1
|
Klein P, Kaminski RM, Koepp M, Löscher W. New epilepsy therapies in development. Nat Rev Drug Discov 2024; 23:682-708. [PMID: 39039153 DOI: 10.1038/s41573-024-00981-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2024] [Indexed: 07/24/2024]
Abstract
Epilepsy is a common brain disorder, characterized by spontaneous recurrent seizures, with associated neuropsychiatric and cognitive comorbidities and increased mortality. Although people at risk can often be identified, interventions to prevent the development of the disorder are not available. Moreover, in at least 30% of patients, epilepsy cannot be controlled by current antiseizure medications (ASMs). As a result of considerable progress in epilepsy genetics and the development of novel disease models, drug screening technologies and innovative therapeutic modalities over the past 10 years, more than 200 novel epilepsy therapies are currently in the preclinical or clinical pipeline, including many treatments that act by new mechanisms. Assisted by diagnostic and predictive biomarkers, the treatment of epilepsy is undergoing paradigm shifts from symptom-only ASMs to disease prevention, and from broad trial-and-error treatments for seizures in general to mechanism-based treatments for specific epilepsy syndromes. In this Review, we assess recent progress in ASM development and outline future directions for the development of new therapies for the treatment and prevention of epilepsy.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA.
| | | | - Matthias Koepp
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Wolfgang Löscher
- Translational Neuropharmacology Lab., NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
2
|
Pol E, Côme E, Merlaud Z, Gouhier J, Russeau M, Scotto-Lomassese S, Moutkine I, Marques X, Lévi S. NKCC1 and KCC2 Chloride Transporters Have Different Membrane Dynamics on the Surface of Hippocampal Neurons. Cells 2023; 12:2363. [PMID: 37830575 PMCID: PMC10571912 DOI: 10.3390/cells12192363] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
Na-K-2Cl cotransporter 1 (NKCC1) regulates chloride influx in neurons and thereby GABAA receptor activity in normal and pathological conditions. Here, we characterized in hippocampal neurons the membrane expression, distribution and dynamics of exogenous NKCC1a and NKCC1b isoforms and compared them to those of the chloride extruder K-Cl cotransporter 2 (KCC2). We found that NKCC1a and NKCC1b behave quite similarly. NKCC1a/1b but not KCC2 are present along the axon initial segment where they are confined. Moreover, NKCC1a/1b are detected in the somato-dendritic compartment at a lower level than KCC2, where they form fewer, smaller and less compact clusters at perisynaptic and extrasynaptic sites. Interestingly, ~60% of dendritic clusters of NKCC1a/1b are colocalized with KCC2. They are larger and brighter than those devoid of KCC2, suggesting a particular NKCC1a/1b-KCC2 relationship. In agreement with the reduced dendritic clustering of NKCC1a/1b compared with that of KCC2, NKCC1a/1b are more mobile on the dendrite than KCC2, suggesting weaker cytoskeletal interaction. NKCC1a/b are confined to endocytic zones, where they spend more time than KCC2. However, they spend less time in these compartments than at the synapses, suggesting that they can rapidly leave endocytic zones to increase the membrane pool, which can happen in pathological conditions. Thus, NKCC1a/b have different membrane dynamics and clustering from KCC2, which helps to explain their low level in the neuronal membrane, while allowing a rapid increase in the membrane pool under pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Sabine Lévi
- Institut du Fer à Moulin, Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR-S 1270, Sorbonne Université, 75005 Paris, France; (E.P.); (E.C.); (Z.M.); (J.G.); (M.R.); (S.S.-L.); (I.M.); (X.M.)
| |
Collapse
|
3
|
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: 12.0] [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
|
4
|
Kurki SN, Uvarov P, Pospelov AS, Trontti K, Hübner AK, Srinivasan R, Watanabe M, Hovatta I, Hübner CA, Kaila K, Virtanen MA. Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development. Cereb Cortex 2022; 33:5906-5923. [PMID: 36573432 PMCID: PMC10183754 DOI: 10.1093/cercor/bhac470] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
The Na-K-2Cl cotransporter NKCC1 is widely expressed in cells within and outside the brain. However, our understanding of its roles in brain functions throughout development, as well as in neuropsychiatric and neurological disorders, has been severely hindered by the lack of reliable data on its developmental and (sub)cellular expression patterns. We provide here the first properly controlled analysis of NKCC1 protein expression in various cell types of the mouse brain using custom-made antibodies and an NKCC1 knock-out validated immunohistochemical procedure, with parallel data based on advanced mRNA approaches. NKCC1 protein and mRNA are expressed at remarkably high levels in oligodendrocytes. In immature neurons, NKCC1 protein was located in the somata, whereas in adult neurons, only NKCC1 mRNA could be clearly detected. NKCC1 immunoreactivity is also seen in microglia, astrocytes, developing pericytes, and in progenitor cells of the dentate gyrus. Finally, a differential expression of NKCC1 splice variants was observed, with NKCC1a predominating in non-neuronal cells and NKCC1b in neurons. Taken together, our data provide a cellular basis for understanding NKCC1 functions in the brain and enable the identification of major limitations and promises in the development of neuron-targeting NKCC1-blockers.
Collapse
Affiliation(s)
- Samu N Kurki
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Pavel Uvarov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Alexey S Pospelov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Kalevi Trontti
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Antje K Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Rakenduvadhana Srinivasan
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Masahiko Watanabe
- Hokkaido University Department of Anatomy, Faculty of Medicine, , Sapporo 060–8638 , Japan
| | - Iiris Hovatta
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Christian A Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Kai Kaila
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Mari A Virtanen
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| |
Collapse
|
5
|
NKCC1 Deficiency in Forming Hippocampal Circuits Triggers Neurodevelopmental Disorder: Role of BDNF-TrkB Signalling. Brain Sci 2022; 12:brainsci12040502. [PMID: 35448033 PMCID: PMC9030861 DOI: 10.3390/brainsci12040502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 12/10/2022] Open
Abstract
The time-sensitive GABA shift from excitatory to inhibitory is critical in early neural circuits development and depends upon developmentally regulated expression of cation-chloride cotransporters NKCC1 and KCC2. NKCC1, encoded by the SLC12A2 gene, regulates neuronal Cl− homeostasis by chloride import working opposite KCC2. The high NKCC1/KCC2 expression ratio decreases in early neural development contributing to GABA shift. Human SLC12A2 loss-of-function mutations were recently associated with a multisystem disorder affecting neural development. However, the multisystem phenotype of rodent Nkcc1 knockout models makes neurodevelopment challenging to study. Brain-Derived Neurotrophic Factor (BDNF)-NTRK2/TrkB signalling controls KCC2 expression during neural development, but its impact on NKCC1 is still controversial. Here, we discuss recent evidence supporting BDNF-TrkB signalling controlling Nkcc1 expression and the GABA shift during hippocampal circuit formation. Namely, specific deletion of Ntrk2/Trkb from immature mouse hippocampal dentate granule cells (DGCs) affects their integration and maturation in the hippocampal circuitry and reduces Nkcc1 expression in their target region, the CA3 principal cells, leading to premature GABA shift, ultimately influencing the establishment of functional hippocampal circuitry and animal behaviour in adulthood. Thus, immature DGCs emerge as a potential therapeutic target as GABAergic transmission is vital for specific neural progenitors generating dentate neurogenesis in early development and the mature brain.
Collapse
|
6
|
Löscher W, Kaila K. CNS pharmacology of NKCC1 inhibitors. Neuropharmacology 2021; 205:108910. [PMID: 34883135 DOI: 10.1016/j.neuropharm.2021.108910] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022]
Abstract
The Na-K-2Cl cotransporter NKCC1 and the neuron-specific K-Cl cotransporter KCC2 are considered attractive CNS drug targets because altered neuronal chloride regulation and consequent effects on GABAergic signaling have been implicated in numerous CNS disorders. While KCC2 modulators are not yet clinically available, the loop diuretic bumetanide has been used off-label in attempts to treat brain disorders and as a tool for NKCC1 inhibition in preclinical models. Bumetanide is known to have anticonvulsant and neuroprotective effects under some pathophysiological conditions. However, as shown in several species from neonates to adults (mice, rats, dogs, and by extrapolation in humans), at the low clinical doses of bumetanide approved for diuresis, this drug has negligible access into the CNS, reaching levels that are much lower than what is needed to inhibit NKCC1 in cells within the brain parenchyma. Several drug discovery strategies have been initiated over the last ∼15 years to develop brain-permeant compounds that, ideally, should be selective for NKCC1 to eliminate the diuresis mediated by inhibition of renal NKCC2. The strategies employed to improve the pharmacokinetic and pharmacodynamic properties of NKCC1 blockers include evaluation of other clinically approved loop diuretics; development of lipophilic prodrugs of bumetanide; development of side-chain derivatives of bumetanide; and unbiased high-throughput screening approaches of drug discovery based on large chemical compound libraries. The main outcomes are that (1), non-acidic loop diuretics such as azosemide and torasemide may have advantages as NKCC1 inhibitors vs. bumetanide; (2), bumetanide prodrugs lead to significantly higher brain levels than the parent drug and have lower diuretic activity; (3), the novel bumetanide side-chain derivatives do not exhibit any functionally relevant improvement of CNS accessibility or NKCC1 selectivity vs. bumetanide; (4) novel compounds discovered by high-throughput screening may resolve some of the inherent problems of bumetanide, but as yet this has not been achieved. Thus, further research is needed to optimize the design of brain-permeant NKCC1 inhibitors. In parallel, a major challenge is to identify the mechanisms whereby various NKCC1-expressing cellular targets of these drugs within (e.g., neurons, oligodendrocytes or astrocytes) and outside the brain parenchyma (e.g., the blood-brain barrier, the choroid plexus, and the endocrine system), as well as molecular off-target effects, might contribute to their reported therapeutic and adverse effects.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany.
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Finland
| |
Collapse
|
7
|
Influence of Trace Elements on Neurodegenerative Diseases of The Eye-The Glaucoma Model. Int J Mol Sci 2021; 22:ijms22094323. [PMID: 33919241 PMCID: PMC8122456 DOI: 10.3390/ijms22094323] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is a heterogeneous group of chronic neurodegenerative disorders characterized by a relatively selective, progressive damage to the retinal ganglion cells (RGCs) and their axons, which leads to axon loss and visual field alterations. To date, many studies have shown the role of various elements, mainly metals, in maintaining the balance of prooxidative and antioxidative processes, regulation of fluid and ion flow through cell membranes of the ocular tissues. Based on the earlier and current research results, their relationship with the development and progression of glaucoma seems obvious and is increasingly appreciated. In this review, we aimed to summarize the current evidence on the role of trace elements in the pathogenesis and prevention of glaucomatous diseases. Special attention is also paid to the genetic background associated with glaucoma-related abnormalities of physiological processes that regulate or involve the ions of elements considered as trace elements necessary for the functioning of the cells.
Collapse
|
8
|
Virtanen MA, Uvarov P, Hübner CA, Kaila K. NKCC1, an Elusive Molecular Target in Brain Development: Making Sense of the Existing Data. Cells 2020; 9:cells9122607. [PMID: 33291778 PMCID: PMC7761970 DOI: 10.3390/cells9122607] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
Ionotropic GABA transmission is mediated by anion (mainly Cl−)-permeable GABAA receptors (GABAARs). In immature neurons, GABA exerts depolarizing and sometimes functionally excitatory actions, based on active uptake of Cl− by the Na-K-2Cl cotransporter NKCC1. While functional evidence firmly shows NKCC1-mediated ion transport in immature and diseased neurons, molecular detection of NKCC1 in the brain has turned out to be extremely difficult. In this review, we describe the highly inconsistent data that are available on the cell type-specific expression patterns of the NKCC1 mRNA and protein in the CNS. We discuss the major technical caveats, including a lack of knock-out-controlled immunohistochemistry in the forebrain, possible effects of alternative splicing on the binding of antibodies and RNA probes, and the wide expression of NKCC1 in different cell types, which make whole-tissue analyses of NKCC1 useless for studying its neuronal expression. We also review novel single-cell RNAseq data showing that most of the NKCC1 in the adult CNS may, in fact, be expressed in non-neuronal cells, especially in glia. As future directions, we suggest single-cell NKCC1 mRNA and protein analyses and the use of genetically tagged endogenous proteins or systematically designed novel antibodies, together with proper knock-out controls, for the visualization of endogenous NKCC1 in distinct brain cell types and their subcellular compartments.
Collapse
Affiliation(s)
- Mari A. Virtanen
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland; (M.A.V.); (P.U.)
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Pavel Uvarov
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland; (M.A.V.); (P.U.)
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Christian A. Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany;
| | - Kai Kaila
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland; (M.A.V.); (P.U.)
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-407256759
| |
Collapse
|
9
|
Garneau AP, Slimani S, Fiola MJ, Tremblay LE, Isenring P. Multiple Facets and Roles of Na+-K+-Cl−Cotransport: Mechanisms and Therapeutic Implications. Physiology (Bethesda) 2020; 35:415-429. [DOI: 10.1152/physiol.00012.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The Na+-K+-Cl−cotransporters play key physiological and pathophysiological roles by regulating the membrane potential of many cell types and the movement of fluid across a variety of epithelial or endothelial structures. As such, they should soon become invaluable targets for the treatment of various disorders including pain, epilepsy, brain edema, and hypertension. This review highlights the nature of these roles, the mechanisms at play, and the unresolved issues in the field.
Collapse
Affiliation(s)
- A. P. Garneau
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
- Cardiometabolic Axis, School of Kinesiology and Physical Activity Sciences, University of Montréal, Montréal, Canada
| | - S. Slimani
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| | - M. J. Fiola
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| | - L. E. Tremblay
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| | - P. Isenring
- Department of Medicine, Nephrology Research Group, Laval University, Québec, Canada; and
| |
Collapse
|
10
|
Merli AM, Vieujean S, Massot C, Blétard N, Quesada Calvo F, Baiwir D, Mazzucchelli G, Servais L, Wéra O, Oury C, de Leval L, Sempoux C, Manzini R, Bluemel S, Scharl M, Rogler G, De Pauw E, Coimbra Marques C, Colard A, Vijverman A, Delvenne P, Louis E, Meuwis MA. Solute carrier family 12 member 2 as a proteomic and histological biomarker of dysplasia and neoplasia in ulcerative colitis. J Crohns Colitis 2020; 15:jjaa168. [PMID: 32920643 DOI: 10.1093/ecco-jcc/jjaa168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Ulcerative colitis (UC) patients have a greater risk of developing colorectal cancer through inflammation-dysplasia-carcinoma sequence of transformation. The histopathological diagnosis of dysplasia is therefore of critical clinical relevance, but dysplasia may be difficult to distinguish from inflammatory changes. METHODS A proteomic pilot study on 5 UC colorectal dysplastic patients highlighted proteins differentially distributed between paired dysplastic, inflammatory and normal tissues. The best candidate marker was selected and immunohistochemistry confirmation was performed on AOM/DSS mouse model lesions, 37 UC dysplasia, 14 UC cancers, 23 longstanding UC, 35 sporadic conventional adenomas, 57 sporadic serrated lesions and 82 sporadic colorectal cancers. RESULTS Differential proteomics found 11 proteins significantly more abundant in dysplasia compared to inflammation, including Solute carrier family 12 member 2 (SLC12A2) which was confidently identified with 8 specific peptides and was below the limit of quantitation in both inflammatory and normal colon. SLC12A2 immunohistochemical analysis confirmed the discrimination of preneoplastic and neoplastic lesions from inflammatory lesions in mice, UC and in sporadic contexts. A specific SLC12A2 staining pattern termed "loss of gradient" reached 89% sensitivity, 95% specificity and 92% accuracy for UC-dysplasia diagnosis together with an inter-observer agreement of 95.24% (multirater κfree of 0.90; IC95%: 0.78 - 1.00). Such discrimination could not be obtained by Ki67 staining. This specific pattern was also associated with sporadic colorectal adenomas and cancers. CONCLUSIONS We found a specific SLC12A2 immunohistochemical staining pattern in precancerous and cancerous colonic UC-lesions which could be helpful for diagnosing dysplasia and cancer in UC and non-UC patients.
Collapse
Affiliation(s)
- Angela-Maria Merli
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
| | - Sophie Vieujean
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, Liège, Belgium
| | - Charlotte Massot
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, Liège, Belgium
| | - Noella Blétard
- Pathological Anatomy and Cytology, University Hospital CHU of Liège, Liège, Belgium
| | | | | | | | - Laurence Servais
- Laboratory of Cardiology, GIGA-Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Odile Wéra
- Laboratory of Cardiology, GIGA-Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Cécile Oury
- Laboratory of Cardiology, GIGA-Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Laurence de Leval
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Roberto Manzini
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sena Bluemel
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Edwin De Pauw
- Laboratory of Mass Spectrometry, University of Liège, Liège, Belgium
| | - C Coimbra Marques
- Abdominal Surgery Department, University Hospital CHU of Liège, Liège, Belgium
| | - Arnaud Colard
- Department of Gastroenterology, CHC Clinique Saint-Joseph, Liège, Belgium
| | - Anne Vijverman
- Department of Gastroenterology, CHR Citadelle, Liège, Belgium
| | - Philippe Delvenne
- Pathological Anatomy and Cytology, University Hospital CHU of Liège, Liège, Belgium
| | - Edouard Louis
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, Liège, Belgium
- Equally contributed to this work
| | - Marie-Alice Meuwis
- Laboratory of Translational Gastroenterology, University of Liège, Liège, Belgium
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, Liège, Belgium
- Equally contributed to this work
| |
Collapse
|
11
|
McNeill JK, Walton JC, Ryu V, Albers HE. The Excitatory Effects of GABA within the Suprachiasmatic Nucleus: Regulation of Na-K-2Cl Cotransporters (NKCCs) by Environmental Lighting Conditions. J Biol Rhythms 2020; 35:275-286. [PMID: 32406304 DOI: 10.1177/0748730420924271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The suprachiasmatic nucleus (SCN) contains a pacemaker that generates circadian rhythms and entrains them with the 24-h light-dark cycle (LD). The SCN is composed of 16,000 to 20,000 heterogeneous neurons in bilaterally paired nuclei. γ-amino butyric acid (GABA) is the primary neurochemical signal within the SCN and plays a key role in regulating circadian function. While GABA is the primary inhibitory neurotransmitter in the brain, there is now evidence that GABA can also exert excitatory effects in the adult brain. Cation chloride cotransporters determine the effects of GABA on chloride equilibrium, thereby determining whether GABA produces hyperpolarizing or depolarizing actions following activation of GABAA receptors. The activity of Na-K-2Cl cotransporter1 (NKCC1), the most prevalent chloride influx cotransporter isoform in the brain, plays a critical role in determining whether GABA has depolarizing effects. In the present study, we tested the hypothesis that NKCC1 protein expression in the SCN is regulated by environmental lighting and displays daily and circadian changes in the intact circadian system of the Syrian hamster. In hamsters housed in constant light (LL), the overall NKCC1 immunoreactivity (NKCC1-ir) in the SCN was significantly greater than in hamsters housed in LD or constant darkness (DD), although NKCC1 protein levels in the SCN were not different between hamsters housed in LD and DD. In hamsters housed in LD cycles, no differences in NKCC1-ir within the SCN were observed over the 24-h cycle. NKCC1 protein in the SCN was found to vary significantly over the circadian cycle in hamsters housed in free-running conditions. Overall, NKCC1 protein was greater in the ventral SCN than in the dorsal SCN, although no significant differences were observed across lighting conditions or time of day in either subregion. These data support the hypothesis that NKCC1 protein expression can be regulated by environmental lighting and circadian mechanisms within the SCN.
Collapse
Affiliation(s)
- John K McNeill
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| | - James C Walton
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| | - Vitaly Ryu
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| | - H Elliott Albers
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia
| |
Collapse
|
12
|
Mutai H, Wasano K, Momozawa Y, Kamatani Y, Miya F, Masuda S, Morimoto N, Nara K, Takahashi S, Tsunoda T, Homma K, Kubo M, Matsunaga T. Variants encoding a restricted carboxy-terminal domain of SLC12A2 cause hereditary hearing loss in humans. PLoS Genet 2020; 16:e1008643. [PMID: 32294086 PMCID: PMC7159186 DOI: 10.1371/journal.pgen.1008643] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 01/31/2020] [Indexed: 02/07/2023] Open
Abstract
Hereditary hearing loss is challenging to diagnose because of the heterogeneity of the causative genes. Further, some genes involved in hereditary hearing loss have yet to be identified. Using whole-exome analysis of three families with congenital, severe-to-profound hearing loss, we identified a missense variant of SLC12A2 in five affected members of one family showing a dominant inheritance mode, along with de novo splice-site and missense variants of SLC12A2 in two sporadic cases, as promising candidates associated with hearing loss. Furthermore, we detected another de novo missense variant of SLC12A2 in a sporadic case. SLC12A2 encodes Na+, K+, 2Cl− cotransporter (NKCC) 1 and plays critical roles in the homeostasis of K+-enriched endolymph. Slc12a2-deficient mice have congenital, profound deafness; however, no human variant of SLC12A2 has been reported as associated with hearing loss. All identified SLC12A2 variants mapped to exon 21 or its 3’-splice site. In vitro analysis indicated that the splice-site variant generates an exon 21-skipped SLC12A2 mRNA transcript expressed at much lower levels than the exon 21-included transcript in the cochlea, suggesting a tissue-specific role for the exon 21-encoded region in the carboy-terminal domain. In vitro functional analysis demonstrated that Cl− influx was significantly decreased in all SLC12A2 variants studied. Immunohistochemistry revealed that SLC12A2 is located on the plasma membrane of several types of cells in the cochlea, including the strial marginal cells, which are critical for endolymph homeostasis. Overall, this study suggests that variants affecting exon 21 of the SLC12A2 transcript are responsible for hereditary hearing loss in humans. Sounds are perceived by auditory sensory cells, owing to tissues surrounding them, including the cochlear lateral wall. Part of the cochlear lateral wall, the stria vascularis, is critical for production and maintenance of inner-ear fluid with a high potassium concentration, and for generating the positive voltage in the inner ear, important for sound perception, by stimulating secretion of potassium from marginal cells. The gene SLC12A2 encodes a protein involved in sodium, potassium, and chloride transport essential for proper function of specific cells in the stria vascularis; however, human variants of SLC12A2 have not previously been associated with hearing loss. By comprehensive genetic analysis of protein-coding sequences, we identified four candidate changes in SLC12A2 in four families with congenital, severe-to-profound hearing loss. Intriguingly, all four genetic variants were either within or at the 3’-splice site of the exon 21 which encodes a part of the carboxy terminal intracellular domain of SLC12A2. Experiments in cultured cells showed that skipping or mutation of exon 21 significantly decreased chloride influx mediated by the SLC12A2 protein. Overall, our results strongly indicate that mutations influencing exon 21 of SLC12A2 represent a novel mechanism underlying deafness in humans.
Collapse
Affiliation(s)
- Hideki Mutai
- Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Meguro, Tokyo, Japan
| | - Koichiro Wasano
- Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Meguro, Tokyo, Japan
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Yoshidakonoecho, Kyoto, Japan
| | - Fuyuki Miya
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Sawako Masuda
- Department of Otorhinolaryngology, National Hospital Organization Mie National Hospital, Tsu, Mie, Japan
| | - Noriko Morimoto
- Department of Otorhinolaryngology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Kiyomitsu Nara
- Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Meguro, Tokyo, Japan
| | - Satoe Takahashi
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Medical Science Mathematics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kazuaki Homma
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern University, Evanston, Illinois, United States of America
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Tatsuo Matsunaga
- Division of Hearing and Balance Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Meguro, Tokyo, Japan
- Medical Genetics Center, National Hospital Organization Tokyo Medical Center, Meguro, Tokyo, Japan
- * E-mail:
| |
Collapse
|
13
|
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: 65] [Impact Index Per Article: 13.0] [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
|
14
|
Koumangoye R, Omer S, Delpire E. A dileucine motif in the COOH-terminal domain of NKCC1 targets the cotransporter to the plasma membrane. Am J Physiol Cell Physiol 2019; 316:C545-C558. [PMID: 30865516 DOI: 10.1152/ajpcell.00023.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Na+-K+-2Cl- cotransporter-1 (NKCC1) mediates the electroneutral transport of Na+, K+, and Cl- and is normally localized to the basolateral membrane of polarized epithelial cells. We recently reported the first known solute carrier family 12 member 2 ( SLC12A2) mutation (we call NKCC1-DFX) that causes epithelial dysfunction in an undiagnosed disease program case. The heterozygous mutation leads to truncation of the COOH-terminal tail of the cotransporter, resulting in both mutant and wild-type cotransporters being mistrafficked to the apical membrane of polarized epithelial cells. Here we demonstrate by using consecutive truncations and site-directed mutagenesis of the COOH-terminal domain of NKCC1 that truncation of NKCC1 COOH domain uncouples the cotransporter from the lateral membrane. We identify a dileucine motif that, when mutated, leads to cotransporter accumulation in the cytoplasm and mistrafficking to the apical/subapical region of epithelial cells, thereby recapitulating the phenotype observed with the patient mutation. We show that truncation deletion and LL substitution mutants are trafficked out of the endoplasmic reticulum and trans-Golgi network but accumulate in early and late endosomes where they are degraded.
Collapse
Affiliation(s)
- Rainelli Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Salma Omer
- Department of Anesthesiology, Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine , Nashville, Tennessee
| |
Collapse
|
15
|
Brandt C, Seja P, Töllner K, Römermann K, Hampel P, Kalesse M, Kipper A, Feit PW, Lykke K, Toft-Bertelsen TL, Paavilainen P, Spoljaric I, Puskarjov M, MacAulay N, Kaila K, Löscher W. Bumepamine, a brain-permeant benzylamine derivative of bumetanide, does not inhibit NKCC1 but is more potent to enhance phenobarbital's anti-seizure efficacy. Neuropharmacology 2018; 143:186-204. [DOI: 10.1016/j.neuropharm.2018.09.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/30/2018] [Accepted: 09/16/2018] [Indexed: 01/01/2023]
|
16
|
Azosemide is more potent than bumetanide and various other loop diuretics to inhibit the sodium-potassium-chloride-cotransporter human variants hNKCC1A and hNKCC1B. Sci Rep 2018; 8:9877. [PMID: 29959396 PMCID: PMC6026185 DOI: 10.1038/s41598-018-27995-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/14/2018] [Indexed: 12/31/2022] Open
Abstract
The Na+–K+–2Cl− cotransporter NKCC1 plays a role in neuronal Cl− homeostasis secretion and represents a target for brain pathologies with altered NKCC1 function. Two main variants of NKCC1 have been identified: a full-length NKCC1 transcript (NKCC1A) and a shorter splice variant (NKCC1B) that is particularly enriched in the brain. The loop diuretic bumetanide is often used to inhibit NKCC1 in brain disorders, but only poorly crosses the blood-brain barrier. We determined the sensitivity of the two human NKCC1 splice variants to bumetanide and various other chemically diverse loop diuretics, using the Xenopus oocyte heterologous expression system. Azosemide was the most potent NKCC1 inhibitor (IC50s 0.246 µM for hNKCC1A and 0.197 µM for NKCC1B), being about 4-times more potent than bumetanide. Structurally, a carboxylic group as in bumetanide was not a prerequisite for potent NKCC1 inhibition, whereas loop diuretics without a sulfonamide group were less potent. None of the drugs tested were selective for hNKCC1B vs. hNKCC1A, indicating that loop diuretics are not a useful starting point to design NKCC1B-specific compounds. Azosemide was found to exert an unexpectedly potent inhibitory effect and as a non-acidic compound, it is more likely to cross the blood-brain barrier than bumetanide.
Collapse
|
17
|
Di Cristo G, Awad PN, Hamidi S, Avoli M. KCC2, epileptiform synchronization, and epileptic disorders. Prog Neurobiol 2018; 162:1-16. [DOI: 10.1016/j.pneurobio.2017.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/09/2017] [Accepted: 11/28/2017] [Indexed: 12/31/2022]
|
18
|
Caceres PS, Benedicto I, Lehmann GL, Rodriguez-Boulan EJ. Directional Fluid Transport across Organ-Blood Barriers: Physiology and Cell Biology. Cold Spring Harb Perspect Biol 2017; 9:a027847. [PMID: 28003183 PMCID: PMC5334253 DOI: 10.1101/cshperspect.a027847] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Directional fluid flow is an essential process for embryo development as well as for organ and organism homeostasis. Here, we review the diverse structure of various organ-blood barriers, the driving forces, transporters, and polarity mechanisms that regulate fluid transport across them, focusing on kidney-, eye-, and brain-blood barriers. We end by discussing how cross talk between barrier epithelial and endothelial cells, perivascular cells, and basement membrane signaling contribute to generate and maintain organ-blood barriers.
Collapse
Affiliation(s)
- Paulo S Caceres
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Ignacio Benedicto
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Guillermo L Lehmann
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| | - Enrique J Rodriguez-Boulan
- Margaret Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
19
|
Bialer M, Johannessen SI, Levy RH, Perucca E, Tomson T, White HS. Progress report on new antiepileptic drugs: A summary of the Thirteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIII). Epilepsia 2017; 58:181-221. [PMID: 28111749 DOI: 10.1111/epi.13634] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2016] [Indexed: 01/05/2023]
Abstract
The Thirteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIII) took place in Madrid, Spain, on June 26-29, 2016, and was attended by >200 delegates from 31 countries. The present Progress Report provides an update on experimental and clinical results for drugs presented at the Conference. Compounds for which summary data are presented include an AED approved in 2016 (brivaracetam), 12 drugs in phase I-III clinical development (adenosine, allopregnanolone, bumetanide, cannabidiol, cannabidivarin, 2-deoxy-d-glucose, everolimus, fenfluramine, huperzine A, minocycline, SAGE-217, and valnoctamide) and 6 compounds or classes of compounds for which only preclinical data are available (bumetanide derivatives, sec-butylpropylacetamide, FV-082, 1OP-2198, NAX 810-2, and SAGE-689). Overall, the results presented at the Conference show that considerable efforts are ongoing into discovery and development of AEDs with potentially improved therapeutic profiles compared with existing agents. Many of the drugs discussed in this report show innovative mechanisms of action and many have shown promising results in patients with pharmacoresistant epilepsies, including previously neglected rare and severe epilepsy syndromes.
Collapse
Affiliation(s)
- Meir Bialer
- Faculty of Medicine, School of Pharmacy and David R. Bloom Center for Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Svein I Johannessen
- The National Center for Epilepsy, Sandvika, Norway.,Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | - René H Levy
- Department of Pharmaceutics and Neurological Surgery, University of Washington, Seattle, Washington, U.S.A
| | - Emilio Perucca
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy.,C. Mondino National Neurological Institute, Pavia, Italy
| | - Torbjörn Tomson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, Washington, U.S.A
| |
Collapse
|
20
|
Martín-Aragón Baudel MAS, Poole AV, Darlison MG. Chloride co-transporters as possible therapeutic targets for stroke. J Neurochem 2016; 140:195-209. [PMID: 27861901 DOI: 10.1111/jnc.13901] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/08/2016] [Accepted: 11/08/2016] [Indexed: 02/06/2023]
Abstract
Stroke is one of the major causes of death and disability worldwide. The major type of stroke is an ischaemic one, which is caused by a blockage that interrupts blood flow to the brain. There are currently very few pharmacological strategies to reduce the damage and social burden triggered by this pathology. The harm caused by the interruption of blood flow to the brain unfolds in the subsequent hours and days, so it is critical to identify new therapeutic targets that could reduce neuronal death associated with the spread of the damage. Here, we review some of the key molecular mechanisms involved in the progression of neuronal death, focusing on some new and promising studies. In particular, we focus on the potential of the chloride co-transporter (CCC) family of proteins, mediators of the GABAergic response, both during the early and later stages of stroke, to promote neuroprotection and recovery. Different studies of CCCs during the chronic and recovery phases post-stroke reveal the importance of timing when considering CCCs as potential neuroprotective and/or neuromodulator targets. The molecular regulatory mechanisms of the two main neuronal CCCs, NKCC1 and KCC2, are further discussed as an indirect approach for promoting neuroprotection and neurorehabilitation following an ischaemic insult. Finally, we mention the likely importance of combining different strategies in order to achieve more effective therapies.
Collapse
Affiliation(s)
| | - Amy V Poole
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, UK
| | - Mark G Darlison
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, UK
| |
Collapse
|
21
|
Sedmak G, Jovanov-Milošević N, Puskarjov M, Ulamec M, Krušlin B, Kaila K, Judaš M. Developmental Expression Patterns of KCC2 and Functionally Associated Molecules in the Human Brain. Cereb Cortex 2016; 26:4574-4589. [PMID: 26428952 DOI: 10.1093/cercor/bhv218] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Work on rodents demonstrated that steep upregulation of KCC2, a neuron-specific Cl- extruder of cation-chloride cotransporter (CCC) family, commences in supraspinal structures at around birth, leading to establishment of hyperpolarizing GABAergic responses. We describe spatiotemporal expression profiles of the entire CCC family in human brain. KCC2 mRNA was observed already at 10th postconceptional week (PCW) in amygdala, cerebellum, and thalamus. KCC2-immunoreactive (KCC2-ir) neurons were abundant in subplate at 18 PCW. By 25 PCW, numerous subplate and cortical plate neurons became KCC2-ir. The mRNA expression profiles of α- and β-isoforms of Na-K ATPase, which fuels cation-chloride cotransport, as well of tropomyosin receptor kinase B (TrkB), which promotes developmental upregulation of KCC2, were consistent with data from studies on rodents about their interactions with KCC2. Thus, in human brain, expression of KCC2 and its functionally associated proteins begins in early fetal period. Our work facilitates translation of results on CCC functions from animal studies to human and refutes the view that poor efficacy of anticonvulsants in the term human neonate is attributable to the lack of KCC2. We propose that perinatally low threshold for activation of Ca2+-dependent protease calpain renders neonates susceptible to downregulation of KCC2 by traumatic events, such as perinatal hypoxia ischemia.
Collapse
Affiliation(s)
| | | | - Martin Puskarjov
- Department of Biosciences and Neuroscience Center, University of Helsinki, Viikinkaari 1, Helsinki FI-00014, Finland
| | - Monika Ulamec
- Department of Pathology, Clinical Hospital Center Sisters of Mercy, University of Zagreb School of Medicine, Zagreb 10 000, Croatia
| | - Božo Krušlin
- Department of Pathology, Clinical Hospital Center Sisters of Mercy, University of Zagreb School of Medicine, Zagreb 10 000, Croatia
| | - Kai Kaila
- Department of Biosciences and Neuroscience Center, University of Helsinki, Viikinkaari 1, Helsinki FI-00014, Finland
| | | |
Collapse
|
22
|
Dayioglu E, Buharalioglu CK, Saracoglu F, Akar F. The Effects of Bumetanide on Human Umbilical Artery Contractions. Reprod Sci 2016; 14:246-52. [PMID: 17636238 DOI: 10.1177/1933719107300871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The authors investigate the effect of bumetan ide, an inhibitor of NKCC1 and a loop diuretic, on the tone of human umbilical artery (HUA). Rings of HUA (n = 35) from vaginal deliveries were suspended for isometric tension recordings in organ baths. Cumulative concentration-response curves to serotonin, histamine, and KCl were performed in the absence (control) or in the presence of bumetanide. The relaxant effect of bumetanide was also evaluated in serotonin- and histamine-induced contractions. Bumetanide inhibited HUA tone in serotonin- and histamine-induced contractions with significant changes in the potency (pD(2)) and maximum contractile response (E(max)) values. However, only pD( 2) values for KCl-induced contraction significantly changed in the presence of bumetanide. Bumetanide caused concentration-dependent and sustained relaxations in serotonin-induced contraction; however, there was refractoriness in histamine-induced contraction. These findings raise the possibility that NKCC1 may play a role in the regulation of the umbilical artery tone.
Collapse
Affiliation(s)
- Emel Dayioglu
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | | | | | | |
Collapse
|
23
|
Lykke K, Töllner K, Feit PW, Erker T, MacAulay N, Löscher W. The search for NKCC1-selective drugs for the treatment of epilepsy: Structure-function relationship of bumetanide and various bumetanide derivatives in inhibiting the human cation-chloride cotransporter NKCC1A. Epilepsy Behav 2016; 59:42-9. [PMID: 27088517 DOI: 10.1016/j.yebeh.2016.03.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/11/2016] [Accepted: 03/12/2016] [Indexed: 11/30/2022]
Abstract
The Na(+)-K(+)-Cl(-) cotransporter NKCC1 plays a major role in the regulation of intraneuronal Cl(-) concentration. Abnormal functionality of NKCC1 has been implicated in several brain disorders, including epilepsy. Bumetanide is the only available selective NKCC1 inhibitor, but also inhibits NKCC2, which can cause severe adverse effects during treatment of brain disorders. A NKCC1-selective bumetanide derivative would therefore be a desirable option. In the present study, we used the Xenopus oocyte heterologous expression system to compare the effects of bumetanide and several derivatives on the two major human splice variants of NKCCs, hNKCC1A and hNKCC2A. The derivatives were selected from a series of ~5000 3-amino-5-sulfamoylbenzoic acid derivatives, covering a wide range of structural modifications and diuretic potencies. To our knowledge, such structure-function relationships have not been performed before for NKCC1. Half maximal inhibitory concentrations (IC50s) of bumetanide were 0.68 (hNKCC1A) and 4.0μM (hNKCC2A), respectively, indicating that this drug is 6-times more potent to inhibit hNKCC1A than hNKCC2A. Side chain substitutions in the bumetanide molecule variably affected the potency to inhibit hNKCC1A. This allowed defining the minimal structural requirements necessary for ligand interaction. Unexpectedly, only a few of the bumetanide derivatives examined were more potent than bumetanide to inhibit hNKCC1A, and most of them also inhibited hNKCC2A, with a highly significant correlation between IC50s for the two NKCC isoforms. These data indicate that the structural requirements for inhibition of NKCC1 and NKCC2 are similar, which complicates development of bumetanide-related compounds with high selectivity for NKCC1.
Collapse
Affiliation(s)
- Kasper Lykke
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Peter W Feit
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Thomas Erker
- Department of Medicinal Chemistry, University of Vienna, Vienna, Austria
| | - Nanna MacAulay
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
24
|
Impact of Hybrid and Complex N-Glycans on Cell Surface Targeting of the Endogenous Chloride Cotransporter Slc12a2. Int J Cell Biol 2015; 2015:505294. [PMID: 26351455 PMCID: PMC4553341 DOI: 10.1155/2015/505294] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/29/2015] [Accepted: 07/07/2015] [Indexed: 11/21/2022] Open
Abstract
The Na+K+2Cl− cotransporter-1 (Slc12a2, NKCC1) is widely distributed and involved in cell volume/ion regulation. Functional NKCC1 locates in the plasma membrane of all cells studied, particularly in the basolateral membrane of most polarized cells. Although the mechanisms involved in plasma membrane sorting of NKCC1 are poorly understood, it is assumed that N-glycosylation is necessary. Here, we characterize expression, N-glycosylation, and distribution of NKCC1 in COS7 cells. We show that ~25% of NKCC1 is complex N-glycosylated whereas the rest of it corresponds to core/high-mannose and hybrid-type N-glycosylated forms. Further, ~10% of NKCC1 reaches the plasma membrane, mostly as core/high-mannose type, whereas ~90% of NKCC1 is distributed in defined intracellular compartments. In addition, inhibition of the first step of N-glycan biosynthesis with tunicamycin decreases total and plasma membrane located NKCC1 resulting in almost undetectable cotransport function. Moreover, inhibition of N-glycan maturation with swainsonine or kifunensine increased core/hybrid-type NKCC1 expression but eliminated plasma membrane complex N-glycosylated NKCC1 and transport function. Together, these results suggest that (i) NKCC1 is delivered to the plasma membrane of COS7 cells independently of its N-glycan nature, (ii) most of NKCC1 in the plasma membrane is core/hybrid-type N-glycosylated, and (iii) the minimal proportion of complex N-glycosylated NKCC1 is functionally active.
Collapse
|
25
|
Kaila K, Price TJ, Payne JA, Puskarjov M, Voipio J. Cation-chloride cotransporters in neuronal development, plasticity and disease. Nat Rev Neurosci 2014; 15:637-54. [PMID: 25234263 DOI: 10.1038/nrn3819] [Citation(s) in RCA: 505] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Electrical activity in neurons requires a seamless functional coupling between plasmalemmal ion channels and ion transporters. Although ion channels have been studied intensively for several decades, research on ion transporters is in its infancy. In recent years, it has become evident that one family of ion transporters, cation-chloride cotransporters (CCCs), and in particular K(+)-Cl(-) cotransporter 2 (KCC2), have seminal roles in shaping GABAergic signalling and neuronal connectivity. Studying the functions of these transporters may lead to major paradigm shifts in our understanding of the mechanisms underlying brain development and plasticity in health and disease.
Collapse
Affiliation(s)
- Kai Kaila
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Theodore J Price
- University of Texas at Dallas, School of Behavior and Brain Sciences, Dallas, Texas 75093, USA
| | - John A Payne
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California 95616, USA
| | - Martin Puskarjov
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Juha Voipio
- Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
26
|
Puskarjov M, Kahle KT, Ruusuvuori E, Kaila K. Pharmacotherapeutic targeting of cation-chloride cotransporters in neonatal seizures. Epilepsia 2014; 55:806-18. [PMID: 24802699 PMCID: PMC4284054 DOI: 10.1111/epi.12620] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2014] [Indexed: 12/15/2022]
Abstract
Seizures are a common manifestation of acute neurologic insults in neonates and are often resistant to the standard antiepileptic drugs that are efficacious in children and adults. The paucity of evidence-based treatment guidelines, coupled with a rudimentary understanding of disease pathogenesis, has made the current treatment of neonatal seizures empiric and often ineffective, highlighting the need for novel therapies. Key developmental differences in γ-aminobutyric acid (GABA)ergic neurotransmission between the immature and mature brain, and trauma-induced alterations in the function of the cation-chloride cotransporters (CCCs) NKCC1 and KCC2, probably contribute to the poor efficacy of standard antiepileptic drugs used in the treatment of neonatal seizures. Although CCCs are attractive drug targets, bumetanide and other existing CCC inhibitors are suboptimal because of pharmacokinetic constraints and lack of target specificity. Newer approaches including isoform-specific NKCC1 inhibitors with increased central nervous system penetration, and direct and indirect strategies to enhance KCC2-mediated neuronal chloride extrusion, might allow therapeutic modulation of the GABAergic system for neonatal seizure treatment. A PowerPoint slide summarizing this article is available for download in the Supporting Information section here.
Collapse
Affiliation(s)
- Martin Puskarjov
- Department of Biosciences and Neuroscience Center, University of HelsinkiHelsinki, Finland
| | - Kristopher T Kahle
- Department of Neurosurgery, Harvard Medical School, Massachusetts General HospitalBoston, Massachusetts, U.S.A
| | - Eva Ruusuvuori
- Department of Biosciences and Neuroscience Center, University of HelsinkiHelsinki, Finland
| | - Kai Kaila
- Department of Biosciences and Neuroscience Center, University of HelsinkiHelsinki, Finland
| |
Collapse
|
27
|
Morita Y, Callicott JH, Testa LR, Mighdoll MI, Dickinson D, Chen Q, Tao R, Lipska BK, Kolachana B, Law AJ, Ye T, Straub RE, Weinberger DR, Kleinman JE, Hyde TM. Characteristics of the cation cotransporter NKCC1 in human brain: alternate transcripts, expression in development, and potential relationships to brain function and schizophrenia. J Neurosci 2014; 34:4929-40. [PMID: 24695712 PMCID: PMC3972720 DOI: 10.1523/jneurosci.1423-13.2014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 01/10/2014] [Accepted: 02/14/2014] [Indexed: 02/07/2023] Open
Abstract
Early in development, GABA, an inhibitory neurotransmitter in adults, is excitatory. NKCC1 (SLC12A2) encodes one of two cation chloride cotransporters mediating the conversion of GABA from excitatory to inhibitory. Using 3' and 5' RACE and PCR, we verified previously characterized alternative transcripts of NKCC1a (1-27) and NKCC1b (1-27(Δ21)), identified new NKCC1 transcripts, and explored their expression patterns during human prefrontal cortical development. A novel ultra-short transcript (1-2a) was expressed preferentially in the fetus. Expression of NKCC1b and 1-2a were decreased in schizophrenia compared with controls (NKCC1b: 0.8-fold decrease, p = 0.013; 1-2a: 0.8-fold decrease, p = 0.006). Furthermore, the expression of NKCC1b was associated with NKCC1 polymorphism rs3087889. The minor allele at rs3087889, associated with reduced NKCC1b expression (homozygous for major allele: N = 37; homozygous for minor allele: N = 15; 1.5-fold decrease; p < 0.01), was also associated with a modest increase in schizophrenia risk in a case-control sample (controls: N = 435; cases: N = 397, OR = 1.5). This same allele was then found associated with cognitive (n = 369) and fMRI (n = 313) intermediate phenotypes associated with schizophrenia-working memory (Cohen's d = 0.35), global cognition or g (d = 0.18), and prefrontal inefficiency (d = 0.36) as measured by BOLD fMRI during a working memory task. Together, these preclinical and clinical results suggest that variation in NKCC1 may increase risk for schizophrenia via alterations of mRNA expression at the molecular level and impairment of optimal prefrontal function at the macro or systems level.
Collapse
Affiliation(s)
- Yukitaka Morita
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Department of Psychiatry, Hiroshima City Hospital, Hiroshima City, Hiroshima Prefecture, 730-8518, Japan
| | - Joseph H. Callicott
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Lauren R. Testa
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Michelle I. Mighdoll
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Dwight Dickinson
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Qiang Chen
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Ran Tao
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Barbara K. Lipska
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Bhaskar Kolachana
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
| | - Amanda J. Law
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Neurodevelopmental and Neuropsychiatric Genetics Laboratory, Departments of Psychiatry and Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, and
| | - Tianzhang Ye
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Richard E. Straub
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Daniel R. Weinberger
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
- Departments of Psychiatry, Neurology, Neuroscience and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Joel E. Kleinman
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Thomas M. Hyde
- Clinical Brain Disorders Branch, Genes, Cognition, and Psychosis Program, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
- Departments of Psychiatry, Neurology, Neuroscience and the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
28
|
Markadieu N, Delpire E. Physiology and pathophysiology of SLC12A1/2 transporters. Pflugers Arch 2014; 466:91-105. [PMID: 24097229 PMCID: PMC3877717 DOI: 10.1007/s00424-013-1370-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/21/2013] [Accepted: 09/23/2013] [Indexed: 01/14/2023]
Abstract
The electroneutral Na(+)-K(+)-Cl(-) cotransporters NKCC1 (encoded by the SLC12A2 gene) and NKCC2 (SLC12A1 gene) belong to the Na(+)-dependent subgroup of solute carrier 12 (SLC12) family of transporters. They mediate the electroneutral movement of Na(+) and K(+), tightly coupled to the movement of Cl(-) across cell membranes. As they use the energy of the ion gradients generated by the Na(+)/K(+)-ATPase to transport Na(+), K(+), and Cl(-) from the outside to the inside of a cell, they are considered secondary active transport mechanisms. NKCC-mediated transport occurs in a 1Na(+), 1K(+), and 2Cl(-) ratio, although NKCC1 has been shown to sometimes mediate partial reactions. Both transporters are blocked by bumetanide and furosemide, drugs which are commonly used in clinical medicine. NKCC2 is the molecular target of loop diuretics as it is expressed on the apical membrane of thick ascending limb of Henle epithelial cells, where it mediates NaCl reabsorption. NKCC1, in contrast, is found on the basolateral membrane of Cl(-) secretory epithelial cells, as well as in a variety of non-epithelial cells, where it mediates cell volume regulation and participates in Cl(-) homeostasis. Following their molecular identification two decades ago, much has been learned about their biophysical properties, their mode of operation, their regulation by kinases and phosphatases, and their physiological relevance. However, despite this tremendous amount of new information, there are still so many gaps in our knowledge. This review summarizes information that constitutes consensus in the field, but it also discusses current points of controversy and highlights many unanswered questions.
Collapse
Affiliation(s)
- Nicolas Markadieu
- Department of Anesthesiology, Vanderbilt University School of Medicine, MCN T-4202, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | | |
Collapse
|
29
|
Abbas L, Whitfield TT. Nkcc1 (Slc12a2) is required for the regulation of endolymph volume in the otic vesicle and swim bladder volume in the zebrafish larva. Development 2009; 136:2837-48. [PMID: 19633174 DOI: 10.1242/dev.034215] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Endolymph is the specialised extracellular fluid present inside the inner ear. In mammals, disruptions to endolymph homeostasis can result in either collapse or distension of the endolymphatic compartment in the cochlea, with concomitant hearing loss. The zebrafish little ears (lte) mutant shows a collapse of the otic vesicle in the larva, apparently owing to a loss of endolymphatic fluid in the ear, together with an over-inflation of the swim bladder. Mutant larvae display signs of abnormal vestibular function by circling and swimming upside down. The two available alleles of lte are homozygous lethal: mutant larvae fail to thrive beyond 6 days post-fertilisation. Patterning of the otic vesicle is apparently normal. However, the expression of several genes thought to play a role in endolymph production is downregulated, including the sodium-potassium-chloride cotransporter gene nkcc1 (slc12a2) and several Na(+)/K(+)-ATPase channel subunit genes. We show here that lte mutations correspond to lesions in nkcc1. Each allele has a point mutation that disrupts splicing, leading to frame shifts in the coding region that predict the generation of truncated products. Endolymph collapse in the lte/nkcc1 mutant shows distinct parallels to that seen in mouse Nkcc1 mutants, validating zebrafish as a model for the study of endolymph disorders. The collapse in ear volume can be ameliorated in the to27d allele of lte by injection of a morpholino that blocks splicing at an ectopic site introduced by the mutation. This exemplifies the use of morpholinos as potential therapeutic agents for genetic disease.
Collapse
Affiliation(s)
- Leila Abbas
- MRC Centre for Developmental and Biomedical Genetics and Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | | |
Collapse
|
30
|
Blaesse P, Airaksinen MS, Rivera C, Kaila K. Cation-chloride cotransporters and neuronal function. Neuron 2009; 61:820-38. [PMID: 19323993 DOI: 10.1016/j.neuron.2009.03.003] [Citation(s) in RCA: 567] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 03/04/2009] [Accepted: 03/06/2009] [Indexed: 11/29/2022]
Abstract
Recent years have witnessed a steep increase in studies on the diverse roles of neuronal cation-chloride cotransporters (CCCs). The versatility of CCC gene transcription, posttranslational modification, and trafficking are on par with what is known about ion channels. The cell-specific and subcellular expression patterns of different CCC isoforms have a key role in modifying a neuron's electrophysiological phenotype during development, synaptic plasticity, and disease. While having a major role in controlling responses mediated by GABA(A) and glycine receptors, CCCs also show close interactions with glutamatergic signaling. A cross-talk among CCCs and trophic factors is important in short-term and long-term modification of neuronal properties. CCCs appear to be multifunctional proteins that are also involved in shaping neuronal structure at various stages of development, from stem cells to synaptogenesis. The rapidly expanding work on CCCs promotes our understanding of fundamental mechanisms that control brain development and functions under normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Peter Blaesse
- Department of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 1, FIN-00014, Helsinki, Finland
| | | | | | | |
Collapse
|
31
|
Carmosino M, Giménez I, Caplan M, Forbush B. Exon loss accounts for differential sorting of Na-K-Cl cotransporters in polarized epithelial cells. Mol Biol Cell 2008; 19:4341-51. [PMID: 18667527 DOI: 10.1091/mbc.e08-05-0478] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The renal Na-K-Cl cotransporter (NKCC2) is selectively expressed in the apical membranes of cells of the mammalian kidney, where it is the target of the clinically important loop diuretics. In contrast, the "secretory" NKCC1 cotransporter is localized in the basolateral membranes of many epithelia. To identify the sorting signal(s) that direct trafficking of NKCCs, we generated chimeras between the two isoforms and expressed these constructs in polarized renal epithelial cell lines. This analysis revealed an amino acid stretch in NKCC2 containing apical sorting information. The NKCC1 C terminus contains a dileucine motif that constitutes the smallest essential component of its basolateral sorting signal. NKCC1 lacking this motif behaves as an apical protein. Examination of the NKCC gene structure reveals that this dileucine motif is encoded by an additional exon in NKCC1 absent in NKCC2. Phylogenetic analysis of this exon suggests that the evolutionary loss of this exon from the gene encoding the basolateral NKCC1 constitutes a novel mechanism that accounts for the apical sorting of the protein encoded by the NKCC2 gene.
Collapse
Affiliation(s)
- Monica Carmosino
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | |
Collapse
|
32
|
Muñoz A, Méndez P, DeFelipe J, Alvarez-Leefmans FJ. Cation-chloride cotransporters and GABA-ergic innervation in the human epileptic hippocampus. Epilepsia 2007; 48:663-73. [PMID: 17319917 DOI: 10.1111/j.1528-1167.2007.00986.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Intracellular chloride concentration, [Cl(-)](i), determines the polarity of GABA(A)-induced neuronal Cl(-) currents. In neurons, [Cl(-)](i) is set by the activity of Na(+), K(+), 2Cl(-) cotransporters (NKCC) such as NKCC1, which physiologically accumulate Cl(-) in the cell, and Cl(-) extruding K(+), Cl(-) cotransporters like KCC2. Alterations in the balance of NKCC1 and KCC2 activity may determine the switch from hyperpolarizing to depolarizing effects of GABA, reported in the subiculum of epileptic patients with hippocampal sclerosis. We studied the expression of NKCC (putative NKCC1) and KCC2 in human normal temporal neocortex by Western blot analysis and in normal and epileptic regions of the subiculum and the hippocampus proper using immunocytochemistry. Western blot analysis revealed NKCC and KCC2 proteins in adult human neocortical membranes similar to those in rat neocortex. NKCC and KCC2 immunolabeling of pyramidal and nonpyramidal cells was found in normal and epileptic hippocampal formation. In the transition between the subiculum with sclerotic regions of CA1, known to exhibit epileptogenic activity, double immunolabeling of NKCC and KCC2 revealed that approximately 20% of the NKCC-immunoreactive neurons do not express KCC2. In these same areas some neurons were distinctly hyperinnervated by parvalbumin (PV) positive hypertrophic basket formations that innervated mostly neurons expressing NKCC (74%) and to a lesser extent NKCC-immunonegative neurons (26%). Hypertrophic basket formations also innervated KCC2-positive (76%) and -negative (24%) neurons. The data suggest that changes in the relative expression of NKCC1 and KCC2 in neurons having aberrant GABA-ergic hyperinnervation may contribute to epileptiform activity in the subicular regions adjacent to sclerotic areas of the hippocampus.
Collapse
|
33
|
Gillen CM, Blair CR, Heilman NR, Somple M, Stulberg M, Thombre R, Watson N, Gillen KM, Itagaki H. The cation-chloride cotransporter, masBSC, is widely expressed in Manduca sexta tissues. JOURNAL OF INSECT PHYSIOLOGY 2006; 52:661-8. [PMID: 16730744 DOI: 10.1016/j.jinsphys.2006.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 02/20/2006] [Accepted: 02/21/2006] [Indexed: 05/09/2023]
Abstract
Cation-chloride cotransporters, including the Na-K-Cl cotransporter, play an important role in epithelial ion transport in insects. We have determined the tissue distribution of Manduca sexta bumetanide sensitive cotransporter (masBSC), a putative Na-K-Cl cotransporter that was originally cloned from M. sexta Malpighian tubules. We developed a polyclonal antibody (M6) against a C-terminal fragment of masBSC. masBSC protein was detected by M6 at an apparent molecular mass of approximately 220kDa in M. sexta foregut, midgut, hindgut, Malpighian tubule, salivary gland, fat body, trachea, and nerve cord. Higher expression was observed in the foregut than in other tissues. M6 stained the apical membrane of midgut epithelial cells in cross-sections of third instar larvae. The transcript of masBSC was detected by RT-PCR in midgut, Malpighian tubule, hindgut, trachea, nerve cord, and fat bodies. Taken together, these findings demonstrate that masBSC is widely expressed in M. sexta. While the specific function of masBSC remains unknown, the wide distribution indicates a role of masBSC in a broad range of tissues.
Collapse
|
34
|
Pedersen SF, O'Donnell ME, Anderson SE, Cala PM. Physiology and pathophysiology of Na+/H+ exchange and Na+ -K+ -2Cl- cotransport in the heart, brain, and blood. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1-25. [PMID: 16484438 DOI: 10.1152/ajpregu.00782.2005] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Maintenance of a stable cell volume and intracellular pH is critical for normal cell function. Arguably, two of the most important ion transporters involved in these processes are the Na+/H+ exchanger isoform 1 (NHE1) and Na+ -K+ -2Cl- cotransporter isoform 1 (NKCC1). Both NHE1 and NKCC1 are stimulated by cell shrinkage and by numerous other stimuli, including a wide range of hormones and growth factors, and for NHE1, intracellular acidification. Both transporters can be important regulators of cell volume, yet their activity also, directly or indirectly, affects the intracellular concentrations of Na+, Ca2+, Cl-, K+, and H+. Conversely, when either transporter responds to a stimulus other than cell shrinkage and when the driving force is directed to promote Na+ entry, one consequence may be cell swelling. Thus stimulation of NHE1 and/or NKCC1 by a deviation from homeostasis of a given parameter may regulate that parameter at the expense of compromising others, a coupling that may contribute to irreversible cell damage in a number of pathophysiological conditions. This review addresses the roles of NHE1 and NKCC1 in the cellular responses to physiological and pathophysiological stress. The aim is to provide a comprehensive overview of the mechanisms and consequences of stress-induced stimulation of these transporters with focus on the heart, brain, and blood. The physiological stressors reviewed are metabolic/exercise stress, osmotic stress, and mechanical stress, conditions in which NHE1 and NKCC1 play important physiological roles. With respect to pathophysiology, the focus is on ischemia and severe hypoxia where the roles of NHE1 and NKCC1 have been widely studied yet remain controversial and incompletely elucidated.
Collapse
Affiliation(s)
- S F Pedersen
- Department of Biochemistry, Institute of Molecular Biology and Physiology, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
35
|
Donev RM, Morgan BP. A quantitative method for comparison of expression of alternatively spliced genes using different primer pairs. ACTA ACUST UNITED AC 2005; 66:23-31. [PMID: 16376430 DOI: 10.1016/j.jbbm.2005.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2005] [Revised: 11/09/2005] [Accepted: 11/10/2005] [Indexed: 11/26/2022]
Abstract
In this paper we describe a novel two-step method for comparison of expression of alternatively spliced genes by quantitative PCR (QPCR) applying different primer pairs. As a model system we used rat decay accelerating factor (DAF; CD55) mRNA, which comprises three different isoforms: soluble (sDAF), transmembrane (tmDAF) and glycosyl-phosphatidylinositol (GPI) anchored (gpiDAF) forms. The first step was to prepare solid phase specific for each mRNA isoform and purify the three DAF-forms from total RNA. We then assessed amplification efficiency of primer pairs designed to recognise each of the isoforms using equimolar amounts of the three purified DAF mRNAs. The final step in our assay was to compare expression of the three DAF-isoforms in testis by QPCR taking into account the efficiency of their amplification to enable quantification. The RNA capture/QPCR method we described here can be used for quantifying the expression ratios of alternatively spliced mRNAs from a single gene or for direct comparison of expression of different genes.
Collapse
Affiliation(s)
- Rossen M Donev
- Department of Medical Biochemistry and Immunology, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | | |
Collapse
|
36
|
Gamba G. Molecular Physiology and Pathophysiology of Electroneutral Cation-Chloride Cotransporters. Physiol Rev 2005; 85:423-93. [PMID: 15788703 DOI: 10.1152/physrev.00011.2004] [Citation(s) in RCA: 579] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Electroneutral cation-Cl−cotransporters compose a family of solute carriers in which cation (Na+or K+) movement through the plasma membrane is always accompanied by Cl−in a 1:1 stoichiometry. Seven well-characterized members include one gene encoding the thiazide-sensitive Na+−Cl−cotransporter, two genes encoding loop diuretic-sensitive Na+−K+−2Cl−cotransporters, and four genes encoding K+−Cl−cotransporters. These membrane proteins are involved in several physiological activities including transepithelial ion absorption and secretion, cell volume regulation, and setting intracellular Cl−concentration below or above its electrochemical potential equilibrium. In addition, members of this family play an important role in cardiovascular and neuronal pharmacology and pathophysiology. Some of these cotransporters serve as targets for loop diuretics and thiazide-type diuretics, which are among the most commonly prescribed drugs in the world, and inactivating mutations of three members of the family cause inherited diseases such as Bartter's, Gitelman's, and Anderman's diseases. Major advances have been made in the past decade as consequences of molecular identification of all members in this family. This work is a comprehensive review of the knowledge that has evolved in this area and includes molecular biology of each gene, functional properties of identified cotransporters, structure-function relationships, and physiological and pathophysiological roles of each cotransporter.
Collapse
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
37
|
Watson RM, Griaznova OI, Long CM, Holland MJ. Increased sample capacity for genotyping and expression profiling by kinetic polymerase chain reaction. Anal Biochem 2005; 329:58-67. [PMID: 15136167 DOI: 10.1016/j.ab.2004.02.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2003] [Indexed: 11/28/2022]
Abstract
We fabricated and evaluated high-throughput kinetic thermal cyclers with 768-reaction capacity for kinetic polymerase chain reaction (kPCR)-based genotyping and kinetic reverse transcription (kRT)-PCR-based transcript quantitation. The system uses dye-based detection with ethidium bromide and a single DNA polymerase-based PCR or RT-PCR assay. Allele-specific detection of the two most common hereditary hemochromotosis mutant alleles, C282Y and H63D, was reliably measured by kPCR using human DNA templates as low as 10 genome equivalents per assay. Transcript profiling was performed for 16 yeast transcripts ranging in intracellular abundance over four orders of magnitude. Standard deviations of the PCR cycle threshold values determined from multiple kRT-PCR assays in three different instruments ranged from 0.11 to 0.97 PCR cycles and were reproducible, transcript specific, and instrument independent. The effects of the sin3, gal11, and snf2 knockout mutations on expression of 385 yeast genes were evaluated by kRT-PCR and compared to published values determined by high-density oligonucleotide array and/or microarray analysis for snf2 and sin3. The 768-reaction kinetic thermalcyclers, each with a capacity for more than a half million assays per year, are well suited to genomics applications such as single nucleotide polymorphism/disease association studies and genomewide transcription profiling where high sensitivity and accuracy are required.
Collapse
Affiliation(s)
- Robert M Watson
- Program in Core Research, Roche Molecular Systems, Inc., 1145 Atlantic Avenue, Alameda, CA 94501, USA
| | | | | | | |
Collapse
|
38
|
Flatman PW. Regulation of Na-K-2Cl cotransport in red cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 559:77-88. [PMID: 18727229 DOI: 10.1007/0-387-23752-6_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Affiliation(s)
- Peter W Flatman
- Membrane Biology Group, College of Medicine and Veterinary Medicine, The University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD Scotland, UK.
| |
Collapse
|
39
|
Expression and function of chloride transporters during development of inhibitory neurotransmission in the auditory brainstem. J Neurosci 2003. [PMID: 12764101 DOI: 10.1523/jneurosci.23-10-04134.2003] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glycine and GABA, the dominant inhibitory neurotransmitters in the CNS, assume a depolarizing role in early development, leading to increased cytoplasmic Ca2+ levels and action potentials. The effect is thought to be of some significance for maturation. The depolarization is caused by Cl- efflux, and chloride transporters contribute to the phenomenon by raising the intracellular Cl- concentration ([Cl-]i) above equilibrium, thereby generating an outward-directed electrochemical gradient for Cl-. In mature neurons, the [Cl-]i is reduced below equilibrium, thus rendering glycine activity hyperpolarizing. Here, we investigated the temporal expression of the K-Cl cotransporter KCC2 and the Na-K-Cl cotransporter NKCC1 in the lateral superior olive (LSO) of rats and mice. The two cation cotransporters normally extrude and accumulate Cl-, respectively. As evidenced by several methods, KCC2 mRNA was present in LSO neurons during both the depolarizing and hyperpolarizing periods. Western blots confirmed a constant level of KCC2 in the brainstem, and immunohistochemistry showed that the protein is diffusely distributed within neonatal LSO neurons, becoming integrated into the plasma membrane only with increasing age. The glycine reversal potential in KCC2 knock-out mice differed significantly from that determined in wild-type controls at postnatal day 12 (P12) but not at P3, demonstrating that KCC2 is not active in neonates, despite its early presence. NKCC1 mRNA was not detected during the depolarizing phase in the LSO, implying that this transporter does not contribute to the high [Cl-]i. Our results reveal major differences in the development of [Cl-]i regulation mechanisms seen in brainstem versus forebrain regions.
Collapse
|