1
|
Poblano J, Castillo-Tobías I, Berlanga L, Tamayo-Ordoñez MC, Del Carmen Rodríguez-Salazar M, Silva-Belmares SY, Aguayo-Morales H, Cobos-Puc LE. Drugs targeting APOE4 that regulate beta-amyloid aggregation in the brain: Therapeutic potential for Alzheimer's disease. Basic Clin Pharmacol Toxicol 2024; 135:237-249. [PMID: 39020526 DOI: 10.1111/bcpt.14055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/21/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024]
Abstract
Alzheimer's disease is characterized by progressive cognitive decline, and behavioural and psychological symptoms of dementia are common. The APOE ε4 allele, a genetic risk factor, significantly increases susceptibility to the disease. Despite efforts to effectively treat the disease, only seven drugs are approved for its treatment, and only two of these prevent its progression. This highlights the need to identify new pharmacological options. This review focuses on mimetic peptides, small molecule correctors and HAE-4 antibodies that target ApoE. These drugs reduce β-amyloid-induced neurodegeneration in preclinical models. In addition, loop diuretics such as bumetanide and furosemide show the potential to reduce the prevalence of Alzheimer's disease in humans, and antidepressants such as imipramine improve cognitive function in individuals diagnosed with Alzheimer's disease. Consistent with this, both classes of drugs have been shown to exert neuroprotective effects by inhibiting ApoE4-catalysed Aβ aggregation in preclinical models. Moreover, peroxisome proliferator-activated receptor ligands, particularly pioglitazone and rosiglitazone, reduce ApoE4-induced neurodegeneration in animal models. However, they do not prevent the cognitive decline in APOE ε4 allele carriers. Finally, ApoE4 impairs the integrity of the blood-brain barrier and haemostasis. On this basis, ApoE4 modulation is a promising avenue for the treatment of late-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Joan Poblano
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | - Ileana Castillo-Tobías
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | - Lia Berlanga
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | | | | | | | - Hilda Aguayo-Morales
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | - Luis E Cobos-Puc
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| |
Collapse
|
2
|
Delpire E, Terker AS, Gagnon KB. Pharmacology of Compounds Targeting Cation-Chloride Cotransporter Physiology. Handb Exp Pharmacol 2024; 283:249-284. [PMID: 37563251 PMCID: PMC10823342 DOI: 10.1007/164_2023_692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Transporters of the solute carrier family 12 (SLC12) carry inorganic cations such as Na+ and/or K+ alongside Cl across the plasma membrane of cells. These tightly coupled, electroneutral, transporters are expressed in almost all tissues/organs in the body where they fulfil many critical functions. The family includes two key transporters participating in salt reabsorption in the kidney: the Na-K-2Cl cotransporter-2 (NKCC2), expressed in the loop of Henle, and the Na-Cl cotransporter (NCC), expressed in the distal convoluted tubule. NCC and NKCC2 are the targets of thiazides and "loop" diuretics, respectively, drugs that are widely used in clinical medicine to treat hypertension and edema. Bumetanide, in addition to its effect as a loop diuretic, has recently received increasing attention as a possible therapeutic agent for neurodevelopmental disorders. This chapter also describes how over the past two decades, the pharmacology of Na+ independent transporters has expanded significantly to provide novel tools for research. This work has indeed led to the identification of compounds that are 100-fold to 1000-fold more potent than furosemide, the first described inhibitor of K-Cl cotransport, and identified compounds that possibly directly stimulate the function of the K-Cl cotransporter. Finally, the recent cryo-electron microscopy revolution has begun providing answers as to where and how pharmacological agents bind to and affect the function of the transporters.
Collapse
Affiliation(s)
- Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Andrew S Terker
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kenneth B Gagnon
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
3
|
Graber-Naidich A, Lee J, Younes K, Greicius MD, Le Guen Y, He Z. Loop diuretics association with Alzheimer's disease risk. FRONTIERS IN AGING 2023; 4:1211571. [PMID: 37822457 PMCID: PMC10563814 DOI: 10.3389/fragi.2023.1211571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/07/2023] [Indexed: 10/13/2023]
Abstract
Objectives: To investigate whether exposure history to two common loop diuretics, bumetanide and furosemide, affects the risk of developing Alzheimer's disease (AD) after accounting for socioeconomic status and congestive heart failure. Methods: Individuals exposed to bumetanide or furosemide were identified in the Stanford University electronic health record using the de-identified Observational Medical Outcomes Partnership platform. We matched the AD case cohort to a control cohort (1:20 case:control) on gender, race, ethnicity, and hypertension, and controlled for variables that could potentially be collinear with bumetanide exposure and/or AD diagnosis. Among individuals older than 65 years, 5,839 AD cases and 116,103 matched controls were included. A total of 1,759 patients (54 cases and 1,705 controls) were exposed to bumetanide. Results: After adjusting for socioeconomic status and other confounders, the exposure of bumetanide and furosemide was significantly associated with reduced AD risk (respectively, bumetanide odds ratio [OR] = 0.23; 95% confidence interval [CI], 0.15-0.36; p = 4.0 × 10-11; furosemide OR = 0.42; 95% CI, 0.38-0.47; p < 2.0 × 10-16). Discussion: Our study replicates in an independent sample that a history of bumetanide exposure is associated with reduced AD risk while also highlighting an association of the most common loop diuretic (furosemide) with reduced AD risk. These associations need to be additionally replicated, and the mechanism of action remains to be investigated.
Collapse
Affiliation(s)
- Anna Graber-Naidich
- Quantitative Sciences Unit, Department of Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA, United States
| | - Justin Lee
- Quantitative Sciences Unit, Department of Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA, United States
| | - Kyan Younes
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Michael D. Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Yann Le Guen
- Quantitative Sciences Unit, Department of Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA, United States
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Zihuai He
- Quantitative Sciences Unit, Department of Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA, United States
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| |
Collapse
|
4
|
Boyarko B, Podvin S, Greenberg B, Momper JD, Huang Y, Gerwick WH, Bang AG, Quinti L, Griciuc A, Kim DY, Tanzi RE, Feldman HH, Hook V. Evaluation of bumetanide as a potential therapeutic agent for Alzheimer's disease. Front Pharmacol 2023; 14:1190402. [PMID: 37601062 PMCID: PMC10436590 DOI: 10.3389/fphar.2023.1190402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/28/2023] [Indexed: 08/22/2023] Open
Abstract
Therapeutics discovery and development for Alzheimer's disease (AD) has been an area of intense research to alleviate memory loss and the underlying pathogenic processes. Recent drug discovery approaches have utilized in silico computational strategies for drug candidate selection which has opened the door to repurposing drugs for AD. Computational analysis of gene expression signatures of patients stratified by the APOE4 risk allele of AD led to the discovery of the FDA-approved drug bumetanide as a top candidate agent that reverses APOE4 transcriptomic brain signatures and improves memory deficits in APOE4 animal models of AD. Bumetanide is a loop diuretic which inhibits the kidney Na+-K+-2Cl- cotransporter isoform, NKCC2, for the treatment of hypertension and edema in cardiovascular, liver, and renal disease. Electronic health record data revealed that patients exposed to bumetanide have lower incidences of AD by 35%-70%. In the brain, bumetanide has been proposed to antagonize the NKCC1 isoform which mediates cellular uptake of chloride ions. Blocking neuronal NKCC1 leads to a decrease in intracellular chloride and thus promotes GABAergic receptor mediated hyperpolarization, which may ameliorate disease conditions associated with GABAergic-mediated depolarization. NKCC1 is expressed in neurons and in all brain cells including glia (oligodendrocytes, microglia, and astrocytes) and the vasculature. In consideration of bumetanide as a repurposed drug for AD, this review evaluates its pharmaceutical properties with respect to its estimated brain levels across doses that can improve neurologic disease deficits of animal models to distinguish between NKCC1 and non-NKCC1 mechanisms. The available data indicate that bumetanide efficacy may occur at brain drug levels that are below those required for inhibition of the NKCC1 transporter which implicates non-NKCC1 brain mechansims for improvement of brain dysfunctions and memory deficits. Alternatively, peripheral bumetanide mechanisms may involve cells outside the central nervous system (e.g., in epithelia and the immune system). Clinical bumetanide doses for improved neurological deficits are reviewed. Regardless of mechanism, the efficacy of bumetanide to improve memory deficits in the APOE4 model of AD and its potential to reduce the incidence of AD provide support for clinical investigation of bumetanide as a repurposed AD therapeutic agent.
Collapse
Affiliation(s)
- Ben Boyarko
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Sonia Podvin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Barry Greenberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jeremiah D. Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, United States
- Departments of Neurology and Pathology, University of California, San Francisco, San Francisco, CA, United States
| | - William H. Gerwick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, United States
| | - Anne G. Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys, San Diego, CA, United States
| | - Luisa Quinti
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Ana Griciuc
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Howard H. Feldman
- Department of Neurosciences and Department of Pharmacology, University of California, San Diego, San Diego, United States
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, La Jolla, CA, United States
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences and Department of Pharmacology, University of California, San Diego, San Diego, United States
| |
Collapse
|
5
|
Pressey JC, de Saint-Rome M, Raveendran VA, Woodin MA. Chloride transporters controlling neuronal excitability. Physiol Rev 2023; 103:1095-1135. [PMID: 36302178 DOI: 10.1152/physrev.00025.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Synaptic inhibition plays a crucial role in regulating neuronal excitability, which is the foundation of nervous system function. This inhibition is largely mediated by the neurotransmitters GABA and glycine that activate Cl--permeable ion channels, which means that the strength of inhibition depends on the Cl- gradient across the membrane. In neurons, the Cl- gradient is primarily mediated by two secondarily active cation-chloride cotransporters (CCCs), NKCC1 and KCC2. CCC-mediated regulation of the neuronal Cl- gradient is critical for healthy brain function, as dysregulation of CCCs has emerged as a key mechanism underlying neurological disorders including epilepsy, neuropathic pain, and autism spectrum disorder. This review begins with an overview of neuronal chloride transporters before explaining the dependent relationship between these CCCs, Cl- regulation, and inhibitory synaptic transmission. We then discuss the evidence for how CCCs can be regulated, including by activity and their protein interactions, which underlie inhibitory synaptic plasticity. For readers who may be interested in conducting experiments on CCCs and neuronal excitability, we have included a section on techniques for estimating and recording intracellular Cl-, including their advantages and limitations. Although the focus of this review is on neurons, we also examine how Cl- is regulated in glial cells, which in turn regulate neuronal excitability through the tight relationship between this nonneuronal cell type and synapses. Finally, we discuss the relatively extensive and growing literature on how CCC-mediated neuronal excitability contributes to neurological disorders.
Collapse
Affiliation(s)
- Jessica C Pressey
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Miranda de Saint-Rome
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vineeth A Raveendran
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Melanie A Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Lam P, Newland J, Faull RLM, Kwakowsky A. Cation-Chloride Cotransporters KCC2 and NKCC1 as Therapeutic Targets in Neurological and Neuropsychiatric Disorders. Molecules 2023; 28:1344. [PMID: 36771011 PMCID: PMC9920462 DOI: 10.3390/molecules28031344] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Neurological diseases including Alzheimer's, Huntington's disease, Parkinson's disease, Down syndrome and epilepsy, and neuropsychiatric disorders such as schizophrenia, are conditions that affect not only individuals but societies on a global scale. Current therapies offer a means for small symptomatic relief, but recently there has been increasing demand for therapeutic alternatives. The γ-aminobutyric acid (GABA)ergic signaling system has been investigated for developing new therapies as it has been noted that any dysfunction or changes to this system can contribute to disease progression. Expression of the K-Cl-2 (KCC2) and N-K-C1-1 (NKCC1) cation-chloride cotransporters (CCCs) has recently been linked to the disruption of GABAergic activity by affecting the polarity of GABAA receptor signaling. KCC2 and NKCC1 play a part in multiple neurological and neuropsychiatric disorders, making them a target of interest for potential therapies. This review explores current research suggesting the pathophysiological role and therapeutic importance of KCC2 and NKCC1 in neuropsychiatric and neurological disorders.
Collapse
Affiliation(s)
- Patricia Lam
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Julia Newland
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Richard L. M. Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
7
|
Nasirinezhad F, Zarepour L, Hadjighassem M, Gharaylou Z, Majedi H, Ramezani F. Analgesic Effect of Bumetanide on Neuropathic Pain in Patients With Spinal Cord Injury. Basic Clin Neurosci 2021; 12:409-420. [PMID: 34917299 PMCID: PMC8666921 DOI: 10.32598/bcn.12.3.2049.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/15/2019] [Accepted: 01/21/2020] [Indexed: 11/20/2022] Open
Abstract
Introduction The current study evaluated the analgesic effects of bumetanide as an adjunctive in the management of neuropathic pain following Spinal Cord Injury (SCI). The peripheral expression of Na-K-Cl Cotransporter-1 (NKCC1) and K-Cl Cotransporter-2 (KCC2) genes in polymorphonuclear lymphocytes (PMLs) was assessed as a possible biomarker indicating central mechanisms underlying the observed response. Methods Through an open-label, single-arm, pilot trial of bumetanide (2 mg/d), an add-on treatment was conducted on 14 SCI patients for 19 weeks. This study consisted of 3 phases: pre-treatment (1 month), titration (3 weeks), and active treatment (4 months). Ultimately, 9 patients completed the study. The primary outcome variables were the endpoint pain score using the Numeric Rating Scale (NRS), and also the short-form of the McGill pain questionnaire. Secondary endpoints included the short-form of the health survey that assesses the quality of life. Blood samples were collected and used for determining the expression of NKCC1 and KCC2 genes in transcription and translation levels. Results Bumetanide treatment significantly decreased average pain intensity according to the NRS and the short-form of the McGill pain questionnaire scores. Baseline expression of KCC2 protein was low between groups and increased significantly following treatment (P<0.05). In the current study, pain improvement was accompanied by the greater mean change from the baseline (improvement) for the overall quality of life. Conclusion These data highlighted the analgesic effect of bumetanide on neuropathic pain and indicated the potential role of the upregulation of KCC2 protein and involvement of GABAergic disinhibition in producing neuropathic pain.
Collapse
Affiliation(s)
- Farinaz Nasirinezhad
- Department of Physiology, Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Zarepour
- Department of Physiology, Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Gharaylou
- Brain and Spinal cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossin Majedi
- Brain and Spinal cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Department of Physiology, Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
The Alteration of Chloride Homeostasis/GABAergic Signaling in Brain Disorders: Could Oxidative Stress Play a Role? Antioxidants (Basel) 2021; 10:antiox10081316. [PMID: 34439564 PMCID: PMC8389245 DOI: 10.3390/antiox10081316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022] Open
Abstract
In neuronal precursors and immature neurons, the depolarizing (excitatory) effect of γ-Aminobutyric acid (GABA) signaling is associated with elevated [Cl−]i; as brain cells mature, a developmental switch occurs, leading to the decrease of [Cl−]i and to the hyperpolarizing (inhibitory) effect of GABAergic signaling. [Cl−]i is controlled by two chloride co-transporters: NKCC1, which causes Cl− to accumulate into the cells, and KCC2, which extrudes it. The ontogenetic upregulation of the latter determines the above-outlined switch; however, many other factors contribute to the correct [Cl−]i in mature neurons. The dysregulation of chloride homeostasis is involved in seizure generation and has been associated with schizophrenia, Down’s Syndrome, Autism Spectrum Disorder, and other neurodevelopmental disorders. Recently, much effort has been put into developing new drugs intended to inhibit NKCC1 activity, while no attention has been paid to the origin of [Cl−]i dysregulation. Our study examines the pathophysiology of Cl− homeostasis and focuses on the impact of oxidative stress (OS) and inflammation on the activity of Cl− co-transporters, highlighting the relevance of OS in numerous brain abnormalities and diseases. This hypothesis supports the importance of primary prevention during pregnancy. It also integrates the therapeutic framework addressed to restore normal GABAergic signaling by counteracting the alteration in chloride homeostasis in central nervous system (CNS) cells, aiming at limiting the use of drugs that potentially pose a health risk.
Collapse
|
9
|
Borgogno M, Savardi A, Manigrasso J, Turci A, Portioli C, Ottonello G, Bertozzi SM, Armirotti A, Contestabile A, Cancedda L, De Vivo M. Design, Synthesis, In Vitro and In Vivo Characterization of Selective NKCC1 Inhibitors for the Treatment of Core Symptoms in Down Syndrome. J Med Chem 2021; 64:10203-10229. [PMID: 34137257 PMCID: PMC8311653 DOI: 10.1021/acs.jmedchem.1c00603] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Intracellular chloride concentration [Cl-]i is defective in several neurological disorders. In neurons, [Cl-]i is mainly regulated by the action of the Na+-K+-Cl- importer NKCC1 and the K+-Cl- exporter KCC2. Recently, we have reported the discovery of ARN23746 as the lead candidate of a novel class of selective inhibitors of NKCC1. Importantly, ARN23746 is able to rescue core symptoms of Down syndrome (DS) and autism in mouse models. Here, we describe the discovery and extensive characterization of this chemical class of selective NKCC1 inhibitors, with focus on ARN23746 and other promising derivatives. In particular, we present compound 40 (ARN24092) as a backup/follow-up lead with in vivo efficacy in a mouse model of DS. These results further strengthen the potential of this new class of compounds for the treatment of core symptoms of brain disorders characterized by the defective NKCC1/KCC2 expression ratio.
Collapse
Affiliation(s)
- Marco Borgogno
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Dulbecco Telethon Institute, 38123 Rome, Italy
| | - Jacopo Manigrasso
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Alessandra Turci
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Università degli Studi di Genova, via Balbi, 5, 16126 Genoa, Italy
| | - Corinne Portioli
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Giuliana Ottonello
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Sine Mandrup Bertozzi
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy.,Dulbecco Telethon Institute, 38123 Rome, Italy
| | - Marco De Vivo
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| |
Collapse
|
10
|
Kim HR, Rajagopal L, Meltzer HY, Martina M. Depolarizing GABA A current in the prefrontal cortex is linked with cognitive impairment in a mouse model relevant for schizophrenia. SCIENCE ADVANCES 2021; 7:eaba5032. [PMID: 33789887 PMCID: PMC8011979 DOI: 10.1126/sciadv.aba5032] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/12/2021] [Indexed: 05/06/2023]
Abstract
Cognitive impairment in schizophrenia (CIAS) is the most critical predictor of functional outcome. Limited understanding of the cellular mechanisms of CIAS hampers development of more effective treatments. We found that in subchronic phencyclidine (scPCP)-treated mice, an animal model that mimics CIAS, the reversal potential of GABAA currents in pyramidal neurons of the infralimbic prefrontal cortex (ILC) shifts from hyperpolarizing to depolarizing, the result of increased expression of the chloride transporter NKCC1. Further, we found that in scPCP mice, the NKCC1 antagonist bumetanide normalizes GABAA current polarity ex vivo and improves performance in multiple cognitive tasks in vivo. This behavioral effect was mimicked by selective, bilateral, NKCC1 knockdown in the ILC. Thus, we show that depolarizing GABAA currents in the ILC contributes to cognitive impairments in scPCP mice and suggest that bumetanide, an FDA-approved drug, has potential to treat or prevent CIAS and other components of the schizophrenia syndrome.
Collapse
Affiliation(s)
- Haram R Kim
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 300 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Lakshmi Rajagopal
- Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, 300 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Herbert Y Meltzer
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 300 E. Chicago Avenue, Chicago, IL 60611, USA.
- Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, 300 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Marco Martina
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 300 E. Chicago Avenue, Chicago, IL 60611, USA.
- Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, 300 E. Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|
11
|
Pergakis M, Badjatia N, Simard JM. An update on the pharmacological management and prevention of cerebral edema: current therapeutic strategies. Expert Opin Pharmacother 2021; 22:1025-1037. [PMID: 33467932 DOI: 10.1080/14656566.2021.1876663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Cerebral edema is a common complication of multiple neurological diseases and is a strong predictor of outcome, especially in traumatic brain injury and large hemispheric infarction.Areas Covered: Traditional and current treatments of cerebral edema include treatment with osmotherapy or decompressive craniectomy at the time of clinical deterioration. The authors discuss preclinical and clinical models of a variety of neurological disease states that have identified receptors, ion transporters, and channels involved in the development of cerebral edema as well as modulation of these receptors with promising agents.Expert opinion: Further study is needed on the safety and efficacy of the agents discussed. IV glibenclamide has shown promise in preclinical and clinical trials of cerebral edema in large hemispheric infarct and traumatic brain injury. Consideration of underlying pathophysiology and pharmacodynamics is vital, as the synergistic use of agents has the potential to drastically mitigate cerebral edema and secondary brain injury thusly transforming our treatment paradigms.
Collapse
Affiliation(s)
- Melissa Pergakis
- Program in Trauma Department of Neurology University of Maryland School of Medicine,Baltimore MD USA
| | - Neeraj Badjatia
- Program in Trauma Department of Neurology University of Maryland School of Medicine,Baltimore MD USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Josiah SS, Meor Azlan NF, Zhang J. Targeting the WNK-SPAK/OSR1 Pathway and Cation-Chloride Cotransporters for the Therapy of Stroke. Int J Mol Sci 2021; 22:1232. [PMID: 33513812 PMCID: PMC7865768 DOI: 10.3390/ijms22031232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/05/2023] Open
Abstract
Stroke is one of the major culprits responsible for morbidity and mortality worldwide, and the currently available pharmacological strategies to combat this global disease are scanty. Cation-chloride cotransporters (CCCs) are expressed in several tissues (including neurons) and extensively contribute to the maintenance of numerous physiological functions including chloride homeostasis. Previous studies have implicated two CCCs, the Na+-K+-Cl- and K+-Cl- cotransporters (NKCCs and KCCs) in stroke episodes along with their upstream regulators, the with-no-lysine kinase (WNKs) family and STE20/SPS1-related proline/alanine rich kinase (SPAK) or oxidative stress response kinase (OSR1) via a signaling pathway. As the WNK-SPAK/OSR1 pathway reciprocally regulates NKCC and KCC, a growing body of evidence implicates over-activation and altered expression of NKCC1 in stroke pathology whilst stimulation of KCC3 during and even after a stroke event is neuroprotective. Both inhibition of NKCC1 and activation of KCC3 exert neuroprotection through reduction in intracellular chloride levels and thus could be a novel therapeutic strategy. Hence, this review summarizes the current understanding of functional regulations of the CCCs implicated in stroke with particular focus on NKCC1, KCC3, and WNK-SPAK/OSR1 signaling and discusses the current and potential pharmacological treatments for stroke.
Collapse
Affiliation(s)
| | | | - Jinwei Zhang
- Hatherly Laboratories, Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Exeter EX4 4PS, UK; (S.S.J.); (N.F.M.A.)
| |
Collapse
|
13
|
Zarepour L, Gharaylou Z, Hadjighassem M, Shafaghi L, Majedi H, Behzad E, Hosseindoost S, Ramezani F, Nasirinezhad F. Preliminary study of analgesic effect of bumetanide on neuropathic pain in patients with spinal cord injury. J Clin Neurosci 2020; 81:477-484. [PMID: 33222966 DOI: 10.1016/j.jocn.2020.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/07/2020] [Accepted: 10/03/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND/OBJECTIVE The current study evaluated the analgesic effects of bumetanide as an adjunctive treatment in managing neuropathic pain following spinal cord injury. The peripheral expression level of Na-K-Cl-cotransporter-1 (NKCC1) and K-Cl-cotransporter-2 (KCC2) genes in polymorphonuclear lymphocytes (PMLs) assessed as a possible biomarker indicating central underlying mechanisms. METHODS This open-label, single-arm, pilot trial of bumetanide (2 mg/day) is an add-on treatment conducted in 14 SCI patients for 19 weeks. The whole duration consisted of three phases: pre-treatment (1 month), titration (3 weeks), and active treatment (4 months). Ultimately, nine patients completed the study. The primary outcome variables were the endpoint pain score measured by the numeric rating scale (NRS), and the short-form McGill Pain Questionnaire. Secondary endpoints included the Short-Form Health Survey that measures the quality of life. Blood samples were collected and used for determining the expression of NKCC1 and KCC2 genes in transcription and translation levels. RESULTS Bumetanide treatment significantly reduced average pain intensity according to the NRS and the short form of the McGill Pain Questionnaire scores. The baseline expression of KCC2 protein was low between groups and increased significantly following treatment (P < 0.05). Through the current study, pain improvement accompanied by the more significant mean change from the baseline for the overall quality of life. CONCLUSION These data might be a piece of preliminary evidence for the analgesic effect of bumetanide on neuropathic pain and could support the potential role of the upregulation of KCC2 protein and involvement of GABAergic disinhibition in producing neuropathic pain.
Collapse
Affiliation(s)
- Leila Zarepour
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Gharaylou
- Multiple Sclerosis Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Lida Shafaghi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Majedi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Behzad
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saereh Hosseindoost
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farinaz Nasirinezhad
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Murillo-de-Ozores AR, Chávez-Canales M, de los Heros P, Gamba G, Castañeda-Bueno M. Physiological Processes Modulated by the Chloride-Sensitive WNK-SPAK/OSR1 Kinase Signaling Pathway and the Cation-Coupled Chloride Cotransporters. Front Physiol 2020; 11:585907. [PMID: 33192599 PMCID: PMC7606576 DOI: 10.3389/fphys.2020.585907] [Citation(s) in RCA: 30] [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: 07/21/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
The role of Cl- as an intracellular signaling ion has been increasingly recognized in recent years. One of the currently best described roles of Cl- in signaling is the modulation of the With-No-Lysine (K) (WNK) - STE20-Proline Alanine rich Kinase (SPAK)/Oxidative Stress Responsive Kinase 1 (OSR1) - Cation-Coupled Cl- Cotransporters (CCCs) cascade. Binding of a Cl- anion to the active site of WNK kinases directly modulates their activity, promoting their inhibition. WNK activation due to Cl- release from the binding site leads to phosphorylation and activation of SPAK/OSR1, which in turn phosphorylate the CCCs. Phosphorylation by WNKs-SPAK/OSR1 of the Na+-driven CCCs (mediating ions influx) promote their activation, whereas that of the K+-driven CCCs (mediating ions efflux) promote their inhibition. This results in net Cl- influx and feedback inhibition of WNK kinases. A wide variety of alterations to this pathway have been recognized as the cause of several human diseases, with manifestations in different systems. The understanding of WNK kinases as Cl- sensitive proteins has allowed us to better understand the mechanistic details of regulatory processes involved in diverse physiological phenomena that are reviewed here. These include cell volume regulation, potassium sensing and intracellular signaling in the renal distal convoluted tubule, and regulation of the neuronal response to the neurotransmitter GABA.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Chávez-Canales
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paola de los Heros
- Unidad de Investigación UNAM-INC, Research Division, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
15
|
Savardi A, Borgogno M, Narducci R, La Sala G, Ortega JA, Summa M, Armirotti A, Bertorelli R, Contestabile A, De Vivo M, Cancedda L. Discovery of a Small Molecule Drug Candidate for Selective NKCC1 Inhibition in Brain Disorders. Chem 2020; 6:2073-2096. [PMID: 32818158 PMCID: PMC7427514 DOI: 10.1016/j.chempr.2020.06.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/14/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023]
Abstract
Aberrant expression ratio of Cl− transporters, NKCC1 and KCC2, is implicated in several brain conditions. NKCC1 inhibition by the FDA-approved diuretic drug, bumetanide, rescues core symptoms in rodent models and/or clinical trials with patients. However, bumetanide has a strong diuretic effect due to inhibition of the kidney Cl− transporter NKCC2, creating critical drug compliance issues and health concerns. Here, we report the discovery of a new chemical class of selective NKCC1 inhibitors and the lead drug candidate ARN23746. ARN23746 restores the physiological intracellular Cl− in murine Down syndrome neuronal cultures, has excellent solubility and metabolic stability, and displays no issues with off-target activity in vitro. ARN23746 recovers core symptoms in mouse models of Down syndrome and autism, with no diuretic effect, nor overt toxicity upon chronic treatment in adulthood. ARN23746 is ready for advanced preclinical/manufacturing studies toward the first sustainable therapeutics for the neurological conditions characterized by impaired Cl− homeostasis. NKCC1 is a promising target for the treatment of brain disorders The newly discovered ARN23746 presents selective NKCC1 versus NKCC2 and KCC2 inhibition ARN23746 restores altered neuronal chloride homeostasis in vitro ARN23746 rescues core behaviors in DS and ASD mice with no diuretic effect or toxicity
In the last few decades, drug development for brain disorders has struggled to deliver effective small molecules as novel breakthrough classes of drugs. Discovery of effective chemical compounds for brain disorders has been greatly hampered by the fact that the few currently clinically used drugs were identified by serendipity, and these drugs’ mechanism of action is often poorly understood. Here, by leveraging drug repurposing as a means to quickly and safely evaluate the new pharmacological target NKCC1 and its implications in brain disorders in animal models and patients, we report an integrated strategy for the rational design and discovery of a novel, selective, and safe NKCC1 inhibitor, active in vivo. This compound has the potential to become a clinical drug candidate to treat several neurological conditions in patients. Eventually, this integrated drug-discovery strategy has the prospective to revive the appeal of drug-discovery programs in the challenging field of neuroscience.
Collapse
Affiliation(s)
- Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Università degli Studi di Genova, Via Balbi, 5, 16126 Genoa, Italy
| | - Marco Borgogno
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Roberto Narducci
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Giuseppina La Sala
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Jose Antonio Ortega
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Maria Summa
- In Vivo Pharmacology Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Rosalia Bertorelli
- In Vivo Pharmacology Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Contestabile
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Marco De Vivo
- Molecular Modeling and Drug Discovery Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Corresponding author
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Dulbecco Telethon Institute, Via Orus 2, 35129 Padova, Italy
- Corresponding author
| |
Collapse
|
16
|
Tao D, Liu F, Sun X, Qu H, Zhao S, Zhou Z, Xiao T, Zhao C, Zhao M. Bumetanide: A review of its neuroplasticity and behavioral effects after stroke. Restor Neurol Neurosci 2020; 37:397-407. [PMID: 31306143 DOI: 10.3233/rnn-190926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Stroke often leads to neuronal injury and neurological functional deficits. Whilst spontaneous neurogenesis and axon regeneration are induced by ischemic stroke, effective pharmacological treatments are also essential for the improvement of neuroplasticity and functional recovery after stroke. However, no pharmacological therapy has been demonstrated to be able to effectively improve the functional recovery after stroke. Bumetanide is a specific Na+-K+-Cl- co-transporter inhibitor which can maintain chloride homeostasis in neurons. Therefore, many studies have focused on this drug's effect in stroke recovery in recent years. Here, we first review the function of Na+-K+-Cl- co-transporter in neurons, then how bumetanide's role in reducing brain damage, promoting neuroplasticity, leading to functional recovery after stroke, is elucidated. Finally, we discuss current limitations of bumetanide's efficiency and their potential solutions. These results may provide new avenues for further exploring mechanisms of post-stroke functional recovery as well as promising therapeutic targets for functional disability rehabilitation after ischemic stroke.
Collapse
Affiliation(s)
- Dongxia Tao
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Fangxi Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaoyu Sun
- Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Huiling Qu
- Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Shanshan Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Zhike Zhou
- Geriatrics, The First Hospital of China Medical University, Shenyang, China
| | - Ting Xiao
- Dermatology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, China
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Mei Zhao
- Cardiology, The Shengjing Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
17
|
Maguire JL. Get With the (Developmental) Program. Epilepsy Curr 2020; 20:102-104. [PMID: 32313506 PMCID: PMC7160877 DOI: 10.1177/1535759720901606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Impaired Regulation of KCC2 Phosphorylation Leads to Neuronal Network Dysfunction and Neurodevelopmental Pathology Pisella LI, Gaiarsa JL, Diabira D, et al. Sci Signal. 2019:12(603):eaay0300. doi:10.1126/scisignal.aay0300. KCC2 is a vital neuronal K+/Cl− cotransporter that is implicated in the etiology of numerous neurological diseases. In normal cells, KCC2 undergoes developmental dephosphorylation at Thr906 and Thr1007. We engineered mice with heterozygous phosphomimetic mutations T906E and T1007E (KCC2E/+) to prevent the normal developmental dephosphorylation of these sites. Immature (postnatal day 15) but not juvenile (postnatal day 30) KCC2E/+ mice exhibited altered GABAergic inhibition, an increased glutamate/GABA synaptic ratio, and greater susceptibility to seizure. KCC2E/+ mice also had abnormal ultrasonic vocalizations at postnatal days 10 to 12 and impaired social behavior at postnatal day 60. Postnatal bumetanide treatment restored network activity by postnatal day 15 but failed to restore social behavior by postnatal day 60. Our data indicate that posttranslational KCC2 regulation controls the GABAergic developmental sequence in vivo, indicating that deregulation of KCC2 could be a risk factor for the emergence of neurological pathology. Developmental Regulation of KCC2 Phosphorylation Has Long-Term Impacts on Cognitive Function Moore YE, Conway LC, Wobst HJ, et al. Front Mol Neurosci. 2019;12:173. doi:10.3389/fnmol.2019.00173. The GABAA receptor-mediated currents shift from excitatory to inhibitory during postnatal brain development in rodents. A postnatal increase in KCC2 protein expression is considered to be the sole mechanism controlling the developmental onset of hyperpolarizing synaptic transmission, but here we identify a key role for KCC2 phosphorylation in the developmental EGABA shift. Preventing phosphorylation of KCC2 in vivo at either residue serine 940 (S940), or at residues threonine 906 and threonine 1007 (T906/T1007), delayed or accelerated the postnatal onset of KCC2 function, respectively. Several models of neurodevelopmental disorders including Rett syndrome, Fragile × and Down syndrome exhibit delayed postnatal onset of hyperpolarizing GABAergic inhibition, but whether the timing of the onset of hyperpolarizing synaptic inhibition during development plays a role in establishing adulthood cognitive function is unknown; we have used the distinct KCC2-S940A and KCC2-T906A/T1007A knock-in mouse models to address this issue. Altering KCC2 function resulted in long-term abnormalities in social behavior and memory retention. Tight regulation of KCC2 phosphorylation is therefore required for the typical timing of the developmental onset of hyperpolarizing synaptic inhibition, and it plays a fundamental role in the regulation of adulthood cognitive function.
Collapse
|
18
|
Waddington JL, Zhen X, O'Tuathaigh CMP. Developmental Genes and Regulatory Proteins, Domains of Cognitive Impairment in Schizophrenia Spectrum Psychosis and Implications for Antipsychotic Drug Discovery: The Example of Dysbindin-1 Isoforms and Beyond. Front Pharmacol 2020; 10:1638. [PMID: 32063853 PMCID: PMC7000454 DOI: 10.3389/fphar.2019.01638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
Alongside positive and negative symptomatology, deficits in working memory, attention, selective learning processes, and executive function have been widely documented in schizophrenia spectrum psychosis. These cognitive abnormalities are strongly associated with impairment across multiple function domains and are generally treatment-resistant. The DTNBP1 (dystrobrevin-binding protein-1) gene, encoding dysbindin, is considered a risk factor for schizophrenia and is associated with variation in cognitive function in both clinical and nonclinical samples. Downregulation of DTNBP1 expression in dorsolateral prefrontal cortex and hippocampal formation of patients with schizophrenia has been suggested to serve as a primary pathophysiological process. Described as a "hub," dysbindin is an important regulatory protein that is linked with multiple complexes in the brain and is involved in a wide variety of functions implicated in neurodevelopment and neuroplasticity. The expression pattern of the various dysbindin isoforms (-1A, -1B, -1C) changes depending upon stage of brain development, tissue areas and subcellular localizations, and can involve interaction with different protein partners. We review evidence describing how sequence variation in DTNBP1 isoforms has been differentially associated with schizophrenia-associated symptoms. We discuss results linking these isoform proteins, and their interacting molecular partners, with cognitive dysfunction in schizophrenia, including evidence from drosophila through to genetic mouse models of dysbindin function. Finally, we discuss preclinical evidence investigating the antipsychotic potential of molecules that influence dysbindin expression and functionality. These studies, and other recent work that has extended this approach to other developmental regulators, may facilitate identification of novel molecular pathways leading to improved antipsychotic treatments.
Collapse
Affiliation(s)
- John L Waddington
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Colm M P O'Tuathaigh
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Medical Education Unit, School of Medicine, Brookfield Health Sciences Complex, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
Gharaylou Z, Shafaghi L, Oghabian MA, Yoonessi A, Tafakhori A, Shahsavand Ananloo E, Hadjighassem M. Longitudinal Effects of Bumetanide on Neuro-Cognitive Functioning in Drug-Resistant Epilepsy. Front Neurol 2019; 10:483. [PMID: 31133976 PMCID: PMC6517515 DOI: 10.3389/fneur.2019.00483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/23/2019] [Indexed: 12/25/2022] Open
Abstract
Antiepileptic drugs (AEDs) have repeatedly shown inconsistent and almost contradictory effects on the neurocognitive system, from substantial impairments in processing speed to the noticeable improvement in working memory and executive functioning. Previous studies have provided a novel insight into the cognitive improvement by bumetanide as a potential antiepileptic drug. Through the current investigation, we evaluated the longitudinal effects of bumetanide, an NKCC1 co-transporter antagonist, on the brain microstructural organization as a probable underlying component for cognitive performance. Microstructure assessment was completed using SPM for the whole brain assay and Freesurfer/TRACULA for the automatic probabilistic tractography analysis. Primary cognitive operations including selective attention and processing speed, working memory capacity and spatial memory were evaluated in 12 patients with a confirmed diagnosis of refractory epilepsy. Participants treated with bumetanide (2 mg/ day) in two divided doses as an adjuvant therapy to their regular AEDs for 6 months, which followed by the re-assessment of their cognitive functions and microstructural organizations. Seizure frequency reduced in eight patients which accompanied by white matter reconstruction; fractional anisotropy (FA) increased in the cingulum-cingulate gyrus (CCG), anterior thalamic radiation (ATR), and temporal part of the superior longitudinal fasciculus (SLFt) in correlation with the clinical response. The voxel-based analysis in responder patients revealed increased FA in the left hippocampus, right cerebellum, and right medial temporal lobe, while mean diffusivity (MD) values reduced in the right occipital lobe and cerebellum. Microstructural changes in SLFt and ATR accompanied by a reduction in the error rate in the spatial memory test. These primary results have provided preliminary evidence for the effect of bumetanide on cognitive functioning through microstructural changes in patients with drug-resistant epilepsy.
Collapse
Affiliation(s)
- Zeinab Gharaylou
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Lida Shafaghi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oghabian
- Neuroimaging and Analysis Group, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Yoonessi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Imam Khomeini Hospital, Iranian Center of Neurological Research, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahmoudreza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Kharod SC, Kang SK, Kadam SD. Off-Label Use of Bumetanide for Brain Disorders: An Overview. Front Neurosci 2019; 13:310. [PMID: 31068771 PMCID: PMC6491514 DOI: 10.3389/fnins.2019.00310] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/19/2019] [Indexed: 01/17/2023] Open
Abstract
Bumetanide (BTN or BUM) is a FDA-approved potent loop diuretic (LD) that acts by antagonizing sodium-potassium-chloride (Na-K-Cl) cotransporters, NKCC1 (SLc12a2) and NKCC2. While NKCC1 is expressed both in the CNS and in systemic organs, NKCC2 is kidney-specific. The off-label use of BTN to modulate neuronal transmembrane Cl− gradients by blocking NKCC1 in the CNS has now been tested as an anti-seizure agent and as an intervention for neurological disorders in pre-clinical studies with varying results. BTN safety and efficacy for its off-label use has also been tested in several clinical trials for neonates, children, adolescents, and adults. It failed to meet efficacy criteria for hypoxic-ischemic encephalopathy (HIE) neonatal seizures. In contrast, positive outcomes in temporal lobe epilepsy (TLE), autism, and schizophrenia trials have been attributed to BTN in studies evaluating its off-label use. NKCC1 is an electroneutral neuronal Cl− importer and the dominance of NKCC1 function has been proposed as the common pathology for HIE seizures, TLE, autism, and schizophrenia. Therefore, the use of BTN to antagonize neuronal NKCC1 with the goal to lower internal Cl− levels and promote GABAergic mediated hyperpolarization has been proposed. In this review, we summarize the data and results for pre-clinical and clinical studies that have tested off-label BTN interventions and report variable outcomes. We also compare the data underlying the developmental expression profile of NKCC1 and KCC2, highlight the limitations of BTN’s brain-availability and consider its actions on non-neuronal cells.
Collapse
Affiliation(s)
- Shivani C Kharod
- Neuroscience Laboratory, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
| | - Seok Kyu Kang
- Neuroscience Laboratory, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
| | - Shilpa D Kadam
- Neuroscience Laboratory, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States.,Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
21
|
Hsu Y, Chang Y, Liu Y, Wang K, Chen H, Lee D, Yang S, Tsai C, Lien C, Chern Y. Enhanced Na
+
‐K
+
‐2Cl
‐
cotransporter 1 underlies motor dysfunction in huntington's disease. Mov Disord 2019; 34:845-857. [DOI: 10.1002/mds.27651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 01/17/2023] Open
Affiliation(s)
- Yi‐Ting Hsu
- PhD Program for Translational MedicineChina Medical University and Academia Sinica Taipei Taiwan
- Department of NeurologyChina Medical University Hospital Taichung Taiwan
| | - Ya‐Gin Chang
- Institute of NeuroscienceNational Yang‐Ming University Taipei Taiwan
- Taiwan International Graduate Program in Interdisciplinary NeuroscienceNational Yang‐Ming University and Academia Sinica Taipei Taiwan
| | - Yu‐Chao Liu
- Institute of NeuroscienceNational Yang‐Ming University Taipei Taiwan
| | - Kai‐Yi Wang
- Institute of NeuroscienceNational Yang‐Ming University Taipei Taiwan
| | - Hui‐Mei Chen
- Institute of Biomedical SciencesAcademia Sinica Taipei Taiwan
| | - Ding‐Jin Lee
- PhD Program for Translational MedicineChina Medical University and Academia Sinica Taipei Taiwan
| | - Sung‐Sen Yang
- Division of Nephrology, Department of Medicine, Tri‐Service General HospitalNational Defense Medical Center Taipei Taiwan
| | - Chon‐Haw Tsai
- PhD Program for Translational MedicineChina Medical University and Academia Sinica Taipei Taiwan
- Department of NeurologyChina Medical University Hospital Taichung Taiwan
| | - Cheng‐Chang Lien
- Institute of NeuroscienceNational Yang‐Ming University Taipei Taiwan
- Brain Research CenterNational Yang‐Ming University Taipei Taiwan
| | - Yijuang Chern
- PhD Program for Translational MedicineChina Medical University and Academia Sinica Taipei Taiwan
- Institute of NeuroscienceNational Yang‐Ming University Taipei Taiwan
- Institute of Biomedical SciencesAcademia Sinica Taipei Taiwan
| |
Collapse
|
22
|
Vogel KR, Ainslie GR, Walters DC, McConnell A, Dhamne SC, Rotenberg A, Roullet JB, Gibson KM. Succinic semialdehyde dehydrogenase deficiency, a disorder of GABA metabolism: an update on pharmacological and enzyme-replacement therapeutic strategies. J Inherit Metab Dis 2018; 41:699-708. [PMID: 29460030 PMCID: PMC6041169 DOI: 10.1007/s10545-018-0153-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
Abstract
We present an update to the status of research on succinic semialdehyde dehydrogenase (SSADH) deficiency (SSADHD), a rare disorder of GABA metabolism. This is an unusual disorder featuring the accumulation of both GABA and its neuromodulatory analog, gamma-hydroxybutyric acid (GHB), and recent studies have advanced the potential clinical application of NCS-382, a putative GHB receptor antagonist. Animal studies have provided proof-of-concept that enzyme replacement therapy could represent a long-term therapeutic option. The characterization of neuronal stem cells (NSCs) derived from aldehyde dehydrogenase 5a1-/- (aldh5a1-/-) mice, the murine model of SSADHD, has highlighted NSC utility as an in vitro system in which to study therapeutics and associated toxicological properties. Gene expression analyses have revealed that transcripts encoding GABAA receptors are down-regulated and may remain largely immature in aldh5a1-/- brain, characterized by excitatory as opposed to inhibitory outputs, the latter being the expected action in the mature central nervous system. This indicates that agents altering chloride channel activity may be therapeutically relevant in SSADHD. The most recent therapeutic prospects include mTOR (mechanistic target of rapamycin) inhibitors, drugs that have received attention with the elucidation of the effects of elevated GABA on autophagy. The outlook for novel therapeutic trials in SSADHD continues to improve.
Collapse
Affiliation(s)
- Kara R Vogel
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | | | - Dana C Walters
- Department of Pharmacotherapy, College of Pharmacy, Washington State University, 412 E. Spokane Falls Blvd, Health Sciences Building Room 210, Spokane, WA, 99204, USA
| | | | - Sameer C Dhamne
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexander Rotenberg
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jean-Baptiste Roullet
- Department of Pharmacotherapy, College of Pharmacy, Washington State University, 412 E. Spokane Falls Blvd, Health Sciences Building Room 210, Spokane, WA, 99204, USA
| | - K Michael Gibson
- Department of Pharmacotherapy, College of Pharmacy, Washington State University, 412 E. Spokane Falls Blvd, Health Sciences Building Room 210, Spokane, WA, 99204, USA.
| |
Collapse
|
23
|
So HC, Chau CKL, Chiu WT, Ho KS, Lo CP, Yim SHY, Sham PC. Analysis of genome-wide association data highlights candidates for drug repositioning in psychiatry. Nat Neurosci 2017; 20:1342-1349. [PMID: 28805813 DOI: 10.1038/nn.4618] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Knowledge of psychiatric disease genetics has advanced rapidly during the past decade with the advent of genome-wide association studies (GWAS). However, less progress has been made in harnessing these data to reveal new therapies. Here we propose a framework for drug repositioning by comparing transcriptomes imputed from GWAS data with drug-induced gene expression profiles from the Connectivity Map database and apply this approach to seven psychiatric disorders. We found a number of repositioning candidates, many supported by preclinical or clinical evidence. Repositioning candidates for a number of disorders were also significantly enriched for known psychiatric medications or therapies considered in clinical trials. For example, candidates for schizophrenia were enriched for antipsychotics, while those for bipolar disorder were enriched for both antipsychotics and antidepressants. These findings provide support for the usefulness of GWAS data in guiding drug discovery.
Collapse
Affiliation(s)
- Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Zoology Institute of Zoology and The Chinese University of Hong Kong, China
| | - Carlos Kwan-Long Chau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wan-To Chiu
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Kin-Sang Ho
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Cho-Pong Lo
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | - Pak-Chung Sham
- Department of Psychiatry, University of Hong Kong, Pokfulam, Hong Kong, China.,Centre for Genomic Sciences, University of Hong Kong, Pokfulam, Hong Kong, China.,State Key Laboratory for Cognitive and Brain Sciences, University of Hong Kong, Pokfulam, Hong Kong, China.,Centre for Reproduction, Development and Growth, University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|