , México City, México
| | - Jose J Aceves
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , Mexico City, México
| | - Elvira Galarraga
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City, México
| | - José Bargas
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City, México
| |
Collapse
|
23
|
Rajendran M, Claywell B, Haynes EP, Scales U, Henning CK, Tantama M. Imaging pH Dynamics Simultaneously in Two Cellular Compartments Using a Ratiometric pH-Sensitive Mutant of mCherry. ACS OMEGA 2018; 3:9476-9486. [PMID: 30197999 PMCID: PMC6120727 DOI: 10.1021/acsomega.8b00655] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/06/2018] [Indexed: 05/09/2023]
Abstract
The regulation of pH is essential for proper organelle function, and organelle-specific changes in pH often reflect the dynamics of physiological signaling and metabolism. For example, mitochondrial energy production depends on the proton gradient maintained between the alkaline mitochondrial matrix and neutral cytosol. However, we still lack a quantitative understanding of how pH dynamics are coupled between compartments and how pH gradients are regulated at organelle boundaries. Genetically encoded pH sensors are well suited to address this problem because they can be targeted to specific subcellular locations and they facilitate live, single-cell analysis. However, most of these pH sensors are derivatives of green and yellow fluorescent proteins that are not spectrally compatible for dual-compartment imaging. Therefore, there is a need for ratiometric red fluorescent protein pH sensors that enable quantitative multicolor imaging of spatially resolved pH dynamics. In this work, we demonstrate that the I158E/Q160A mutant of the red fluorescent protein mCherry is an effective ratiometric pH sensor. It has a pKa of 7.3 and a greater than 3-fold change in ratio signal. To demonstrate its utility in cells, we measured activity and metabolism-dependent pH dynamics in cultured primary neurons and neuroblastoma cells. Furthermore, we were able to image pH changes simultaneously in the cytosol and mitochondria by using the mCherryEA mutant together with the green fluorescent pH sensor, ratiometric-pHluorin. Our results demonstrate the feasibility of studying interorganelle pH dynamics in live cells over time and the broad applicability of these sensors in studying the role of pH regulation in metabolism and signaling.
Collapse
Affiliation(s)
- Megha Rajendran
- Department
of Chemistry, Institute for Integrative Neuroscience, and Institute of Inflammation, Immunology,
and Infectious Disease, Purdue University, 560 Oval Drive,
P.O. Box 68, West Lafayette, Indiana 47907, United States
| | - Benjamin Claywell
- Department
of Chemistry, Institute for Integrative Neuroscience, and Institute of Inflammation, Immunology,
and Infectious Disease, Purdue University, 560 Oval Drive,
P.O. Box 68, West Lafayette, Indiana 47907, United States
| | - Emily P. Haynes
- Department
of Chemistry, Institute for Integrative Neuroscience, and Institute of Inflammation, Immunology,
and Infectious Disease, Purdue University, 560 Oval Drive,
P.O. Box 68, West Lafayette, Indiana 47907, United States
| | - Umi Scales
- Department
of Chemistry, Institute for Integrative Neuroscience, and Institute of Inflammation, Immunology,
and Infectious Disease, Purdue University, 560 Oval Drive,
P.O. Box 68, West Lafayette, Indiana 47907, United States
| | - Chace K. Henning
- Department
of Chemistry, Institute for Integrative Neuroscience, and Institute of Inflammation, Immunology,
and Infectious Disease, Purdue University, 560 Oval Drive,
P.O. Box 68, West Lafayette, Indiana 47907, United States
| | - Mathew Tantama
- Department
of Chemistry, Institute for Integrative Neuroscience, and Institute of Inflammation, Immunology,
and Infectious Disease, Purdue University, 560 Oval Drive,
P.O. Box 68, West Lafayette, Indiana 47907, United States
- E-mail: . Phone: 765-494-5312
| |
Collapse
|
24
|
Tiapko O, Groschner K. TRPC3 as a Target of Novel Therapeutic Interventions. Cells 2018; 7:cells7070083. [PMID: 30037143 PMCID: PMC6071100 DOI: 10.3390/cells7070083] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 01/25/2023] Open
Abstract
TRPC3 is one of the classical members of the mammalian transient receptor potential (TRP) superfamily of ion channels. TRPC3 is a molecule with intriguing sensory features including the direct recognition of and activation by diacylglycerols (DAG). Although TRPC3 channels are ubiquitously expressed, they appear to control functions of the cardiovascular system and the brain in a highly specific manner. Moreover, a role of TRPC3 in immunity, cancer, and tissue remodeling has been proposed, generating much interest in TRPC3 as a target for pharmacological intervention. Advances in the understanding of molecular architecture and structure-function relations of TRPC3 have been the foundations for novel therapeutic approaches, such as photopharmacology and optochemical genetics of TRPC3. This review provides an account of advances in therapeutic targeting of TRPC3 channels.
Collapse
Affiliation(s)
- Oleksandra Tiapko
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria.
| | - Klaus Groschner
- Gottfried-Schatz-Research-Center-Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/D04, 8010 Graz, Austria.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW High-fat, low-carbohydrate ketogenic diets have been used for almost a century for the treatment of epilepsy. Used traditionally for the treatment of refractory pediatric epilepsies, in recent years the use of ketogenic diets has experienced a revival to include the treatment of adulthood epilepsies as well as conditions ranging from autism to chronic pain and cancer. Despite the ability of ketogenic diet therapy to suppress seizures refractory to antiepileptic drugs and reports of lasting seizure freedom, the underlying mechanisms are poorly understood. This review explores new insights into mechanisms mobilized by ketogenic diet therapies. RECENT FINDINGS Ketogenic diets act through a combination of mechanisms, which are linked to the effects of ketones and glucose restriction, and to interactions with receptors, channels, and metabolic enzymes. Decanoic acid, a component of medium-chain triclycerides, contributes to seizure control through direct α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor inhibition, whereas drugs targeting lactate dehydrogenase reduce seizures through inhibition of a metabolic pathway. Ketogenic diet therapy also affects DNA methylation, a novel epigenetic mechanism of the diet. SUMMARY Ketogenic diet therapy combines several beneficial mechanisms that provide broad benefits for the treatment of epilepsy with the potential to not only suppress seizures but also to modify the course of the epilepsy.
Collapse
|
26
|
Szeto V, Chen NH, Sun HS, Feng ZP. The role of K ATP channels in cerebral ischemic stroke and diabetes. Acta Pharmacol Sin 2018; 39:683-694. [PMID: 29671418 PMCID: PMC5943906 DOI: 10.1038/aps.2018.10] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/19/2018] [Indexed: 12/18/2022]
Abstract
ATP-sensitive potassium (KATP) channels are ubiquitously expressed on the plasma membrane of cells in multiple organs, including the heart, pancreas and brain. KATP channels play important roles in controlling and regulating cellular functions in response to metabolic state, which are inhibited by ATP and activated by Mg-ADP, allowing the cell to couple cellular metabolic state (ATP/ADP ratio) to electrical activity of the cell membrane. KATP channels mediate insulin secretion in pancreatic islet beta cells, and controlling vascular tone. Under pathophysiological conditions, KATP channels play cytoprotective role in cardiac myocytes and neurons during ischemia and/or hypoxia. KATP channel is a hetero-octameric complex, consisting of four pore-forming Kir6.x and four regulatory sulfonylurea receptor SURx subunits. These subunits are differentially expressed in various cell types, thus determining the sensitivity of the cells to specific channel modifiers. Sulfonylurea class of antidiabetic drugs blocks KATP channels, which are neuroprotective in stroke, can be one of the high stoke risk factors for diabetic patients. In this review, we discussed the potential effects of KATP channel blockers when used under pathological conditions related to diabetics and cerebral ischemic stroke.
Collapse
Affiliation(s)
- Vivian Szeto
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Nai-hong Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hong-shuo Sun
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
- Surgery
- Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Zhong-ping Feng
- Departments of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
27
|
Barry D, Ellul S, Watters L, Lee D, Haluska R, White R. The ketogenic diet in disease and development. Int J Dev Neurosci 2018; 68:53-58. [DOI: 10.1016/j.ijdevneu.2018.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/31/2018] [Accepted: 04/15/2018] [Indexed: 02/08/2023] Open
Affiliation(s)
- Denis Barry
- Department of Anatomy Trinity Biomedical Sciences InstituteTrinity College DublinDublin, 2Ireland
| | - Sarah Ellul
- Department of Anatomy Trinity Biomedical Sciences InstituteTrinity College DublinDublin, 2Ireland
| | - Lindsey Watters
- Department of Anatomy Trinity Biomedical Sciences InstituteTrinity College DublinDublin, 2Ireland
| | - David Lee
- Department of Anatomy Trinity Biomedical Sciences InstituteTrinity College DublinDublin, 2Ireland
| | - Robert Haluska
- Department of BiologyWestfield State University577 Western AvenueWestfieldMA01085United States
| | - Robin White
- Department of BiologyWestfield State University577 Western AvenueWestfieldMA01085United States
| |
Collapse
|
28
|
Salgado-Puga K, Rodríguez-Colorado J, Prado-Alcalá RA, Peña-Ortega F. Subclinical Doses of ATP-Sensitive Potassium Channel Modulators Prevent Alterations in Memory and Synaptic Plasticity Induced by Amyloid-β. J Alzheimers Dis 2018; 57:205-226. [PMID: 28222502 DOI: 10.3233/jad-160543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In addition to coupling cell metabolism and excitability, ATP-sensitive potassium channels (KATP) are involved in neural function and plasticity. Moreover, alterations in KATP activity and expression have been observed in Alzheimer's disease (AD) and during amyloid-β (Aβ)-induced pathology. Thus, we tested whether KATP modulators can influence Aβ-induced deleterious effects on memory, hippocampal network function, and plasticity. We found that treating animals with subclinical doses (those that did not change glycemia) of a KATP blocker (Tolbutamide) or a KATP opener (Diazoxide) differentially restrained Aβ-induced memory deficit, hippocampal network activity inhibition, and long-term synaptic plasticity unbalance (i.e., inhibition of LTP and promotion of LTD). We found that the protective effect of Tolbutamide against Aβ-induced memory deficit was strong and correlated with the reestablishment of synaptic plasticity balance, whereas Diazoxide treatment produced a mild protection against Aβ-induced memory deficit, which was not related to a complete reestablishment of synaptic plasticity balance. Interestingly, treatment with both KATP modulators renders the hippocampus resistant to Aβ-induced inhibition of hippocampal network activity. These findings indicate that KATP are involved in Aβ-induced pathology and they heighten the potential role of KATP modulation as a plausible therapeutic strategy against AD.
Collapse
Affiliation(s)
- Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Javier Rodríguez-Colorado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| |
Collapse
|
29
|
Martínez-François JR, Fernández-Agüera MC, Nathwani N, Lahmann C, Burnham VL, Danial NN, Yellen G. BAD and K ATP channels regulate neuron excitability and epileptiform activity. eLife 2018; 7:32721. [PMID: 29368690 PMCID: PMC5785210 DOI: 10.7554/elife.32721] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/12/2018] [Indexed: 12/17/2022] Open
Abstract
Brain metabolism can profoundly influence neuronal excitability. Mice with genetic deletion or alteration of Bad (BCL-2 agonist of cell death) exhibit altered brain-cell fuel metabolism, accompanied by resistance to acutely induced epileptic seizures; this seizure protection is mediated by ATP-sensitive potassium (KATP) channels. Here we investigated the effect of BAD manipulation on KATP channel activity and excitability in acute brain slices. We found that BAD’s influence on neuronal KATP channels was cell-autonomous and directly affected dentate granule neuron (DGN) excitability. To investigate the role of neuronal KATP channels in the anticonvulsant effects of BAD, we imaged calcium during picrotoxin-induced epileptiform activity in entorhinal-hippocampal slices. BAD knockout reduced epileptiform activity, and this effect was lost upon knockout or pharmacological inhibition of KATP channels. Targeted BAD knockout in DGNs alone was sufficient for the antiseizure effect in slices, consistent with a ‘dentate gate’ function that is reinforced by increased KATP channel activity.
Collapse
Affiliation(s)
| | | | - Nidhi Nathwani
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Carolina Lahmann
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Veronica L Burnham
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Nika N Danial
- Department of Neurobiology, Harvard Medical School, Boston, United States.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, United States
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
30
|
Castro JP, Wardelmann K, Grune T, Kleinridders A. Mitochondrial Chaperones in the Brain: Safeguarding Brain Health and Metabolism? Front Endocrinol (Lausanne) 2018; 9:196. [PMID: 29755410 PMCID: PMC5932182 DOI: 10.3389/fendo.2018.00196] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/10/2018] [Indexed: 12/31/2022] Open
Abstract
The brain orchestrates organ function and regulates whole body metabolism by the concerted action of neurons and glia cells in the central nervous system. To do so, the brain has tremendously high energy consumption and relies mainly on glucose utilization and mitochondrial function in order to exert its function. As a consequence of high rate metabolism, mitochondria in the brain accumulate errors over time, such as mitochondrial DNA (mtDNA) mutations, reactive oxygen species, and misfolded and aggregated proteins. Thus, mitochondria need to employ specific mechanisms to avoid or ameliorate the rise of damaged proteins that contribute to aberrant mitochondrial function and oxidative stress. To maintain mitochondria homeostasis (mitostasis), cells evolved molecular chaperones that shuttle, refold, or in coordination with proteolytic systems, help to maintain a low steady-state level of misfolded/aggregated proteins. Their importance is exemplified by the occurrence of various brain diseases which exhibit reduced action of chaperones. Chaperone loss (expression and/or function) has been observed during aging, metabolic diseases such as type 2 diabetes and in neurodegenerative diseases such as Alzheimer's (AD), Parkinson's (PD) or even Huntington's (HD) diseases, where the accumulation of damage proteins is evidenced. Within this perspective, we propose that proper brain function is maintained by the joint action of mitochondrial chaperones to ensure and maintain mitostasis contributing to brain health, and that upon failure, alter brain function which can cause metabolic diseases.
Collapse
Affiliation(s)
- José Pedro Castro
- Department of Molecular Toxicology, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- *Correspondence: José Pedro Castro, ; André Kleinridders,
| | - Kristina Wardelmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Central Regulation of Metabolism, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - André Kleinridders
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Central Regulation of Metabolism, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Germany
- *Correspondence: José Pedro Castro, ; André Kleinridders,
| |
Collapse
|
31
|
Abstract
Various mechanisms in the mammalian body provide resilience against food deprivation and dietary stress. The ketone body β-hydroxybutyrate (BHB) is synthesized in the liver from fatty acids and represents an essential carrier of energy from the liver to peripheral tissues when the supply of glucose is too low for the body's energetic needs, such as during periods of prolonged exercise, starvation, or absence of dietary carbohydrates. In addition to its activity as an energetic metabolite, BHB is increasingly understood to have cellular signaling functions. These signaling functions of BHB broadly link the outside environment to epigenetic gene regulation and cellular function, and their actions may be relevant to a variety of human diseases as well as human aging.
Collapse
Affiliation(s)
- John C Newman
- Buck Institute for Research on Aging, Novato, California 94945; ,
- Gladstone Institutes, San Francisco, California 94158
- Division of Geriatrics, University of California, San Francisco, California 94143
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, California 94945; ,
- Gladstone Institutes, San Francisco, California 94158
- Division of Geriatrics, University of California, San Francisco, California 94143
| |
Collapse
|
32
|
Bergström U, Lindfors C, Svedberg M, Johansen JE, Häggkvist J, Schalling M, Wibom R, Katz A, Nilsson IAK. Reduced metabolism in the hypothalamus of the anorectic anx/anx mouse. J Endocrinol 2017; 233:15-24. [PMID: 28130409 DOI: 10.1530/joe-16-0383] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/27/2017] [Indexed: 12/13/2022]
Abstract
The anorectic anx/anx mouse exhibits a mitochondrial complex I dysfunction that is related to aberrant expression of hypothalamic neuropeptides and transmitters regulating food intake. Hypothalamic activity, i.e. neuronal firing and transmitter release, is dependent on glucose utilization and energy metabolism. To better understand the role of hypothalamic activity in anorexia, we assessed carbohydrate and high-energy phosphate metabolism, in vivo and in vitro, in the anx/anx hypothalamus. In the fasted state, hypothalamic glucose uptake in the anx/anx mouse was reduced by ~50% of that seen in wild-type (wt) mice (P < 0.05). Under basal conditions, anx/anx hypothalamus ATP and glucose 6-P contents were similar to those in wt hypothalamus, whereas phosphocreatine was elevated (~2-fold; P < 0.001) and lactate was reduced (~35%; P < 0.001). The anx/anx hypothalamus had elevated total AMPK (~25%; P < 0.05) and GLUT4 (~60%; P < 0.01) protein contents, whereas GLUT1 and GLUT3 were similar to that of wt hypothalamus. Interestingly, the activation state of AMPK (ratio of phosphorylated AMPK/total AMPK) was significantly decreased in hypothalamus of the anx/anx mouse (~60% of that in wt; P < 0.05). Finally, during metabolic stress (ischemia), accumulation of lactate (measure of glycolysis) and IMP and AMP (breakdown products of ATP) were ~50% lower in anx/anx vs wt hypothalamus. These data demonstrate that carbohydrate and high-energy phosphate utilization in the anx/anx hypothalamus are diminished under basal and stress conditions. The decrease in hypothalamic metabolism may contribute to the anorectic behavior of the anx/anx mouse, i.e. its inability to regulate food intake in accordance with energy status.
Collapse
Affiliation(s)
- Ulrika Bergström
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Charlotte Lindfors
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Marie Svedberg
- Department of Clinical NeuroscienceCenter for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Jeanette E Johansen
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Jenny Häggkvist
- Department of Clinical NeuroscienceCenter for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Rolf Wibom
- Department of Medical Biochemistry and BiophysicsKarolinska Institutet, Stockholm, Sweden
| | - Abram Katz
- Department of Physical TherapyAriel University, Ariel, Israel
| | - Ida A K Nilsson
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
33
|
|
34
|
Nagatomo K, Suga S, Saitoh M, Kogawa M, Kobayashi K, Yamamoto Y, Yamada K. Dopamine D1 Receptor Immunoreactivity on Fine Processes of GFAP-Positive Astrocytes in the Substantia Nigra Pars Reticulata of Adult Mouse. Front Neuroanat 2017; 11:3. [PMID: 28203148 PMCID: PMC5285371 DOI: 10.3389/fnana.2017.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/13/2017] [Indexed: 12/20/2022] Open
Abstract
Substantia nigra pars reticulata (SNr), the major output nucleus of the basal ganglia, receives dopamine from dendrites extending from dopaminergic neurons of the adjacent nucleus pars compacta (SNc), which is known for its selective degeneration in Parkinson's disease. As a recipient for dendritically released dopamine, the dopamine D1 receptor (D1R) is a primary candidate due to its very dense immunoreactivity in the SNr. However, the precise location of D1R remains unclear at the cellular level in the SNr except for that reported on axons/axon terminals of presumably striatal GABAergic neurons. To address this, we used D1R promotor-controlled, mVenus-expressing transgenic mice. When cells were acutely dissociated from SNr of mouse brain, prominent mVenus fluorescence was detected in fine processes of glia-like cells, but no such fluorescence was detected from neurons in the same preparation, except for the synaptic bouton-like structure on the neurons. Double immunolabeling of SNr cells dissociated from adult wild-type mice brain further revealed marked D1R immunoreactivity in the processes of glial fibrillary acidic protein (GFAP)-positive astrocytes. Such D1R imunoreactivity was significantly stronger in the SNr astrocytes than that in those of the visual cortex in the same preparation. Interestingly, GFAP-positive astrocytes dissociated from the striatum demonstrated D1R immunoreactivity, either remarkable or minimal, similarly to that shown in neurons in this nucleus. In contrast, in the SNr and visual cortex, only weak D1R immunoreactivity was detected in the neurons tested. These results suggest that the SNr astrocyte may be a candidate recipient for dendritically released dopamine. Further study is required to fully elucidate the physiological roles of divergent dopamine receptor immunoreactivity profiles in GFAP-positive astrocytes.
Collapse
Affiliation(s)
- Katsuhiro Nagatomo
- Department of Physiology, Hirosaki University Graduate School of Medicine Aomori, Japan
| | - Sechiko Suga
- Department of Physiology, Hirosaki University Graduate School of MedicineAomori, Japan; Department of Emergency Medical Technology, Hirosaki University of Health and WelfareAomori, Japan
| | - Masato Saitoh
- Laboratory of Veterinary Anatomy and Cell Biology, Faculty of Agriculture, Iwate University Iwate, Japan
| | - Masahito Kogawa
- Department of Physiology, Hirosaki University Graduate School of Medicine Aomori, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine Fukushima, Japan
| | - Yoshio Yamamoto
- Laboratory of Veterinary Anatomy and Cell Biology, Faculty of Agriculture, Iwate University Iwate, Japan
| | - Katsuya Yamada
- Department of Physiology, Hirosaki University Graduate School of Medicine Aomori, Japan
| |
Collapse
|
35
|
Fogle KJ, Hertzler JI, Shon JH, Palladino MJ. The ATP-sensitive K channel is seizure protective and required for effective dietary therapy in a model of mitochondrial encephalomyopathy. J Neurogenet 2016; 30:247-258. [PMID: 27868454 DOI: 10.1080/01677063.2016.1252765] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Effective therapies are lacking for mitochondrial encephalomyopathies (MEs). MEs are devastating diseases that predominantly affect the energy-demanding tissues of the nervous system and muscle, causing symptoms such as seizures, cardiomyopathy, and neuro- and muscular degeneration. Even common anti-epileptic drugs which are frequently successful in ameliorating seizures in other diseases tend to have a lower success rate in ME, highlighting the need for novel drug targets, especially those that may couple metabolic sensitivity to neuronal excitability. Furthermore, alternative epilepsy therapies such as dietary modification are gaining in clinical popularity but have not been thoroughly studied in ME. Using the Drosophila ATP61 model of ME, we have studied dietary therapy throughout disease progression and found that it is highly effective against the seizures of ME, especially a high fat/ketogenic diet, and that the benefits are dependent upon a functional KATP channel complex. Further experiments with KATP show that it is seizure-protective in this model, and that pharmacological promotion of its open state also ameliorates seizures. These studies represent important steps forward in the development of novel therapies for a class of diseases that is notoriously difficult to treat, and lay the foundation for mechanistic studies of currently existing therapies in the context of metabolic disease.
Collapse
Affiliation(s)
- Keri J Fogle
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - J Ian Hertzler
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Joy H Shon
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Michael J Palladino
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| |
Collapse
|
36
|
Yamada K, Takahashi S, Karube F, Fujiyama F, Kobayashi K, Nishi A, Momiyama T. Neuronal circuits and physiological roles of the basal ganglia in terms of transmitters, receptors and related disorders. J Physiol Sci 2016; 66:435-446. [PMID: 26979514 PMCID: PMC5045844 DOI: 10.1007/s12576-016-0445-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/02/2016] [Indexed: 01/11/2023]
Abstract
The authors have reviewed recent research advances in basal ganglia circuitry and function, as well as in related disorders from multidisciplinary perspectives derived from the results of morphological, electrophysiological, behavioral, biochemical and molecular biological studies. Based on their expertise in their respective fields, as denoted in the text, the authors discuss five distinct research topics, as follows: (1) area-specific dopamine receptor expression of astrocytes in basal ganglia, (2) the role of physiologically released dopamine in the striatum, (3) control of behavioral flexibility by striatal cholinergic interneurons, (4) regulation of phosphorylation states of DARPP-32 by protein phosphatases and (5) physiological perspective on deep brain stimulation with optogenetics and closed-loop control for ameliorating parkinsonism.
Collapse
Affiliation(s)
- Katsuya Yamada
- Department of Physiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Susumu Takahashi
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Fuyuki Karube
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Fumino Fujiyama
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Fukushima Medical University, Fukushima, Japan
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Kurume, Japan
| | - Toshihiko Momiyama
- Department of Pharmacology, Jikei University School of Medicine, Nishi-Shinbashi Campus, Minato-ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
37
|
Forte N, Medrihan L, Cappetti B, Baldelli P, Benfenati F. 2-Deoxy-d-glucose enhances tonic inhibition through the neurosteroid-mediated activation of extrasynaptic GABA A receptors. Epilepsia 2016; 57:1987-2000. [PMID: 27735054 DOI: 10.1111/epi.13578] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The inhibition of glycolysis exerts potent antiseizure effects, as demonstrated by the efficacy of ketogenic and low-glucose/nonketogenic diets in the treatment of drug-resistant epilepsy. ATP-sensitive potassium (KATP ) channels have been initially identified as the main determinant of the reduction of neuronal hyperexcitability. However, a plethora of other mechanisms have been proposed. Herein, we report the ability of 2-deoxy-d-glucose (2-DG), a glucose analog that inhibits glycolytic enzymes, of potentiating γ-aminobutyric acid (GABA)ergic tonic inhibition via neurosteroid-mediated activation of extrasynaptic GABAA receptors. METHODS Acute effects of 2-DG on the ATP-sensitive potassium currents, GABAergic tonic inhibition, firing activity, and interictal events were assessed in hippocampal slices by whole-cell patch-clamp and local field potential recordings of dentate gyrus granule cells. RESULTS Acute application of 2-DG activates two distinct outward conductances: a KATP channel-mediated current and a bicuculline-sensitive tonic current. The effect of 2-DG on such GABAergic tonic currents was fully prevented by either finasteride or PK11195, which are specific inhibitors of the neurosteroidogenesis pathway acting via different mechanisms. Moreover, the oxidized form of vitamin C, dehydroascorbic acid, known for its ability to induce neurosteroidogenesis, also activated a bicuculline-sensitive tonic current in a manner indistinguishable from that of 2-DG. Finally, we found that the enhancement of KATP current by 2-DG primarily regulates intrinsic firing rate of granule cells, whereas the increase of the GABAergic tonic current plays a key role in reducing the frequency of interictal events evoked by treatment of hippocampal slices with the convulsive agent 4-aminopyridine. SIGNIFICANCE We demonstrated, for the first time, that 2-DG potentiates the extrasynaptic tonic GABAergic current through activation of neurosteroidogenesis. Such tonic inhibition represents the main conductance responsible for the antiseizure action of this glycolytic inhibitor.
Collapse
Affiliation(s)
- Nicola Forte
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Lucian Medrihan
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Beatrice Cappetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Pietro Baldelli
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
38
|
Lutas A, Lahmann C, Soumillon M, Yellen G. The leak channel NALCN controls tonic firing and glycolytic sensitivity of substantia nigra pars reticulata neurons. eLife 2016; 5. [PMID: 27177420 PMCID: PMC4902561 DOI: 10.7554/elife.15271] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/12/2016] [Indexed: 12/20/2022] Open
Abstract
Certain neuron types fire spontaneously at high rates, an ability that is crucial for their function in brain circuits. The spontaneously active GABAergic neurons of the substantia nigra pars reticulata (SNr), a major output of the basal ganglia, provide tonic inhibition of downstream brain areas. A depolarizing 'leak' current supports this firing pattern, but its molecular basis remains poorly understood. To understand how SNr neurons maintain tonic activity, we used single-cell RNA sequencing to determine the transcriptome of individual mouse SNr neurons. We discovered that SNr neurons express the sodium leak channel, NALCN, and that SNr neurons lacking NALCN have impaired spontaneous firing. In addition, NALCN is involved in the modulation of excitability by changes in glycolysis and by activation of muscarinic acetylcholine receptors. Our findings suggest that disruption of NALCN could impair the basal ganglia circuit, which may underlie the severe motor deficits in humans carrying mutations in NALCN. DOI:http://dx.doi.org/10.7554/eLife.15271.001 Some neurons in the brain produce electrical signals (or “fire”) spontaneously, without receiving any other signals from the senses or from other neurons. This spontaneous activity has a number of important roles. For example, in a part of the brain known as the substantia nigra pars reticulata (SNr), spontaneously active neurons frequently produce electrical signals that reduce electrical activity in other brain areas. A current of positively charged ions constantly flows into the spontaneously active SNr neurons and enables them to fire constantly. Ions enter neurons through proteins called ion channels that are embedded in the surface of the neuron. Like all proteins, ion channels are made by “transcribing” genes to form molecules of RNA that are then “translated” to produce the basic sequence of the protein. Lutas et al. have now used single-cell RNA sequencing to study SNr neurons from mice and investigate which ion channel the positive ion current flows through. The RNA sequences revealed that the neurons have the gene for an ion channel known as NALCN. Recordings of the firing rate of neurons in slices of mouse brain showed that SNr neurons without this channel did not fire as often as SNr neurons with the channel. In addition, neurotransmitters (chemicals that alter the ability of neurons to fire) and changes in cell metabolism had less of an effect on the firing rate of SNr neurons that lacked the NALCN channel than they do on normal neurons. These findings may help explain why people with mutations in the NALCN gene have movement disorders, as the substantia nigra pars reticulata plays an important role in orchestrating complex movements. Future work is now needed to understand how a change in NALCN activity affects the other brain areas that SNr neurons connect to. DOI:http://dx.doi.org/10.7554/eLife.15271.002
Collapse
Affiliation(s)
- Andrew Lutas
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Carolina Lahmann
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | | | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
39
|
Obata T, Nakashima M. Opening of ATP-sensitive K(+) (KATP) channels enhance hydroxyl radical generation induced by MPP(+) in rat striatum. J Neurol Sci 2016; 366:180-183. [PMID: 27288802 DOI: 10.1016/j.jns.2016.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 11/15/2022]
Abstract
The present study examined whether opening of adenosine triphosphate (ATP) sensitive K(+) (KATP) channels can enhance 1-methyl-4-phenylpyridinium (MPP(+))-induced hydroxyl radical (OH) generation in rat striatum. Rats were anesthetized, and sodium salicylate in Ringer's solution (0.5nmol/ml per min) was infused through a microdialysis probe to detect the generation of OH as reflected by the non-enzymatic formation of 2.3-dihydroxybenzoic acid (DHBA) in the striatum. MPP(+) (5mM) enhanced generation of OH with concomitant increased efflux of dopamine (DA). Cromakalim (100μM), a KATP channel opener, through the microdialysis probe significantly increased both DA efflux and OH formation induced by MPP(+). Another KATP channel opener, nicorandil (1mM), also increased the level DA or DHBA, but these changes were not significant. However, in the presence of glibenclamide (10μM), a KATP channel antagonist, and the increase of MPP(+)-induced DA or DHBA were not observed. Cromakalim (10, 50 and 100μM) increased MPP(+)-induced DHBA formation in a concentration-dependent manner. However, the effects of cromakalim in the presence of glibenclamide were abolished. These results suggest that opening of KATP channels may cause OH generation by MPP(+).
Collapse
Affiliation(s)
- Toshio Obata
- School of Nursing, Faculty of Health Sciences, Osaka Aoyama University, 2-11-1 Niina, Mino City, Japan.
| | - Michiko Nakashima
- Department of Nursing, School of Health Sciences, Asahi University, 1851 Hozumi, Mizuho City, Gifu, Japan
| |
Collapse
|
40
|
Abstract
KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.
Collapse
Affiliation(s)
- Monique N Foster
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - William A Coetzee
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
41
|
Quinn JC. Complex Membrane Channel Blockade: A Unifying Hypothesis for the Prodromal and Acute Neuropsychiatric Sequelae Resulting from Exposure to the Antimalarial Drug Mefloquine. J Parasitol Res 2015; 2015:368064. [PMID: 26576290 PMCID: PMC4630403 DOI: 10.1155/2015/368064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/28/2015] [Indexed: 12/18/2022] Open
Abstract
The alkaloid toxin quinine and its derivative compounds have been used for many centuries as effective medications for the prevention and treatment of malaria. More recently, synthetic derivatives, such as the quinoline derivative mefloquine (bis(trifluoromethyl)-(2-piperidyl)-4-quinolinemethanol), have been widely used to combat disease caused by chloroquine-resistant strains of the malaria parasite, Plasmodium falciparum. However, the parent compound quinine, as well as its more recent counterparts, suffers from an incidence of adverse neuropsychiatric side effects ranging from mild mood disturbances and anxiety to hallucinations, seizures, and psychosis. This review considers how the pharmacology, cellular neurobiology, and membrane channel kinetics of mefloquine could lead to the significant and sometimes life-threatening neurotoxicity associated with mefloquine exposure. A key role for mefloquine blockade of ATP-sensitive potassium channels and connexins in the substantia nigra is considered as a unifying hypothesis for the pathogenesis of severe neuropsychiatric events after mefloquine exposure in humans.
Collapse
Affiliation(s)
- Jane C. Quinn
- Plant and Animal Toxicology Group, School of Animal and Veterinary Sciences, Graham Centre for Agricultural Innovation, Charles Sturt University, Boorooma Street, Wagga Wagga, NSW 2650, Australia
| |
Collapse
|
42
|
Lee CR, Patel JC, O'Neill B, Rice ME. Inhibitory and excitatory neuromodulation by hydrogen peroxide: translating energetics to information. J Physiol 2015; 593:3431-46. [PMID: 25605547 DOI: 10.1113/jphysiol.2014.273839] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/16/2015] [Indexed: 12/21/2022] Open
Abstract
Historically, brain neurochemicals have been broadly classified as energetic or informational. However, increasing evidence implicates metabolic substrates and byproducts as signalling agents, which blurs the boundary between energy and information, and suggests the introduction of a new category for 'translational' substances that convey changes in energy state to information. One intriguing example is hydrogen peroxide (H2 O2 ), which is a small, readily diffusible molecule. Produced during mitochondrial respiration, this reactive oxygen species, can mediate dynamic regulation of neuronal activity and transmitter release by activating inhibitory ATP-sensitive K(+) (KATP ) channels, as well as a class of excitatory non-selective cation channels, TRPM2. Studies using ex vivo guinea pig brain slices have revealed that activity-generated H2 O2 can act via KATP channels to inhibit dopamine release in dorsal striatum and dopamine neuron activity in the substantia nigra pars compacta. In sharp contrast, endogenously generated H2 O2 enhances the excitability of GABAergic projection neurons in the dorsal striatum and substantia nigra pars reticulata by activating TRPM2 channels. These studies suggest that the balance of excitation vs. inhibition produced in a given cell by metabolically generated H2 O2 will be dictated by the relative abundance of H2 O2 -sensitive ion channel targets that receive this translational signal.
Collapse
Affiliation(s)
- Christian R Lee
- Department of Neurosurgery, New York University School of Medicine, New York, NY, 10016, USA
| | - Jyoti C Patel
- Department of Neurosurgery, New York University School of Medicine, New York, NY, 10016, USA
| | - Brian O'Neill
- Department of Neurosurgery, New York University School of Medicine, New York, NY, 10016, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA
| | - Margaret E Rice
- Department of Neurosurgery, New York University School of Medicine, New York, NY, 10016, USA.,Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA
| |
Collapse
|