Evaluation of bumetanide as a potential therapeutic agent for Alzheimer’s disease

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.

[1]  Yingwei Wang,et al.  Spatiotemporal expression patterns of genes coding for plasmalemmal chloride transporters and channels in neurological diseases , 2023, Molecular Brain.

[2]  M. Gorospe,et al.  Hydroxychloroquine lowers Alzheimer’s disease and related dementias risk and rescues molecular phenotypes related to Alzheimer’s disease , 2022, Molecular Psychiatry.

[3]  Mari A. Virtanen,et al.  Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development , 2022, Cerebral cortex.

[4]  R. Bateman,et al.  Lecanemab in Early Alzheimer's Disease. , 2022, The New England journal of medicine.

[5]  William T. Ralvenius,et al.  APOE4 impairs myelination via cholesterol dysregulation in oligodendrocytes , 2022, Nature.

[6]  A. Saykin,et al.  Myelin repair in Alzheimer’s disease: a review of biological pathways and potential therapeutics , 2022, Translational Neurodegeneration.

[7]  McKenzie Prillaman Alzheimer’s drug slows mental decline in trial — but is it a breakthrough? , 2022, Nature.

[8]  A. Koyama,et al.  Population pharmacokinetic‐pharmacodynamic analyses of amyloid positron emission tomography and plasma biomarkers for lecanemab in subjects with early Alzheimer's disease , 2022, CPT: pharmacometrics & systems pharmacology.

[9]  R. Rissman,et al.  Dysregulation of Neuropeptide and Tau Peptide Signatures in Human Alzheimer’s Disease Brain , 2022, ACS chemical neuroscience.

[10]  L. Schneider,et al.  Making the Case for Accelerated Withdrawal of Aducanumab. , 2022, Journal of Alzheimer's disease : JAD.

[11]  R. Faull,et al.  Beta-Amyloid (Aβ1-42) Increases the Expression of NKCC1 in the Mouse Hippocampus , 2022, Molecules.

[12]  F. Barkhof,et al.  Two Randomized Phase 3 Studies of Aducanumab in Early Alzheimer’s Disease , 2022, The Journal of Prevention of Alzheimer's Disease.

[13]  A. Alia,et al.  Evaluation of suprachiasmatic nucleus in Alzheimer’s disease with non-invasive magnetic resonance methods , 2022, Neural regeneration research.

[14]  K. Kaila,et al.  The NKCC1 ion transporter modulates microglial phenotype and inflammatory response to brain injury in a cell-autonomous manner , 2022, PLoS biology.

[15]  Mina Esmail Zadeh Nojoo Kambar,et al.  Alzheimer's disease drug development pipeline: 2022 , 2022, Alzheimer's & dementia.

[16]  K. Kaila,et al.  CNS pharmacology of NKCC1 inhibitors , 2021, Neuropharmacology.

[17]  A. Sporbert,et al.  Microglia sense neuronal activity via GABA in the early postnatal hippocampus. , 2021, Cell reports.

[18]  Gui-hong Zhang,et al.  Microglia in Alzheimer’s Disease: A Target for Therapeutic Intervention , 2021, Frontiers in Cellular Neuroscience.

[19]  P. Scheltens,et al.  The Alzheimer’s disease drug development landscape , 2021, Alzheimer's Research & Therapy.

[20]  M. De Vivo,et al.  Pharmacological tools to target NKCC1 in brain disorders. , 2021, Trends in pharmacological sciences.

[21]  Fayzan F. Chaudhry,et al.  Experimental and real-world evidence supporting the computational repurposing of bumetanide for APOE4-related Alzheimer’s disease , 2021, Nature Aging.

[22]  P. Tariot,et al.  The case for low-level BACE1 inhibition for the prevention of Alzheimer disease , 2021, Nature Reviews Neurology.

[23]  A. Atri,et al.  Aducanumab: Appropriate Use Recommendations , 2021, The Journal of Prevention of Alzheimer's Disease.

[24]  W. Löscher,et al.  A combination of phenobarbital and the bumetanide derivative bumepamine prevents neonatal seizures and subsequent hippocampal neurodegeneration in a rat model of birth asphyxia , 2021, Epilepsia.

[25]  L. Ferrucci,et al.  Effects of monoclonal antibodies against amyloid-β on clinical and biomarker outcomes and adverse event risks: A systematic review and meta-analysis of phase III RCTs in Alzheimer’s disease , 2021, Ageing Research Reviews.

[26]  M. Mintun,et al.  Donanemab in Early Alzheimer's Disease. , 2021, The New England journal of medicine.

[27]  M. Kivimäki,et al.  The association of APOE ε4 with cognitive function over the adult life course and incidence of dementia: 20 years follow-up of the Whitehall II study , 2021, Alzheimer's research & therapy.

[28]  B. Hyman,et al.  APOE and Alzheimer’s Disease: Advances in Genetics, Pathophysiology, and Therapeutic Approaches. , 2021, The Lancet Neurology.

[29]  G. Perea,et al.  GABAergic signaling to astrocytes in the prefrontal cortex sustains goal-directed behaviors , 2020, Nature Neuroscience.

[30]  W. Löscher,et al.  The search for brain-permeant NKCC1 inhibitors for the treatment of seizures: Pharmacokinetic-pharmacodynamic modelling of NKCC1 inhibition by azosemide, torasemide, and bumetanide in mouse brain , 2020, Epilepsy & Behavior.

[31]  Mari A. Virtanen,et al.  NKCC1, an Elusive Molecular Target in Brain Development: Making Sense of the Existing Data , 2020, Cells.

[32]  W. Löscher,et al.  Phenobarbital and midazolam suppress neonatal seizures in a noninvasive rat model of birth asphyxia, whereas bumetanide is ineffective , 2020, Epilepsia.

[33]  Shanshan Song,et al.  Ion channels and transporters in microglial function in physiology and brain diseases , 2020, Neurochemistry International.

[34]  W. Löscher,et al.  Hydrolytic biotransformation of the bumetanide ester prodrug DIMAEB to bumetanide by esterases in neonatal human and rat serum and neonatal rat brain—A new treatment strategy for neonatal seizures? , 2020, Epilepsia.

[35]  S. R. Datta,et al.  GABA-receptive microglia selectively sculpt developing inhibitory circuits , 2020, Cell.

[36]  Punam Sawant-Pokam,et al.  Preventing neuronal edema increases network excitability after traumatic brain injury. , 2020, The Journal of clinical investigation.

[37]  N. Zuithoff,et al.  Bumetanide for Core Symptoms of Autism Spectrum Disorder (BAMBI): A Single Center, Double-Blinded, Participant-Randomized, Placebo-Controlled, Phase Two, Superiority Trial. , 2020, Journal of the American Academy of Child and Adolescent Psychiatry.

[38]  R. Bertorelli,et al.  Discovery of a Small Molecule Drug Candidate for Selective NKCC1 Inhibition in Brain Disorders , 2020, Chem.

[39]  M. Theus,et al.  Mechanisms of Blood–Brain Barrier Dysfunction in Traumatic Brain Injury , 2020, International journal of molecular sciences.

[40]  O. Forlenza,et al.  Passive antiamyloid immunotherapy for Alzheimer's disease. , 2020, Current opinion in psychiatry.

[41]  T. Al‐Tel,et al.  BACE1 inhibitors: Current status and future directions in treating Alzheimer's disease , 2020, Medicinal research reviews.

[42]  G. Bu,et al.  Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies , 2019, Nature Reviews Neurology.

[43]  S. Kadam,et al.  Off-Label Use of Bumetanide for Brain Disorders: An Overview , 2019, Front. Neurosci..

[44]  B. Molyneaux,et al.  A Novel Na+-K+-Cl− Cotransporter 1 Inhibitor STS66* Reduces Brain Damage in Mice After Ischemic Stroke , 2019, Stroke.

[45]  D. Michaelson,et al.  ApoE4: an emerging therapeutic target for Alzheimer’s disease , 2019, BMC Medicine.

[46]  M. Bozzali,et al.  Ventral tegmental area disruption in Alzheimer’s disease , 2019, Aging.

[47]  O. Fiehn,et al.  Metabolomics Analyses of 14 Classical Neurotransmitters by GC-TOF with LC-MS Illustrates Secretion of 9 Cell-Cell Signaling Molecules from Sympathoadrenal Chromaffin Cells in the Presence of Lithium. , 2019, ACS chemical neuroscience.

[48]  F. Colbourne,et al.  Failure of bumetanide to improve outcome after intracerebral hemorrhage in rat , 2019, PloS one.

[49]  W. Löscher,et al.  Bumepamine, a brain-permeant benzylamine derivative of bumetanide, does not inhibit NKCC1 but is more potent to enhance phenobarbital's anti-seizure efficacy , 2018, Neuropharmacology.

[50]  Jason H. Huang,et al.  Neuroinflammation and blood–brain barrier disruption following traumatic brain injury: Pathophysiology and potential therapeutic targets , 2018, Journal of neuroscience research.

[51]  E. Sigurdsson,et al.  Tau-targeting therapies for Alzheimer disease , 2018, Nature Reviews Neurology.

[52]  K. Lenz,et al.  Microglia and Beyond: Innate Immune Cells As Regulators of Brain Development and Behavioral Function , 2018, Front. Immunol..

[53]  M. Xie,et al.  NKCC1 Inhibition Attenuates Chronic Cerebral Hypoperfusion-Induced White Matter Lesions by Enhancing Progenitor Cells of Oligodendrocyte Proliferation , 2018, Journal of Molecular Neuroscience.

[54]  Nicole R. Zürcher,et al.  Bumetanide for autism: more eye contact, less amygdala activation , 2018, Scientific Reports.

[55]  M. Joghataei,et al.  Effect of bumetanide, a selective NKCC1 inhibitor, on hallucinations of schizophrenic patients; a double-blind randomized clinical trial , 2017, Schizophrenia Research.

[56]  Jun Chen,et al.  Inhibition of Na+-K+-2Cl− cotransporter attenuates blood-brain-barrier disruption in a mouse model of traumatic brain injury , 2017, Neurochemistry International.

[57]  W. Löscher,et al.  Multiple blood-brain barrier transport mechanisms limit bumetanide accumulation, and therapeutic potential, in the mammalian brain , 2017, Neuropharmacology.

[58]  Kai Gu,et al.  Gamma secretase inhibitors: a patent review (2013 - 2015) , 2017, Expert opinion on therapeutic patents.

[59]  I. Dewachter,et al.  Cortical cells reveal APP as a new player in the regulation of GABAergic neurotransmission , 2017, Scientific Reports.

[60]  Y. Ben-Ari,et al.  Effects of bumetanide on neurobehavioral function in children and adolescents with autism spectrum disorders , 2017, Translational Psychiatry.

[61]  M. Joghataei,et al.  Lack of the effect of bumetanide, a selective NKCC1 inhibitor, in patients with schizophrenia: A double‐blind randomized trial , 2017, Psychiatry and clinical neurosciences.

[62]  G. Alves,et al.  Neuroimaging Findings Related to Behavioral Disturbances in Alzheimer's Disease: A Systematic Review. , 2016, Current Alzheimer research.

[63]  N. Jovanov-Milošević,et al.  Developmental Expression Patterns of KCC2 and Functionally Associated Molecules in the Human Brain. , 2016, Cerebral cortex.

[64]  D. Holtzman,et al.  Mechanisms linking circadian clocks, sleep, and neurodegeneration , 2016, Science.

[65]  M. Sur,et al.  Jointly reduced inhibition and excitation underlies circuit-wide changes in cortical processing in Rett syndrome , 2016, Proceedings of the National Academy of Sciences.

[66]  R. Salvi,et al.  Ototoxic effects and mechanisms of loop diuretics , 2016, Journal of otology.

[67]  M. Nedergaard,et al.  NKCC1 up-regulation contributes to early post-traumatic seizures and increased post-traumatic seizure susceptibility , 2016, Brain Structure and Function.

[68]  M. Bozzali,et al.  Quantitative MRI to understand Alzheimer's disease pathophysiology. , 2016, Current opinion in neurology.

[69]  W. Löscher,et al.  The bumetanide prodrug BUM5, but not bumetanide, potentiates the antiseizure effect of phenobarbital in adult epileptic mice , 2016, Epilepsia.

[70]  Z. Fei,et al.  Inhibition of Na+-K+-2Cl- Cotransporter-1 attenuates traumatic brain injury-induced neuronal apoptosis via regulation of Erk signaling , 2016, Neurochemistry International.

[71]  A. Pérez-Samartín,et al.  Axon-to-Glia Interaction Regulates GABAA Receptor Expression in Oligodendrocytes , 2016, Molecular Pharmacology.

[72]  N. Bandeira,et al.  Neuropeptidomics Mass Spectrometry Reveals Signaling Networks Generated by Distinct Protease Pathways in Human Systems , 2015, Journal of The American Society for Mass Spectrometry.

[73]  B. Wang,et al.  A Pilot Study on the Combination of Applied Behavior Analysis and Bumetanide Treatment for Children with Autism. , 2015, Journal of child and adolescent psychopharmacology.

[74]  M. Chonchol,et al.  Renal control of calcium, phosphate, and magnesium homeostasis. , 2015, Clinical journal of the American Society of Nephrology : CJASN.

[75]  B. Griffin,et al.  The effect of organic anion transporter 3 inhibitor probenecid on bumetanide levels in the brain: an integrated in vivo microdialysis study in the rat , 2015, The Journal of pharmacy and pharmacology.

[76]  C. Zhao,et al.  In vivo effects of bumetanide at brain concentrations incompatible with NKCC1 inhibition on newborn DGC structure and spontaneous EEG seizures following hypoxia-induced neonatal seizures , 2015, Neuroscience.

[77]  W. Löscher,et al.  The organic anion transport inhibitor probenecid increases brain concentrations of the NKCC1 inhibitor bumetanide. , 2015, European journal of pharmacology.

[78]  D. Mount Thick ascending limb of the loop of Henle. , 2014, Clinical journal of the American Society of Nephrology : CJASN.

[79]  Basavaraj Hooli,et al.  A three-dimensional human neural cell culture model of Alzheimer’s disease , 2014, Nature.

[80]  J. A. Payne,et al.  Cation-chloride cotransporters in neuronal development, plasticity and disease , 2014, Nature Reviews Neuroscience.

[81]  K. Kaila,et al.  Pharmacotherapeutic targeting of cation-chloride cotransporters in neonatal seizures , 2014, Epilepsia.

[82]  W. Löscher,et al.  A novel prodrug‐based strategy to increase effects of bumetanide in epilepsy , 2014, Annals of neurology.

[83]  Amene Shahrokhi,et al.  Oxytocin-Mediated GABA Inhibition During Delivery Attenuates Autism Pathogenesis in Rodent Offspring , 2014, Science.

[84]  W. Löscher,et al.  Consequences of inhibition of bumetanide metabolism in rodents on brain penetration and effects of bumetanide in chronic models of epilepsy , 2014, The European journal of neuroscience.

[85]  조현철,et al.  Intracerebral Hemorrhage(ICH) 수술 유도 로봇 시스템을 위한 수술 유도 도구 켈리브레이션 , 2014 .

[86]  F. Jensen,et al.  Bumetanide Enhances Phenobarbital Efficacy in a Rat Model of Hypoxic Neonatal Seizures , 2013, PloS one.

[87]  Huaxi Xu,et al.  Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy , 2013, Nature Reviews Neurology.

[88]  M. Hadjighassem,et al.  Bumetanide reduces seizure frequency in patients with temporal lobe epilepsy , 2013, Epilepsia.

[89]  Y. Ben-Ari,et al.  A randomised controlled trial of bumetanide in the treatment of autism in children , 2012, Translational Psychiatry.

[90]  Lydia Ng,et al.  Allen Brain Atlas: an integrated spatio-temporal portal for exploring the central nervous system , 2012, Nucleic Acids Res..

[91]  Junsung Woo,et al.  Astrocytes as GABA-ergic and GABA-ceptive Cells , 2012, Neurochemical Research.

[92]  E. Castrén,et al.  The Loop Diuretic Bumetanide Blocks Posttraumatic p75NTR Upregulation and Rescues Injured Neurons , 2012, The Journal of Neuroscience.

[93]  H. Arrighi,et al.  Prevalence of Apolipoprotein E4 Genotype and Homozygotes (APOE e4/4) among Patients Diagnosed with Alzheimer’s Disease: A Systematic Review and Meta-Analysis , 2011, Neuroepidemiology.

[94]  M. Sabbagh,et al.  New Acetylcholinesterase Inhibitors for Alzheimer's Disease , 2011, International journal of Alzheimer's disease.

[95]  P. Ortiz,et al.  Molecular regulation of NKCC2 in the thick ascending limb. , 2011, American journal of physiology. Renal physiology.

[96]  J. Soul,et al.  Sensitive isotope dilution liquid chromatography/tandem mass spectrometry method for quantitative analysis of bumetanide in serum and brain tissue. , 2011, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[97]  Y. Ben-Ari,et al.  The diuretic bumetanide decreases autistic behaviour in five infants treated during 3 months with no side effects , 2010, Acta paediatrica.

[98]  J. Simard,et al.  Molecular mechanisms of microvascular failure in central nervous system injury--synergistic roles of NKCC1 and SUR1/TRPM4. , 2010, Journal of neurosurgery.

[99]  O. Witte,et al.  Downregulation of Potassium Chloride Cotransporter KCC2 After Transient Focal Cerebral Ischemia , 2010, Stroke.

[100]  K. Staley,et al.  Decreased Seizure Activity in a Human Neonate Treated With Bumetanide, an Inhibitor of the Na+-K+-2Cl- Cotransporter NKCC1 , 2009, Journal of child neurology.

[101]  M. Norenberg,et al.  Na-K-Cl Cotransporter-1 in the Mechanism of Ammonia-induced Astrocyte Swelling* , 2008, Journal of Biological Chemistry.

[102]  G. Buzsáki,et al.  Inhibition and Brain Work , 2007, Neuron.

[103]  R. Miles,et al.  Perturbed Chloride Homeostasis and GABAergic Signaling in Human Temporal Lobe Epilepsy , 2007, The Journal of Neuroscience.

[104]  D. Kintner,et al.  AMPA‐mediated excitotoxicity in oligodendrocytes: role for Na+–K+–Cl− co‐transport and reversal of Na+/Ca2+ exchanger , 2007, Journal of neurochemistry.

[105]  K. Lu,et al.  Bumetanide administration attenuated traumatic brain injury through IL-1 overexpression , 2007, Neurological research.

[106]  K. Lu,et al.  Inhibition of the Na+ -K+ -2Cl- -cotransporter in choroid plexus attenuates traumatic brain injury-induced brain edema and neuronal damage. , 2006, European journal of pharmacology.

[107]  J. Birks,et al.  Cholinesterase inhibitors for Alzheimer's disease. , 2006 .

[108]  M. O’Donnell,et al.  Moderate-to-severe ischemic conditions increase activity and phosphorylation of the cerebral microvascular endothelial cell Na+-K+-Cl- cotransporter. , 2005, American journal of physiology. Cell physiology.

[109]  D. Kintner,et al.  Na-K-Cl Cotransporter-Mediated Intracellular Na+ Accumulation Affects Ca2+ Signaling in Astrocytes in an In Vitro Ischemic Model , 2004, The Journal of Neuroscience.

[110]  Lien Tran,et al.  Bumetanide Inhibition of the Blood-Brain Barrier Na-K-Cl Cotransporter Reduces Edema Formation in the Rat Middle Cerebral Artery Occlusion Model of Stroke , 2004, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[111]  P. Barthó,et al.  Differential distribution of the KCl cotransporter KCC2 in thalamic relay and reticular nuclei , 2004, The European journal of neuroscience.

[112]  J. Tepper,et al.  Cell Type-Specific Differences in Chloride-Regulatory Mechanisms and GABAA Receptor-Mediated Inhibition in Rat Substantia Nigra , 2003, The Journal of Neuroscience.

[113]  Hao Wang,et al.  GABA-mediated trophic effect on oligodendrocytes requires Na-K-2Cl cotransport activity. , 2003, Journal of neurophysiology.

[114]  Juha Voipio,et al.  Cation–chloride co-transporters in neuronal communication, development and trauma , 2003, Trends in Neurosciences.

[115]  J. Voipio,et al.  BDNF-induced TrkB activation down-regulates the K+–Cl− cotransporter KCC2 and impairs neuronal Cl− extrusion , 2002, The Journal of cell biology.

[116]  Y. Ben-Ari Excitatory actions of gaba during development: the nature of the nurture , 2002, Nature Reviews Neuroscience.

[117]  D. Kintner,et al.  Astrocytes from Na(+)-K(+)-Cl(-) cotransporter-null mice exhibit absence of swelling and decrease in EAA release. , 2002, American journal of physiology. Cell physiology.

[118]  W. Hauser,et al.  Are certain diuretics also anticonvulsants? , 2001, Annals of neurology.

[119]  J. A. Payne,et al.  The Neuron-specific K-Cl Cotransporter, KCC2 , 1999, The Journal of Biological Chemistry.

[120]  J. A. Payne,et al.  The K+/Cl− co-transporter KCC2 renders GABA hyperpolarizing during neuronal maturation , 1999, Nature.

[121]  J. Mellor,et al.  Properties of GABAA receptors in cultured rat oligodendrocyte progenitor cells , 1998, Neuropharmacology.

[122]  S. Hebert,et al.  Ionic conductance pathways in the mouse medullary thick ascending limb of Henle. The paracellular pathway and electrogenic Cl- absorption , 1986, The Journal of general physiology.

[123]  W. Colburn,et al.  Pharmacokinetics of bumetanide following intravenous, intramuscular, and oral administrations to normal subjects. , 1984, Journal of pharmaceutical sciences.

[124]  R. D. Brown Effect of bumetanide on the positive endocochlear dc potential of the cat. , 1976, Toxicology and applied pharmacology.

[125]  R. Vergona,et al.  A comparative diuretic and tissue distribution study of bumetanide and furosemide in the dog. , 1976, The Journal of pharmacology and experimental therapeutics.

[126]  M. Burg Tubular chloride transport and the mode of action of some diuretics. , 1976, Kidney international.

[127]  A. J. Duvall,et al.  Early changes in the cochlear duct from ethacrynic ACID: An electronmicroscopic evaluation , 1970, The Laryngoscope.

[128]  K. Blennow,et al.  Anti-Tau Trials for Alzheimer’s Disease: A Report from the EU/US/CTAD Task Force , 2019, The Journal of Prevention of Alzheimer's Disease.

[129]  C. Schwarzer,et al.  Functional characterization of novel bumetanide derivatives for epilepsy treatment , 2019, Neuropharmacology.

[130]  J. Meijer,et al.  Degeneration of the Suprachiasmatic Nucleus in an Alzheimer's Disease Mouse Model Monitored by in vivo Magnetic Resonance Relaxation Measurements and Immunohistochemistry. , 2019, Journal of Alzheimer's disease : JAD.

[131]  Ghulam Md Ashraf,et al.  Cholinesterase Inhibitors for Alzheimer Disease: Multitargeting Strategy based on Anti-Alzheimer's Drugs Repositioning. , 2019, Current pharmaceutical design.

[132]  R. Klein,et al.  Prevalence of ototoxic medication use among older adults in Beaver Dam, Wisconsin , 2018, Journal of the American Association of Nurse Practitioners.

[133]  T. Ikuta,et al.  Memantine for Alzheimer's Disease: An Updated Systematic Review and Meta-analysis. , 2017, Journal of Alzheimer's disease : JAD.

[134]  P. Damier,et al.  Bumetanide to Treat Parkinson Disease: A Report of 4 Cases. , 2016, Clinical neuropharmacology.

[135]  R. Tanzi,et al.  Recapitulating amyloid β and tau pathology in human neural cell culture models: clinical implications. , 2015, US neurology.

[136]  D. Price,et al.  Basic neurochemistry : principles of molecular, cellular and medical neurobiology , 2012 .

[137]  D. M. Robinson,et al.  Memantine: a review of its use in Alzheimer's disease. , 2006, Drugs.

[138]  Wen‐Hui Wang Regulation of ROMK (Kir1.1) channels: new mechanisms and aspects. , 2006, American journal of physiology. Renal physiology.

[139]  P. A. Friedman,et al.  Effects of antidiuretic hormone on cellular conductive pathways in mouse medullary thick ascending limbs of Henle: I. ADH increases transcellular conductance pathways , 2005, The Journal of Membrane Biology.

[140]  G. Giebisch Diuretic action of potassium channel blockers , 2005, European Journal of Clinical Pharmacology.

[141]  S. Hebert,et al.  Effects of antidiuretic hormone on cellular conductive pathways in mouse medullary thick ascending limbs of Henle: II. Determinants of the ADH-mediated increases in transepithelial voltage and in net Cl− absorption , 2005, The Journal of Membrane Biology.

[142]  J. Russell Sodium-potassium-chloride cotransport. , 2000, Physiological reviews.