1
|
Henderson SW, Nourmohammadi S, Hrmova M. Protein Structural Modeling and Transport Thermodynamics Reveal That Plant Cation-Chloride Cotransporters Mediate Potassium-Chloride Symport. Int J Mol Sci 2024; 25:12955. [PMID: 39684666 DOI: 10.3390/ijms252312955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Plant cation-chloride cotransporters (CCCs) are proposed to be Na+-K+-2Cl- transporting membrane proteins, although evolutionarily, they associate more closely with K+-Cl- cotransporters (KCCs). Here, we investigated grapevine (Vitis vinifera L.) VvCCC using 3D protein modeling, bioinformatics, and electrophysiology with a heterologously expressed protein. The 3D protein modeling revealed that the signatures of ion binding sites in plant CCCs resembled those of animal KCCs, which was supported by phylogenomic analyses and ancestral sequence reconstruction. The conserved features of plant CCCs and animal KCCs included predicted K+ and Cl--binding sites and the absence of a Na+-binding site. Measurements with VvCCC-injected Xenopus laevis oocytes with VvCCC localizing to plasma membranes indicated that the oocytes had depleted intracellular Cl- and net 86Rb fluxes, which agreed with thermodynamic predictions for KCC cotransport. The 86Rb uptake by VvCCC-injected oocytes was Cl--dependent, did not require external Na+, and was partially inhibited by the non-specific CCC-blocker bumetanide, implying that these properties are typical of KCC transporters. A loop diuretic-insensitive Na+ conductance in VvCCC-injected oocytes may account for earlier observations of Na+ uptake by plant CCC proteins expressed in oocytes. Our data suggest plant CCC membrane proteins are likely to function as K+-Cl- cotransporters, which opens the avenues to define their biophysical properties and roles in plant physiology.
Collapse
Affiliation(s)
- Sam W Henderson
- School of Agriculture, Food and Wine, Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Waite Campus Precinct, Glen Osmond, Adelaide, SA 5064, Australia
| | - Saeed Nourmohammadi
- School of Agriculture, Food and Wine, Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Waite Campus Precinct, Glen Osmond, Adelaide, SA 5064, Australia
| | - Maria Hrmova
- School of Agriculture, Food and Wine, Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Waite Campus Precinct, Glen Osmond, Adelaide, SA 5064, Australia
| |
Collapse
|
2
|
Dzhala VI, Mail M, Staley KJ. Timing of interventions to control neuronal chloride elevation in a model of neonatal seizures after hippocampal injury. Epilepsia 2024; 65:3391-3405. [PMID: 39212418 PMCID: PMC11573644 DOI: 10.1111/epi.18108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Following hypoxic-ischemic (HI) brain injury, neuronal cytoplasmic chloride concentration ([Cl-]i) increases, potentially contributing to depolarizing γ-aminobutyric acid (GABA) responses, onset of seizures, and the failure of antiepileptic drugs that target inhibitory chloride-permeable GABAA receptors. Post-HI seizures characteristically begin hours after injury, by which time substantial accumulation of [Cl-]i may have already occurred. In immature neurons, a major pathway for Cl- influx is the reversible Na+-K+-2Cl- cotransporter NKCC1. METHODS Spontaneous neuronal network, neuronal [Cl-]i, and GABA activity were determined in hippocampal preparations from neonatal Clomeleon and SuperClomeleon/DLX-cre mice to test whether blocking NKCC1 earlier after oxygen-glucose deprivation (OGD) injury would more effectively ameliorate the increase in [Cl-]i, ictallike epileptiform discharges (ILDs), and the failure of the GABAergic anticonvulsant phenobarbital. RESULTS In vitro, murine intact hippocampi were free of ILDs for 12 h after preparation. Transient OGD resulted in a gradual increase in [Cl-]i, depolarizing action of GABA, and facilitation of neuronal network activity. Spontaneous ILDs began 3-5 h after injury. Blocking NKCC1 with 2-10 μmol·L-1 bumetanide reduced [Cl-]i equally well when applied up to 10 h after injury. Whereas phenobarbital or bumetanide applied separately were less effective when applied later after injury, ILDs were successfully suppressed by the combination of phenobarbital and bumetanide regardless of the number of prior ILDs or delay in application. SIGNIFICANCE The present age-specific group studies demonstrate that after OGD, NKCC1 transport activity significantly contributes to progressive [Cl-]i accumulation, depolarizing action of GABA, and delayed onset of ILDs. In this neonatal model of neuronal injury and ILDs, earlier treatment with bumetanide alone more efficiently recovered control baseline [Cl-]i and depressed epileptiform discharges. However, there was no time dependency to the anti-ictal efficacy of the combination of phenobarbital and bumetanide. These in vitro results suggest that after perinatal injury, early pre-emptive treatment with phenobarbital plus bumetanide would be as efficacious as late treatment after seizures are manifest.
Collapse
Affiliation(s)
- Volodymyr I Dzhala
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michelle Mail
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin J Staley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Bahari F, Dzhala V, Balena T, Lillis KP, Staley KJ. Intraventricular haemorrhage in premature infants: the role of immature neuronal salt and water transport. Brain 2024; 147:3216-3233. [PMID: 38815055 PMCID: PMC11370806 DOI: 10.1093/brain/awae161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 06/01/2024] Open
Abstract
Intraventricular haemorrhage is a common complication of premature birth. Survivors are often left with cerebral palsy, intellectual disability and/or hydrocephalus. Animal models suggest that brain tissue shrinkage, with subsequent vascular stretch and tear, is an important step in the pathophysiology, but the cause of this shrinkage is unknown. Clinical risk factors for intraventricular haemorrhage are biomarkers of hypoxic-ischaemic stress, which causes mature neurons to swell. However, immature neuronal volume might shift in the opposite direction in these conditions. This is because immature neurons express the chloride, salt and water transporter NKCC1, which subserves regulatory volume increases in non-neural cells, whereas mature neurons express KCC2, which subserves regulatory volume decreases. When hypoxic-ischaemic conditions reduce active ion transport and increase the cytoplasmic membrane permeability, the effects of these transporters are diminished. Consequentially, mature neurons swell (cytotoxic oedema), whereas immature neurons might shrink. After hypoxic-ischaemic stress, in vivo and in vitro multi-photon imaging of perinatal transgenic mice demonstrated shrinkage of viable immature neurons, bulk tissue shrinkage and blood vessel displacement. Neuronal shrinkage was correlated with age-dependent membrane salt and water transporter expression using immunohistochemistry. Shrinkage of immature neurons was prevented by prior genetic or pharmacological inhibition of NKCC1 transport. These findings open new avenues of investigation for the detection of acute brain injury by neuroimaging, in addition to prevention of neuronal shrinkage and the ensuing intraventricular haemorrhage, in premature infants.
Collapse
Affiliation(s)
- Fatemeh Bahari
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Volodymyr Dzhala
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Trevor Balena
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Kyle P Lillis
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin J Staley
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Chattopadhayay S, Wanjari P, Talukdar P. Acylhydrazone-based reversibly photoswitchable ion pair transporter with OFF-ON cotransport activity. Chem Sci 2024; 15:d4sc02474e. [PMID: 39355225 PMCID: PMC11440441 DOI: 10.1039/d4sc02474e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
The cellular membrane transport of physiologically important cations and anions is omnipresent and regulates different physiological functions. Whereas a notable number of cation-anion transporters are being developed to transport salts across the membrane, developing an artificial cation-anion symporter with stimulus-responsive activities is an immense obstacle. Herein, for the first time, we report reversibly photoswitchable acylhydrazone-based transporter 2 that has distinctive OFF-ON cation-anion co-transport abilities. The substituent was modified in 1a-1c and 2, to change the to-and-fro movement of the transporter to enhance the ion transport efficiency. Ion transport experiments across the lipid bilayer membrane demonstrate that 1a has the highest transport activity among the series with irreversible photoisomerization properties, whereas 2 has a unique reversible photoisomerization property. A detailed transport study indicated that the E-conformer of compound 2 facilitates Na+/Cl- transport via the symport process by following the carrier mode of ion transport. 23Na NMR and chloride selective electrode assays confirmed the OFF and ON state of ion transport of compound 2 with photoirradiation. An assembly of [(2 E )2 + NaCl] was subjected to geometry optimization to understand the responsible ion binding motif. Geometry optimization followed by the natural bond orbital analysis of 1a Z and 2 Z demonstrated that 1a Z forms comparatively stronger intramolecular H-bonding than 2 Z , making it accessible for reversible photoisomerization.
Collapse
Affiliation(s)
- Sandip Chattopadhayay
- Department of Chemistry, Indian Institute of Science Education and Research Pune Dr Homi Bhabha Road Pashan Pune 411008 Maharashtra India
| | - Paras Wanjari
- Department of Chemistry, Indian Institute of Science Education and Research Pune Dr Homi Bhabha Road Pashan Pune 411008 Maharashtra India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune Dr Homi Bhabha Road Pashan Pune 411008 Maharashtra India
| |
Collapse
|
5
|
Shao B, Fu H, Aprahamian I. A molecular anion pump. Science 2024; 385:544-549. [PMID: 39088617 DOI: 10.1126/science.adp3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/02/2024] [Indexed: 08/03/2024]
Abstract
Pumping ions against a concentration gradient through protein-based transporters is a cornerstone of numerous biological processes. Mimicking this function by using artificial receptors remains a daunting challenge, mainly because of the difficulties in balancing between the requirement for high binding affinities and precise and on-demand ion capture and release properties. We report a trimeric hydrazone photoswitch-based receptor that converts light energy into work by actively transporting chloride anion against a gradient through a dichloromethane liquid membrane, functioning as a molecular pump. The system manifests ease of synthesis, bistability, excellent photoswitching properties, and superb ON-OFF binding properties (difference of up to six orders of magnitude).
Collapse
Affiliation(s)
- Baihao Shao
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Heyifei Fu
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Ivan Aprahamian
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
6
|
Erickson CA, Perez-Cano L, Pedapati EV, Painbeni E, Bonfils G, Schmitt LM, Sachs H, Nelson M, De Stefano L, Westerkamp G, de Souza ALS, Pohl O, Laufer O, Issachar G, Blaettler T, Hyvelin JM, Durham LA. Safety, Tolerability, and EEG-Based Target Engagement of STP1 (PDE3,4 Inhibitor and NKCC1 Antagonist) in a Randomized Clinical Trial in a Subgroup of Patients with ASD. Biomedicines 2024; 12:1430. [PMID: 39062003 PMCID: PMC11274259 DOI: 10.3390/biomedicines12071430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
This study aimed to evaluate the safety and tolerability of STP1, a combination of ibudilast and bumetanide, tailored for the treatment of a clinically and biologically defined subgroup of patients with Autism Spectrum Disorder (ASD), namely ASD Phenotype 1 (ASD-Phen1). We conducted a randomized, double-blind, placebo-controlled, parallel-group phase 1b study with two 14-day treatment phases (registered at clinicaltrials.gov as NCT04644003). Nine ASD-Phen1 patients were administered STP1, while three received a placebo. We assessed safety and tolerability, along with electrophysiological markers, such as EEG, Auditory Habituation, and Auditory Chirp Synchronization, to better understand STP1's mechanism of action. Additionally, we used several clinical scales to measure treatment outcomes. The results showed that STP1 was well-tolerated, with electrophysiological markers indicating a significant and dose-related reduction of gamma power in the whole brain and in brain areas associated with executive function and memory. Treatment with STP1 also increased alpha 2 power in frontal and occipital regions and improved habituation and neural synchronization to auditory chirps. Although numerical improvements were observed in several clinical scales, they did not reach statistical significance. Overall, this study suggests that STP1 is well-tolerated in ASD-Phen1 patients and shows indirect target engagement in ASD brain regions of interest.
Collapse
Affiliation(s)
- Craig A. Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Laura Perez-Cano
- Discovery and Data Science (DDS) Unit, STALICLA SL, Moll de Barcelona, s/n, Edif Este, 08039 Barcelona, Spain
| | - Ernest V. Pedapati
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Child Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Eric Painbeni
- Drug Development Unit (DDU), STALICLA SA, Campus Biotech Innovation Park, Avenue de Sécheron 15, 1202 Geneva, Switzerland
| | - Gregory Bonfils
- Drug Development Unit (DDU), STALICLA SA, Campus Biotech Innovation Park, Avenue de Sécheron 15, 1202 Geneva, Switzerland
| | - Lauren M. Schmitt
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Hannah Sachs
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Meredith Nelson
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lisa De Stefano
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Grace Westerkamp
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Adriano L. S. de Souza
- Drug Development Unit (DDU), STALICLA SA, Campus Biotech Innovation Park, Avenue de Sécheron 15, 1202 Geneva, Switzerland
| | - Oliver Pohl
- Drug Development Unit (DDU), STALICLA SA, Campus Biotech Innovation Park, Avenue de Sécheron 15, 1202 Geneva, Switzerland
| | | | | | - Thomas Blaettler
- Drug Development Unit (DDU), STALICLA SA, Campus Biotech Innovation Park, Avenue de Sécheron 15, 1202 Geneva, Switzerland
| | - Jean-Marc Hyvelin
- Drug Development Unit (DDU), STALICLA SA, Campus Biotech Innovation Park, Avenue de Sécheron 15, 1202 Geneva, Switzerland
| | - Lynn A. Durham
- Drug Development Unit (DDU), STALICLA SA, Campus Biotech Innovation Park, Avenue de Sécheron 15, 1202 Geneva, Switzerland
| |
Collapse
|
7
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Löscher W, Gramer M, Römermann K. Heterogeneous brain distribution of bumetanide following systemic administration in rats. Biopharm Drug Dispos 2024; 45:138-148. [PMID: 38823029 DOI: 10.1002/bdd.2390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/19/2024] [Accepted: 05/16/2024] [Indexed: 06/03/2024]
Abstract
Bumetanide is used widely as a tool and off-label treatment to inhibit the Na-K-2Cl cotransporter NKCC1 in the brain and thereby to normalize intra-neuronal chloride levels in several brain disorders. However, following systemic administration, bumetanide only poorly penetrates into the brain parenchyma and does not reach levels sufficient to inhibit NKCC1. The low brain penetration is a consequence of both the high ionization rate and plasma protein binding, which restrict brain entry by passive diffusion, and of brain efflux transport. In previous studies, bumetanide was determined in the whole brain or a few brain regions, such as the hippocampus. However, the blood-brain barrier and its efflux transporters are heterogeneous across brain regions, so it cannot be excluded that bumetanide reaches sufficiently high brain levels for NKCC1 inhibition in some discrete brain areas. Here, bumetanide was determined in 14 brain regions following i.v. administration of 10 mg/kg in rats. Because bumetanide is much more rapidly eliminated by rats than humans, its metabolism was reduced by pretreatment with piperonyl butoxide. Significant, up to 5-fold differences in regional bumetanide levels were determined with the highest levels in the midbrain and olfactory bulb and the lowest levels in the striatum and amygdala. Brain:plasma ratios ranged between 0.004 (amygdala) and 0.022 (olfactory bulb). Regional brain levels were significantly correlated with local cerebral blood flow. However, regional bumetanide levels were far below the IC50 (2.4 μM) determined previously for rat NKCC1. Thus, these data further substantiate that the reported effects of bumetanide in rodent models of brain disorders are not related to NKCC1 inhibition in the brain.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Translational Neuropharmacology Laboratory, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Martina Gramer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
9
|
Bovee S, Klump GM, Köppl C, Pyott SJ. The Stria Vascularis: Renewed Attention on a Key Player in Age-Related Hearing Loss. Int J Mol Sci 2024; 25:5391. [PMID: 38791427 PMCID: PMC11121695 DOI: 10.3390/ijms25105391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Age-related hearing loss (HL), or presbycusis, is a complex and heterogeneous condition, affecting a significant portion of older adults and involving various interacting mechanisms. Metabolic presbycusis, a type of age-related HL, is characterized by the dysfunction of the stria vascularis, which is crucial for maintaining the endocochlear potential necessary for hearing. Although attention on metabolic presbycusis has waned in recent years, research continues to identify strial pathology as a key factor in age-related HL. This narrative review integrates past and recent research, bridging findings from animal models and human studies, to examine the contributions of the stria vascularis to age-related HL. It provides a brief overview of the structure and function of the stria vascularis and then examines mechanisms contributing to age-related strial dysfunction, including altered ion transport, changes in pigmentation, inflammatory responses, and vascular atrophy. Importantly, this review outlines the contribution of metabolic mechanisms to age-related HL, highlighting areas for future research. It emphasizes the complex interdependence of metabolic and sensorineural mechanisms in the pathology of age-related HL and highlights the importance of animal models in understanding the underlying mechanisms. The comprehensive and mechanistic investigation of all factors contributing to age-related HL, including cochlear metabolic dysfunction, remains crucial to identifying the underlying mechanisms and developing personalized, protective, and restorative treatments.
Collapse
Affiliation(s)
- Sonny Bovee
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany; (S.B.); (G.M.K.); (C.K.)
| | - Georg M. Klump
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany; (S.B.); (G.M.K.); (C.K.)
- Cluster of Excellence “Hearing4all”, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Research Centre Neurosensory Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Christine Köppl
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany; (S.B.); (G.M.K.); (C.K.)
- Cluster of Excellence “Hearing4all”, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Research Centre Neurosensory Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Sonja J. Pyott
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- The Research School of Behavioural and Cognitive Neurosciences, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
10
|
Dhapola R, Medhi B, HariKrishnaReddy D. Insight into the pathophysiological advances and molecular mechanisms underlying cerebral stroke: current status. Mol Biol Rep 2024; 51:649. [PMID: 38733445 DOI: 10.1007/s11033-024-09597-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
Molecular pathways involved in cerebral stroke are diverse. The major pathophysiological events that are observed in stroke comprises of excitotoxicity, oxidative stress, mitochondrial damage, endoplasmic reticulum stress, cellular acidosis, blood-brain barrier disruption, neuronal swelling and neuronal network mutilation. Various biomolecules are involved in these pathways and several major proteins are upregulated and/or suppressed following stroke. Different types of receptors, ion channels and transporters are activated. Fluctuations in levels of various ions and neurotransmitters have been observed. Cells involved in immune responses and various mediators involved in neuro-inflammation get upregulated progressing the pathogenesis of the disease. Despite of enormity of the problem, there is not a single therapy that can limit infarction and neurological disability due to stroke. This is because of poor understanding of the complex interplay between these pathophysiological processes. This review focuses upon the past to present research on pathophysiological events that are involved in stroke and various factors that are leading to neuronal death following cerebral stroke. This will pave a way to researchers for developing new potent therapeutics that can aid in the treatment of cerebral stroke.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
11
|
Laghmani K. Protein Quality Control of NKCC2 in Bartter Syndrome and Blood Pressure Regulation. Cells 2024; 13:818. [PMID: 38786040 PMCID: PMC11120568 DOI: 10.3390/cells13100818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Mutations in NKCC2 generate antenatal Bartter syndrome type 1 (type 1 BS), a life-threatening salt-losing nephropathy characterized by arterial hypotension, as well as electrolyte abnormalities. In contrast to the genetic inactivation of NKCC2, inappropriate increased NKCC2 activity has been associated with salt-sensitive hypertension. Given the importance of NKCC2 in salt-sensitive hypertension and the pathophysiology of prenatal BS, studying the molecular regulation of this Na-K-2Cl cotransporter has attracted great interest. Therefore, several studies have addressed various aspects of NKCC2 regulation, such as phosphorylation and post-Golgi trafficking. However, the regulation of this cotransporter at the pre-Golgi level remained unknown for years. Similar to several transmembrane proteins, export from the ER appears to be the rate-limiting step in the cotransporter's maturation and trafficking to the plasma membrane. The most compelling evidence comes from patients with type 5 BS, the most severe form of prenatal BS, in whom NKCC2 is not detectable in the apical membrane of thick ascending limb (TAL) cells due to ER retention and ER-associated degradation (ERAD) mechanisms. In addition, type 1 BS is one of the diseases linked to ERAD pathways. In recent years, several molecular determinants of NKCC2 export from the ER and protein quality control have been identified. The aim of this review is therefore to summarize recent data regarding the protein quality control of NKCC2 and to discuss their potential implications in BS and blood pressure regulation.
Collapse
Affiliation(s)
- Kamel Laghmani
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France;
- CNRS, ERL8228, F-75006 Paris, France
| |
Collapse
|
12
|
Shivankar BR, Bhandare VV, Joshi K, Patil VS, Dhotare PS, Sonawane KD, Krishnamurty S. Investigation of cathinone analogs targeting human dopamine transporter using molecular modeling. J Biomol Struct Dyn 2024:1-16. [PMID: 38698732 DOI: 10.1080/07391102.2024.2335303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/20/2024] [Indexed: 05/05/2024]
Abstract
In a step towards understanding the structure-property relationship among Synthetic Cathinones (SCs), a combined methodology based on Density Functional Theory (DFT), Administration, Distribution, Metabolism, Excretion, and Toxicity (ADMET) predictions, docking and molecular dynamics simulations have been applied to correlate physicochemical descriptors of various SCs to their biological activity. The results from DFT and molecular docking studies correlate well with each other explaining the biological activity trends of the studied SCs. Quantum mechanical descriptors viz. polarizability, electron affinity, ionization potential, chemical hardness, electronegativity, molecular electrostatic potential, and ion interaction studies unravel the distinguishingly reactive nature of Group D (pyrrolidine substituted) and Group E (methylenedioxy and pyrrolidine substituted) compounds. According to ADMET analysis, Group D and Group E molecules have a higher probability of permeating through the blood-brain barrier. Molecular docking results indicate that Phe76, Ala77, Asp79, Val152, Tyr156, Phe320, and Phe326 constitute the binding pocket residues of hDAT in which the most active ligands MDPV, MDPBP, and MDPPP are bound. Finally, to validate the derived quantum chemical descriptors and docking results, Molecular Dynamics (MD) simulations are performed with homology-modelled hDAT (human dopamine transporter). The MD simulation results revealed that the majority of SCs remain stable within the hDAT protein's active sites via non-bonded interactions after 100 ns long simulations. The findings from DFT, ADMET analysis, molecular docking, and molecular dynamics simulation studies complement each other suggesting that pyrrolidine-substituted SCs (Group D and E), specifically, MPBP and PVN are proven potent SCs along with MDPV, validating various experimental observations.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Bhavana R Shivankar
- Physical Chemistry Division, CSIR-National Chemical Laboratory, Pune, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - Krati Joshi
- Physical Chemistry Division, CSIR-National Chemical Laboratory, Pune, India
| | - Vishal S Patil
- Department of Pharmacology and Toxicology, KLE College of Pharmacy Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi, India
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, India
| | | | | | - Sailaja Krishnamurty
- Physical Chemistry Division, CSIR-National Chemical Laboratory, Pune, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
13
|
Stritzke A, Ismail R, Rose MS, Lyon AW, Fenton TR. Cord-Blood Derived Chemistry Reference Values in Preterm Infants for Sodium, Chloride, Potassium, Glucose, and Creatinine. Am J Perinatol 2024; 41:722-729. [PMID: 34983069 DOI: 10.1055/a-1730-8536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
OBJECTIVE International guidelines recommend that preterm infants should be supported to maintain their serum electrolytes within "normal" ranges. In term babies, cord blood values differed in pathological pregnancies from healthy ones. STUDY DESIGN We examined cord blood sodium, chloride, potassium, glucose, and creatinine to derive maturity-related reference intervals. We examined associations with gestational age, delivery mode, singleton versus multiple, and prenatal maternal adverse conditions. We compared preterm cord values to term, and to adult reference ranges. RESULTS There were 591 infants, 537 preterm and 54 term. Preterm cord glucose levels were steady (3.7 ± 1.1 mmol/L), while sodium, chloride, and creatinine increased over GA by 0.17, 0.14 mmol/L/week, and 1.07 µmol/L/week, respectively (p < 0.003). Average preterm cord potassium and chloride were higher than the term (p < 0.05). Compared with adult reference intervals, cord preterm reference intervals were higher for chloride (100-111 vs. 98-106 mmol/L), lower for creatinine (29-84 vs. 62-115 µmol/L), and more variable for potassium (2.7-7.9 vs. 3.5-5.0 mmol/L) and sodium (130-141 vs. 136-145 mmol/L). Cesarean section was associated with higher potassium and lower glucose, multiple births with higher chloride and creatinine and lower glucose, and SGA with lower glucose. CONCLUSION Cord blood values varied across the GA range with increases in sodium, chloride, and creatinine, while glucose remained steady. Average preterm reference values were higher than term values for potassium and chloride. Preterm reference values differed from published adults' reference values. The changes across GA and by delivery mode, SGA, and being a multiple, which may have direct implications for neonatal care and fluid management. KEY POINTS · Cord blood electrolyte, creatinine, and glucose values vary across neonatal gestational age.. · Average preterm cord values of potassium and chloride were higher than term values.. · Cord reference values differ by delivery mode, growth, and multiple impacting neonatal care decisions..
Collapse
Affiliation(s)
- Amelie Stritzke
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Rana Ismail
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - M Sarah Rose
- Research Excellence Support Team, Alberta Health Services, Calgary, Calgary, Canada
| | - Andrew W Lyon
- Pathology and Laboratory Medicine, Saint Pauls Hospital Laboratory, Saskatoon, Saskatchewan, Canada
| | - Tanis R Fenton
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
- Community Health Sciences, University of Calgary, Calgary, Canada
- Nutrition Services, Alberta Health Services, Calgary, Canada
| |
Collapse
|
14
|
Mucciolo S, Desiderato A, Mastrodonato M, Lana P, Arruda Freire C, Prodocimo V. First Insights into Body Localization of an Osmoregulation-Related Cotransporter in Estuarine Annelids. BIOLOGY 2024; 13:235. [PMID: 38666847 PMCID: PMC11048583 DOI: 10.3390/biology13040235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024]
Abstract
The expression of the Na+-K+-2Cl- cotransporter (NKCC), widely associated with cell volume regulation, has never been directly demonstrated in annelids. Its putative presence was firstly recovered in silico, and then using immunofluorescence, its signal was retrieved for the first time in different tissues of four species of estuarine annelids from southern Brazil that are regularly subjected to salinity fluctuations. We tested two euryhaline species (wide salinity tolerance), the nereidids Alitta yarae and Laeonereis acuta (habitat salinity: ~10-28 psu), and two stenohaline species (restricted salinity tolerance), the nephtyid Nephtys fluviatilis (habitat salinity: ~6-10 psu), and the melinnid Isolda pulchella (habitat salinity: ~28-35 psu). All four species showed specific immunofluorescent labelling for NKCC-like expression. However, the expression of an NKCC-like protein was not homogeneous among them. The free-living/burrowers (both euryhaline nereidids and the stenohaline nephtyid) displayed a widespread signal for an NKCC-like protein along their bodies, in contrast to the stenohaline sedentary melinnid, in which the signal was restricted to the branchiae and the internal tissues of the body. The results are compatible with NKCC involvement in cell volume, especially in annelids that face wide variations in salinity in their habitats.
Collapse
Affiliation(s)
- Serena Mucciolo
- Department of Invertebrate Zoology and Hydrobiology, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
- Laboratório de Bentos, Centro de Estudos do Mar, Universidade Federal do Paraná, Av. Beira Mar s/n, Pontal do Paraná 83255-976, Paraná, Brazil;
| | - Andrea Desiderato
- Department of Invertebrate Zoology and Hydrobiology, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| | - Maria Mastrodonato
- Dipartimento di Bioscienze Biotecnologie e Ambiente, Campus Universitario “E. Quagliariello”, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy;
| | - Paulo Lana
- Laboratório de Bentos, Centro de Estudos do Mar, Universidade Federal do Paraná, Av. Beira Mar s/n, Pontal do Paraná 83255-976, Paraná, Brazil;
| | - Carolina Arruda Freire
- Laboratório de Fisiologia Comparativa de Osmorregulação, Departamento de Fisiologia, Setor de Ciências Biológicas, Campus Politécnico, Universidade Federal do Paraná, Av. Cel. Francisco H. dos Santos 100, Curitiba 81530-000, Paraná, Brazil; (C.A.F.); (V.P.)
| | - Viviane Prodocimo
- Laboratório de Fisiologia Comparativa de Osmorregulação, Departamento de Fisiologia, Setor de Ciências Biológicas, Campus Politécnico, Universidade Federal do Paraná, Av. Cel. Francisco H. dos Santos 100, Curitiba 81530-000, Paraná, Brazil; (C.A.F.); (V.P.)
| |
Collapse
|
15
|
Lu W, Wen J. Neuroinflammation and Post-Stroke Depression: Focus on the Microglia and Astrocytes. Aging Dis 2024:AD.2024.0214-1. [PMID: 38421829 DOI: 10.14336/ad.2024.0214-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
Post-stroke depression (PSD), a frequent and disabling complication of stroke, has a strong impact on almost thirty percent of stroke survivors. The pathogenesis of PSD is not completely clear so far. Neuroinflammation following stroke is one of underlying mechanisms that involves in the pathophysiology of PSD and plays an important function in the development of depression and is regarded as a sign of depression. During the neuroinflammation after ischemic stroke onset, both astrocytes and microglia undergo a series of morphological and functional changes and play pro-inflammatory or anti-inflammatory effect in the pathological process of stroke. Importantly, astrocytes and microglia exert dual roles in the pathological process of PSD due to the phenotypic transformation. We summarize the latest evidence of neuroinflammation involving in PSD in this review, focus on the phenotypic transformation of microglia and astrocytes following ischemic stroke and reveal the dual roles of both microglia and astrocytes in the PSD via modulating the neuroinflammation.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
16
|
Takeyasu M, Kozai K, Sugita M. Involvement of sodium-glucose cotransporter-1 activities in maintaining oscillatory Cl - currents from mouse submandibular acinar cells. J Comp Physiol B 2024; 194:21-32. [PMID: 38308715 PMCID: PMC10940492 DOI: 10.1007/s00360-024-01532-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024]
Abstract
In salivary acinar cells, cholinergic stimulation induces elevations of cytosolic [Ca2+]i to activate the apical exit of Cl- through TMEM16A Cl- channels, which acts as a driving force for fluid secretion. To sustain the Cl- secretion, [Cl-]i must be maintained to levels that are greater than the electrochemical equilibrium mainly by Na+-K+-2Cl- cotransporter-mediated Cl- entry in basolateral membrane. Glucose transporters carry glucose into the cytoplasm, enabling the cells to produce ATP to maintain Cl- and fluid secretion. Sodium-glucose cotransporter-1 is a glucose transporter highly expressed in acinar cells. The salivary flow is suppressed by the sodium-glucose cotransporter-1 inhibitor phlorizin. However, it remains elusive how sodium-glucose cotransporter-1 contributes to maintaining salivary fluid secretion. To examine if sodium-glucose cotransporter-1 activity is required for sustaining Cl- secretion to drive fluid secretion, we analyzed the Cl- currents activated by the cholinergic agonist, carbachol, in submandibular acinar cells while comparing the effect of phlorizin on the currents between the whole-cell patch and the gramicidin-perforated patch configurations. Phlorizin suppressed carbachol-induced oscillatory Cl- currents by reducing the Cl- efflux dependent on the Na+-K+-2Cl- cotransporter-mediated Cl- entry in addition to affecting TMEM16A activity. Our results suggest that the sodium-glucose cotransporter-1 activity is necessary for maintaining the oscillatory Cl- secretion supported by the Na+-K+-2Cl- cotransporter activity in real time to drive fluid secretion. The concerted effort of sodium-glucose cotransporter-1, Na+-K+-2Cl- cotransporter, and apically located Cl- channels might underlie the efficient driving of Cl- secretion in different secretory epithelia from a variety of animal species.
Collapse
Affiliation(s)
- Misa Takeyasu
- Department of Physiology and Oral Physiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8553, Japan
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Katsuyuki Kozai
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Makoto Sugita
- Department of Physiology and Oral Physiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8553, Japan.
| |
Collapse
|
17
|
Yang C, Wang Y, Li Y, Wang X, Hua W, Yang Z, Wang H. Sub-dose anesthetics combined with chloride regulators protect the brain against chronic ischemia-hypoxia injury. CNS Neurosci Ther 2024; 30:e14379. [PMID: 37545014 PMCID: PMC10848060 DOI: 10.1111/cns.14379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Cerebral ischemia-hypoxia leads to excitotoxicity-mediated neuronal damage and cognitive dysfunction, especially in the elderly. Excessive intracellular [Cl- ]i accumulation weakens γ-aminobutyric acid (GABA) compensatory effects. Sub-anesthetic dose of propofol protected the brain against ischemia-hypoxia, which was abolished by blocking Cl- efflux transporter K+ /Cl- cotransporter 2 (KCC2). We aimed to determine whether low-dose anesthetic combined with [Cl- ]i regulators could restore the compensatory GABAergic system and improve cognitive function. METHODS Chronic cerebral hypoxia (CCH) model was established by bilateral carotid artery ligation in aged rats. Sub-dose of anesthetics (propofol and sevoflurane) with or without KCC2 agonist N-ethylmaleimide (NEM) or Na+ /K+ /Cl- cotransporter 1 (NKCC1) antagonist bumetanide (BTN) was administered systemically 30 days post-surgery. Primary rat hippocampal neuronal cultures were subjected to hypoxic injury with or without drug treatment. Memory function, hippocampal neuronal survival, GABAergic system functioning, and brain-derived neurotrophic factor (BDNF) expressions were evaluated. RESULTS Sub-anesthetic dose of combined propofol (1.2 μg mL-1 ) and sevoflurane [0.7 MAC (minimum alveolar concentration)] did not aggravate the hypoxic brain injury in rats or cell damage in neuronal cultures. Adding either BTN or NEM protected against hypoxic injury, associated with improved cognitive function in vivo, less intracellular accumulation of [Cl- ]i , reduced cell death, restored GABAergic compensation, and increased BDNF expression both in vivo and in vitro. CONCLUSION Sub-anesthetic dose of propofol and sevoflurane is a recommended anesthesia regimen in at-risk patients. Restoration of [Cl- ]i homeostasis and GABAergic could further reduce the brain damage caused by ischemia-hypoxia.
Collapse
Affiliation(s)
- Chenyi Yang
- Nankai UniversityTianjinChina
- Nankai University Affinity the Third Central HospitalTianjinChina
- The Third Central Clinical College of Tianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical DiseasesTianjinChina
- Artificial Cell Engineering Technology Research CenterTianjinChina
- Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Ye Wang
- The Third Central Clinical College of Tianjin Medical UniversityTianjinChina
| | - Yun Li
- The Third Central Clinical College of Tianjin Medical UniversityTianjinChina
| | - Xinyi Wang
- Nankai University Affinity the Third Central HospitalTianjinChina
- The Third Central Clinical College of Tianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical DiseasesTianjinChina
- Artificial Cell Engineering Technology Research CenterTianjinChina
- Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Wei Hua
- Nankai University Affinity the Third Central HospitalTianjinChina
- The Third Central Clinical College of Tianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical DiseasesTianjinChina
- Artificial Cell Engineering Technology Research CenterTianjinChina
- Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| | | | - Haiyun Wang
- Nankai UniversityTianjinChina
- Nankai University Affinity the Third Central HospitalTianjinChina
- The Third Central Clinical College of Tianjin Medical UniversityTianjinChina
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical DiseasesTianjinChina
- Artificial Cell Engineering Technology Research CenterTianjinChina
- Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| |
Collapse
|
18
|
Xiao L, Yang Q, Tan J, Ma B, Chen D. Engineering a Cl - -Modulated Light-Driven Na + Pump. Chemistry 2023; 29:e202302543. [PMID: 37833829 DOI: 10.1002/chem.202302543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/30/2023] [Accepted: 10/13/2023] [Indexed: 10/15/2023]
Abstract
Microbial Na+ -pumping rhodopsin (NaR) is a promising optogenetic tool due to its unique ability to transport Na+ . Like most rhodopsin-based tools, NaR is limited to light-based control. In this study, our objective was to develop a novel mode of modulation for NaR beyond light control. By introducing a potential Cl- binding site near the putative Na+ release cavity, we engineered Nonlabens dokdonensis rhodopsin 2 (NdR2) to be modulated by Cl- , an essential chemical in organisms. The engineered NdR2 demonstrated an approximately two-fold increase in Na+ pump activity in the presence of 100 mM Cl- compared to Cl- -free solution. Increasing Cl- concentration decreased the lifetimes of the M and O intermediates accordingly. The analysis of competitive ion uptake suggested the bound Cl- may increase the Na+ affinity and selectivity. This chemical modulation allows for more diverse and precise control over cellular processes, advancing the development of next-generation optogenetic tools. Notably, our Cl- -modulated NdR2 establishes an innovative mechanism for linking Cl- to Na+ -related processes, with potential applications in optogenetic therapies for related diseases.
Collapse
Affiliation(s)
- Lan Xiao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qifan Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jingjing Tan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baofu Ma
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Deliang Chen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
19
|
Emeish WFA, Abd-ElHafeez HH, Alghamdi AAA, Ahmed M, Khalifa MO, El-Mansi AA, Abou-Elhamd AS, Khormi MM, Alkashif K, Soliman SA. Morphological changes in intraepithelial and stromal telocytes in Cyprinus carpio in response to salinity stress. Sci Rep 2023; 13:19987. [PMID: 37968439 PMCID: PMC10651833 DOI: 10.1038/s41598-023-43279-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/21/2023] [Indexed: 11/17/2023] Open
Abstract
Telocytes establish connections and communicate with various types of cells and structures. Few experimental studies have been performed on telocytes. In this study, we investigated the effect of salinity stress on telocytes in relation to osmoregulatory, immune, and stem cells. After exposing the common carp to 0.2 (control), 6, 10, or 14 ppt salinity, we extracted and fixed gill samples in glutaraldehyde, processed and embedded the samples in resin, and prepared semi-thin and ultrathin sections. Two types of telocytes were identified: intraepithelial and stromal telocytes. Intraepithelial telocytes were found to form part of the cellular lining of the lymphatic space and shed secretory vesicles into this space. Stromal telocytes were observed to shed their secretory vesicles into the secondary circulatory vessels. Both intraepithelial and stromal telocytes were enlarged and exhibited increased secretory activities as salinity increased. They exerted their effects via direct contact and paracrine signaling. The following changes were observed in samples from fish exposed to high salinity levels: chloride cells underwent hypertrophy, and their mitochondria became cigar-shaped; pavement cells were enlarged, and their micro-ridges became thin and elongated; stromal telocytes established contact with stem cells and skeletal myoblasts; skeletal muscle cells underwent hypertrophy; and macrophages and rodlet cells increased in number. In conclusion, our findings indicate that intraepithelial and stromal telocytes respond to salinity stress by activating cellular signaling and that they play major roles in osmoregulation, immunity, and regeneration.
Collapse
Affiliation(s)
- Walaa F A Emeish
- Department of Fish Diseases, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Hanan H Abd-ElHafeez
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | | | - Madeha Ahmed
- Department of Histology, Faculty of Veterinary Medicine, Sohag University, Sohagt, 82524, Egypt
| | - Mahmoud Osman Khalifa
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
- Department of Molecular Bone Biology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Ahmed A El-Mansi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Alaa S Abou-Elhamd
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
- Department of Respiratory Therapy, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Mohsen M Khormi
- Department of Biology, College of Science, Jazan University, Jazan, Saudi Arabia
| | - Khalid Alkashif
- Physiology Department, faculty of Medicine, Merit University, Sohage, Egypt
| | - Soha A Soliman
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt.
| |
Collapse
|
20
|
Humphreys JM, Teixeira LR, Akella R, He H, Kannangara AR, Sekulski K, Pleinis J, Liwocha J, Jiou J, Servage KA, Orth K, Joachimiak L, Rizo J, Cobb MH, Brautigam CA, Rodan AR, Goldsmith EJ. Hydrostatic Pressure Sensing by WNK kinases. Mol Biol Cell 2023; 34:ar109. [PMID: 37585288 PMCID: PMC10559305 DOI: 10.1091/mbc.e23-03-0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
Previous study has demonstrated that the WNK kinases 1 and 3 are direct osmosensors consistent with their established role in cell-volume control. WNK kinases may also be regulated by hydrostatic pressure. Hydrostatic pressure applied to cells in culture with N2 gas or to Drosophila Malpighian tubules by centrifugation induces phosphorylation of downstream effectors of endogenous WNKs. In vitro, the autophosphorylation and activity of the unphosphorylated kinase domain of WNK3 (uWNK3) is enhanced to a lesser extent than in cells by 190 kPa applied with N2 gas. Hydrostatic pressure measurably alters the structure of uWNK3. Data from size exclusion chromatography in line with multi-angle light scattering (SEC-MALS), SEC alone at different back pressures, analytical ultracentrifugation (AUC), NMR, and chemical crosslinking indicate a change in oligomeric structure in the presence of hydrostatic pressure from a WNK3 dimer to a monomer. The effects on the structure are related to those seen with osmolytes. Potential mechanisms of hydrostatic pressure activation of uWNK3 and the relationships of pressure activation to WNK osmosensing are discussed.
Collapse
Affiliation(s)
- John M. Humphreys
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Liliana R. Teixeira
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Radha Akella
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Haixia He
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ashari R. Kannangara
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kamil Sekulski
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John Pleinis
- Department of Internal Medicine, Division of Nephrology and Hypertension and Department of Human Genetics, University of Utah, Salt Lake City UT 84112
| | - Joanna Liwocha
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jenny Jiou
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kelly A. Servage
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lukasz Joachimiak
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Josep Rizo
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Melanie H. Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Chad A. Brautigam
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Aylin R. Rodan
- Department of Internal Medicine, Division of Nephrology and Hypertension and Department of Human Genetics, University of Utah, Salt Lake City UT 84112
| | - Elizabeth J. Goldsmith
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
21
|
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
|
22
|
Benko F, Urminská D, Ďuračka M, Tvrdá E. Signaling Roleplay between Ion Channels during Mammalian Sperm Capacitation. Biomedicines 2023; 11:2519. [PMID: 37760960 PMCID: PMC10525812 DOI: 10.3390/biomedicines11092519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
In order to accomplish their primary goal, mammalian spermatozoa must undergo a series of physiological, biochemical, and functional changes crucial for the acquisition of fertilization ability. Spermatozoa are highly polarized cells, which must swiftly respond to ionic changes on their passage through the female reproductive tract, and which are necessary for male gametes to acquire their functional competence. This review summarizes the current knowledge about specific ion channels and transporters located in the mammalian sperm plasma membrane, which are intricately involved in the initiation of changes within the ionic milieu of the sperm cell, leading to variations in the sperm membrane potential, membrane depolarization and hyperpolarization, changes in sperm motility and capacitation to further lead to the acrosome reaction and sperm-egg fusion. We also discuss the functionality of selected ion channels in male reproductive health and/or disease since these may become promising targets for clinical management of infertility in the future.
Collapse
Affiliation(s)
- Filip Benko
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (D.U.); (E.T.)
| | - Dana Urminská
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (D.U.); (E.T.)
| | - Michal Ďuračka
- AgroBioTech Research Centre, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
| | - Eva Tvrdá
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia; (D.U.); (E.T.)
| |
Collapse
|
23
|
Boyarko B, Podvin S, Greenberg B, Momper JD, Huang Y, Gerwick WH, Bang AG, Quinti L, Griciuc A, Kim DY, Tanzi RE, Feldman HH, Hook V. Evaluation of bumetanide as a potential therapeutic agent for Alzheimer's disease. Front Pharmacol 2023; 14:1190402. [PMID: 37601062 PMCID: PMC10436590 DOI: 10.3389/fphar.2023.1190402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/28/2023] [Indexed: 08/22/2023] Open
Abstract
Therapeutics discovery and development for Alzheimer's disease (AD) has been an area of intense research to alleviate memory loss and the underlying pathogenic processes. Recent drug discovery approaches have utilized in silico computational strategies for drug candidate selection which has opened the door to repurposing drugs for AD. Computational analysis of gene expression signatures of patients stratified by the APOE4 risk allele of AD led to the discovery of the FDA-approved drug bumetanide as a top candidate agent that reverses APOE4 transcriptomic brain signatures and improves memory deficits in APOE4 animal models of AD. Bumetanide is a loop diuretic which inhibits the kidney Na+-K+-2Cl- cotransporter isoform, NKCC2, for the treatment of hypertension and edema in cardiovascular, liver, and renal disease. Electronic health record data revealed that patients exposed to bumetanide have lower incidences of AD by 35%-70%. In the brain, bumetanide has been proposed to antagonize the NKCC1 isoform which mediates cellular uptake of chloride ions. Blocking neuronal NKCC1 leads to a decrease in intracellular chloride and thus promotes GABAergic receptor mediated hyperpolarization, which may ameliorate disease conditions associated with GABAergic-mediated depolarization. NKCC1 is expressed in neurons and in all brain cells including glia (oligodendrocytes, microglia, and astrocytes) and the vasculature. In consideration of bumetanide as a repurposed drug for AD, this review evaluates its pharmaceutical properties with respect to its estimated brain levels across doses that can improve neurologic disease deficits of animal models to distinguish between NKCC1 and non-NKCC1 mechanisms. The available data indicate that bumetanide efficacy may occur at brain drug levels that are below those required for inhibition of the NKCC1 transporter which implicates non-NKCC1 brain mechansims for improvement of brain dysfunctions and memory deficits. Alternatively, peripheral bumetanide mechanisms may involve cells outside the central nervous system (e.g., in epithelia and the immune system). Clinical bumetanide doses for improved neurological deficits are reviewed. Regardless of mechanism, the efficacy of bumetanide to improve memory deficits in the APOE4 model of AD and its potential to reduce the incidence of AD provide support for clinical investigation of bumetanide as a repurposed AD therapeutic agent.
Collapse
Affiliation(s)
- Ben Boyarko
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Sonia Podvin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Barry Greenberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jeremiah D. Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, United States
- Departments of Neurology and Pathology, University of California, San Francisco, San Francisco, CA, United States
| | - William H. Gerwick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, United States
| | - Anne G. Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys, San Diego, CA, United States
| | - Luisa Quinti
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Ana Griciuc
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Howard H. Feldman
- Department of Neurosciences and Department of Pharmacology, University of California, San Diego, San Diego, United States
- Alzheimer’s Disease Cooperative Study, University of California, San Diego, La Jolla, CA, United States
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences and Department of Pharmacology, University of California, San Diego, San Diego, United States
| |
Collapse
|
24
|
Sadegh C, Xu H, Sutin J, Fatou B, Gupta S, Pragana A, Taylor M, Kalugin PN, Zawadzki ME, Alturkistani O, Shipley FB, Dani N, Fame RM, Wurie Z, Talati P, Schleicher RL, Klein EM, Zhang Y, Holtzman MJ, Moore CI, Lin PY, Patel AB, Warf BC, Kimberly WT, Steen H, Andermann ML, Lehtinen MK. Choroid plexus-targeted NKCC1 overexpression to treat post-hemorrhagic hydrocephalus. Neuron 2023; 111:1591-1608.e4. [PMID: 36893755 PMCID: PMC10198810 DOI: 10.1016/j.neuron.2023.02.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/17/2023] [Accepted: 02/13/2023] [Indexed: 03/11/2023]
Abstract
Post-hemorrhagic hydrocephalus (PHH) refers to a life-threatening accumulation of cerebrospinal fluid (CSF) that occurs following intraventricular hemorrhage (IVH). An incomplete understanding of this variably progressive condition has hampered the development of new therapies beyond serial neurosurgical interventions. Here, we show a key role for the bidirectional Na-K-Cl cotransporter, NKCC1, in the choroid plexus (ChP) to mitigate PHH. Mimicking IVH with intraventricular blood led to increased CSF [K+] and triggered cytosolic calcium activity in ChP epithelial cells, which was followed by NKCC1 activation. ChP-targeted adeno-associated viral (AAV)-NKCC1 prevented blood-induced ventriculomegaly and led to persistently increased CSF clearance capacity. These data demonstrate that intraventricular blood triggered a trans-choroidal, NKCC1-dependent CSF clearance mechanism. Inactive, phosphodeficient AAV-NKCC1-NT51 failed to mitigate ventriculomegaly. Excessive CSF [K+] fluctuations correlated with permanent shunting outcome in humans following hemorrhagic stroke, suggesting targeted gene therapy as a potential treatment to mitigate intracranial fluid accumulation following hemorrhage.
Collapse
Affiliation(s)
- Cameron Sadegh
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Huixin Xu
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jason Sutin
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Benoit Fatou
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Suhasini Gupta
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Aja Pragana
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Milo Taylor
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard College, Harvard University, Cambridge, MA 02138, USA
| | - Peter N Kalugin
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Miriam E Zawadzki
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Osama Alturkistani
- Cellular Imaging Core, Boston Children's Hospital, Boston, MA 02115, USA
| | - Frederick B Shipley
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Neil Dani
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Zainab Wurie
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Pratik Talati
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Riana L Schleicher
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Eric M Klein
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Christopher I Moore
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Pei-Yi Lin
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Benjamin C Warf
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA 02115, USA
| | - W Taylor Kimberly
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hanno Steen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mark L Andermann
- Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
25
|
Meldrum Robertson R, MacMillan HA, Andersen MK. A cold and quiet brain: mechanisms of insect CNS arrest at low temperatures. CURRENT OPINION IN INSECT SCIENCE 2023:101055. [PMID: 37201631 DOI: 10.1016/j.cois.2023.101055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 05/20/2023]
Abstract
Exposure to cold causes insects to enter a chill coma at species-specific temperatures and such temperature sensitivity contributes to geographic distribution and phenology. Coma results from abrupt spreading depolarization (SD) of neural tissue in the integrative centers of the CNS. SD abolishes neuronal signaling and the operation of neural circuits, like an off switch for the CNS. Turning off the CNS by allowing ion gradients to collapse will conserve energy and may offset negative consequences of temporary immobility. SD is modified by prior experience via rapid cold hardening (RCH) or cold acclimation which alter properties of Kv channels, Na+/K+-ATPase and Na+/K+/2Cl- cotransporter. The stress hormone octopamine mediates RCH. Future progress depends on developing a more complete understanding of ion homeostasis in and of the insect CNS.
Collapse
Affiliation(s)
| | - Heath A MacMillan
- Department of Biology, Carleton University, Ottawa, ON, Canada, K1S 5B6.
| | - Mads K Andersen
- Department of Biology, Carleton University, Ottawa, ON, Canada, K1S 5B6.
| |
Collapse
|
26
|
Jennings ML. Role of transporters in regulating mammalian intracellular inorganic phosphate. Front Pharmacol 2023; 14:1163442. [PMID: 37063296 PMCID: PMC10097972 DOI: 10.3389/fphar.2023.1163442] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
This review summarizes the current understanding of the role of plasma membrane transporters in regulating intracellular inorganic phosphate ([Pi]In) in mammals. Pi influx is mediated by SLC34 and SLC20 Na+-Pi cotransporters. In non-epithelial cells other than erythrocytes, Pi influx via SLC20 transporters PiT1 and/or PiT2 is balanced by efflux through XPR1 (xenotropic and polytropic retrovirus receptor 1). Two new pathways for mammalian Pi transport regulation have been described recently: 1) in the presence of adequate Pi, cells continuously internalize and degrade PiT1. Pi starvation causes recycling of PiT1 from early endosomes to the plasma membrane and thereby increases the capacity for Pi influx; and 2) binding of inositol pyrophosphate InsP8 to the SPX domain of XPR1 increases Pi efflux. InsP8 is degraded by a phosphatase that is strongly inhibited by Pi. Therefore, an increase in [Pi]In decreases InsP8 degradation, increases InsP8 binding to SPX, and increases Pi efflux, completing a feedback loop for [Pi]In homeostasis. Published data on [Pi]In by magnetic resonance spectroscopy indicate that the steady state [Pi]In of skeletal muscle, heart, and brain is normally in the range of 1–5 mM, but it is not yet known whether PiT1 recycling or XPR1 activation by InsP8 contributes to Pi homeostasis in these organs. Data on [Pi]In in cultured cells are variable and suggest that some cells can regulate [Pi] better than others, following a change in [Pi]Ex. More measurements of [Pi]In, influx, and efflux are needed to determine how closely, and how rapidly, mammalian [Pi]In is regulated during either hyper- or hypophosphatemia.
Collapse
|
27
|
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: 19] [Impact Index Per Article: 9.5] [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
|
28
|
Saito M, Tanaka C, Toyoda H, Kang Y. Subcellular Localization of Homomeric TASK3 Channels and Its Presumed Functional Significances in Trigeminal Motoneurons. Int J Mol Sci 2022; 24:ijms24010344. [PMID: 36613787 PMCID: PMC9820627 DOI: 10.3390/ijms24010344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Somatic expressions of either heteromeric TASK1/3 or homomeric TASK1/1 channels have been reported in various neurons, while expression of homomeric TASK3/3 channels has been re-ported only in dendrites. However, it is not known why homomeric TASK3/3 channels are hardly seen in somata of CNS neurons. Given the absence of somatic TASK3/3 channels, it should be clarified why dendritic expression of TASK3/3 channels is inevitable and necessary and how differentially distributed TASK1/1 and TASK3/3 channels play roles in soma-to-dendritic integration. Here, we addressed these questions. We found that TASK3-transfected HEK293 cells showed decreases in cell volume after being transferred from the cultured medium to HEPES Ringer, suggesting that expressions of TASK3 channels in cell bodies cause an osmolarity problem. Using TASK1- and TASK3-transfected oocytes, we also found that cGMP application slightly suppressed TASK3 currents while it largely enhanced TASK1 currents, alleviating the difference between TASK1 and TASK3 currents at physiological pH. As larger motoneurons have extensive dendritic trees while smaller motoneurons have poor ones, cGMP could integrate Ia-EPSPs to recruit small and large motoneurons synchronously by differentially modulating TASKI and TASK3 channels which were complementary distributed in soma and dendrites of motoneurons in the dorsolateral part of the trigeminal motor nucleus.
Collapse
Affiliation(s)
- Mitsuru Saito
- Department of Oral Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Chie Tanaka
- Department of Neuroscience and Oral Physiology, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
| | - Hiroki Toyoda
- Department of Neuroscience and Oral Physiology, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
- Correspondence: (H.T.); (Y.K.)
| | - Youngnam Kang
- Department of Neuroscience and Oral Physiology, Osaka University Graduate School of Dentistry, Suita 565-0871, Japan
- Department of Behavioral Sciences, Osaka University Graduate School of Human Sciences, Suita 565-0871, Japan
- Correspondence: (H.T.); (Y.K.)
| |
Collapse
|
29
|
Talifu Z, Pan Y, Gong H, Xu X, Zhang C, Yang D, Gao F, Yu Y, Du L, Li J. The role of KCC2 and NKCC1 in spinal cord injury: From physiology to pathology. Front Physiol 2022; 13:1045520. [PMID: 36589461 PMCID: PMC9799334 DOI: 10.3389/fphys.2022.1045520] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The balance of ion concentrations inside and outside the cell is an essential homeostatic mechanism in neurons and serves as the basis for a variety of physiological activities. In the central nervous system, NKCC1 and KCC2, members of the SLC12 cation-chloride co-transporter (CCC) family, participate in physiological and pathophysiological processes by regulating intracellular and extracellular chloride ion concentrations, which can further regulate the GABAergic system. Over recent years, studies have shown that NKCC1 and KCC2 are essential for the maintenance of Cl- homeostasis in neural cells. NKCC1 transports Cl- into cells while KCC2 transports Cl- out of cells, thereby regulating chloride balance and neuronal excitability. An imbalance of NKCC1 and KCC2 after spinal cord injury will disrupt CI- homeostasis, resulting in the transformation of GABA neurons from an inhibitory state into an excitatory state, which subsequently alters the spinal cord neural network and leads to conditions such as spasticity and neuropathic pain, among others. Meanwhile, studies have shown that KCC2 is also an essential target for motor function reconstruction after spinal cord injury. This review mainly introduces the physiological structure and function of NKCC1 and KCC2 and discusses their pathophysiological roles after spinal cord injury.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yunzhu Pan
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Han Gong
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Chunjia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Degang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liangjie Du
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| | - Jianjun Li
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| |
Collapse
|
30
|
Janoš P, Magistrato A. Role of Monovalent Ions in the NKCC1 Inhibition Mechanism Revealed through Molecular Simulations. Int J Mol Sci 2022; 23:ijms232315439. [PMID: 36499764 PMCID: PMC9741434 DOI: 10.3390/ijms232315439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The secondary active Na-K-Cl cotransporter 1 (NKCC1) promotes electroneutral uptake of two chloride ions, one sodium ion and one potassium ion. NKCC1 regulates Cl- homeostasis, thus being implicated in transepithelial water transport and in neuronal excitability. Aberrant NKCC1 transport is linked to a variety of human diseases. The loop diuretic drugs bumetanide, furosemide, azosemide and ethacrynic acid target NKCC1, but are characterized by poor selectivity leading to severe side effects. Despite its therapeutic importance, the molecular details of the NKCC1 inhibition mechanism remain unclear. Using all-atom simulations, we predict a putative binding mode of these drugs to the zebrafish (z) and human (h) NKCC1 orthologs. Although differing in their specific interactions with NKCC1 and/or monovalent ions, all drugs can fit within the same cavity and engage in hydrophobic interactions with M304/M382 in z/hNKCC1, a proposed ion gating residue demonstrated to be key for bumetanide binding. Consistent with experimental evidence, all drugs take advantage of the K+/Na+ ions, which plastically respond to their binding. This study not only provides atomic-level insights useful for drug discovery campaigns of more selective/potent NKCC1 inhibitors aimed to tackle diseases related to deregulated Cl- homeostasis, but it also supplies a paradigmatic example of the key importance of dynamical effects when drug binding is mediated by monovalent ions.
Collapse
|
31
|
Zhao Y, Cao E. Structural Pharmacology of Cation-Chloride Cotransporters. MEMBRANES 2022; 12:1206. [PMID: 36557113 PMCID: PMC9784483 DOI: 10.3390/membranes12121206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Loop and thiazide diuretics have been cornerstones of clinical management of hypertension and fluid overload conditions for more than five decades. The hunt for their molecular targets led to the discovery of cation-chloride cotransporters (CCCs) that catalyze electroneutral movement of Cl- together with Na+ and/or K+. CCCs consist of two 1 Na+-1 K+-2 Cl- (NKCC1-2), one 1 Na+-1 Cl- (NCC), and four 1 K+-1 Cl- (KCC1-4) transporters in human. CCCs are fundamental in trans-epithelia ion secretion and absorption, homeostasis of intracellular Cl- concentration and cell volume, and regulation of neuronal excitability. Malfunction of NKCC2 and NCC leads to abnormal salt and water retention in the kidney and, consequently, imbalance in electrolytes and blood pressure. Mutations in KCC2 and KCC3 are associated with brain disorders due to impairments in regulation of excitability and possibly cell volume of neurons. A recent surge of structures of CCCs have defined their dimeric architecture, their ion binding sites, their conformational changes associated with ion translocation, and the mechanisms of action of loop diuretics and small molecule inhibitors. These breakthroughs now set the stage to expand CCC pharmacology beyond loop and thiazide diuretics, developing the next generation of diuretics with improved potency and specificity. Beyond drugging renal-specific CCCs, brain-penetrable therapeutics are sorely needed to target CCCs in the nervous system for the treatment of neurological disorders and psychiatric conditions.
Collapse
|
32
|
Hu M, Zhou N, Cai W, Xu H. Lysosomal solute and water transport. J Cell Biol 2022; 221:213536. [PMID: 36219209 PMCID: PMC9559593 DOI: 10.1083/jcb.202109133] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
Lysosomes mediate hydrolase-catalyzed macromolecule degradation to produce building block catabolites for reuse. Lysosome function requires an osmo-sensing machinery that regulates osmolytes (ions and organic solutes) and water flux. During hypoosmotic stress or when undigested materials accumulate, lysosomes become swollen and hypo-functional. As a membranous organelle filled with cargo macromolecules, catabolites, ions, and hydrolases, the lysosome must have mechanisms that regulate its shape and size while coordinating content exchange. In this review, we discussed the mechanisms that regulate lysosomal fusion and fission as well as swelling and condensation, with a focus on solute and water transport mechanisms across lysosomal membranes. Lysosomal H+, Na+, K+, Ca2+, and Cl- channels and transporters sense trafficking and osmotic cues to regulate both solute flux and membrane trafficking. We also provide perspectives on how lysosomes may adjust the volume of themselves, the cytosol, and the cytoplasm through the control of lysosomal solute and water transport.
Collapse
Affiliation(s)
- Meiqin Hu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI.,Liangzhu Laboratory & Zhejiang University Medical Center, Hangzhou, China
| | - Nan Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI.,Liangzhu Laboratory & Zhejiang University Medical Center, Hangzhou, China
| | - Weijie Cai
- Liangzhu Laboratory & Zhejiang University Medical Center, Hangzhou, China
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI.,Liangzhu Laboratory & Zhejiang University Medical Center, Hangzhou, China.,Department of Neurology, Second Affiliated Hospital of Zhejiang University Medical School, Hangzhou, China
| |
Collapse
|
33
|
Walch E, Fiacco TA. Honey, I shrunk the extracellular space: Measurements and mechanisms of astrocyte swelling. Glia 2022; 70:2013-2031. [PMID: 35635369 PMCID: PMC9474570 DOI: 10.1002/glia.24224] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 12/16/2022]
Abstract
Astrocyte volume fluctuation is a physiological phenomenon tied closely to the activation of neural circuits. Identification of underlying mechanisms has been challenging due in part to use of a wide range of experimental approaches that vary between research groups. Here, we first review the many methods that have been used to measure astrocyte volume changes directly or indirectly. While the field has recently shifted towards volume analysis using fluorescence microscopy to record cell volume changes directly, established metrics corresponding to extracellular space dynamics have also yielded valuable insights. We then turn to analysis of mechanisms of astrocyte swelling derived from many studies, with a focus on volume changes tied to increases in extracellular potassium concentration ([K+ ]o ). The diverse methods that have been utilized to generate the external [K+ ]o environment highlight multiple scenarios of astrocyte swelling mediated by different mechanisms. Classical potassium buffering theories are tempered by many recent studies that point to different swelling pathways optimized at particular [K+ ]o and that depend on local/transient versus more sustained increases in [K+ ]o .
Collapse
Affiliation(s)
- Erin Walch
- Division of Biomedical Sciences, School of MedicineUniversity of California, RiversideRiversideCaliforniaUSA
| | - Todd A. Fiacco
- Department of Molecular, Cell and Systems BiologyUniversity of California, RiversideRiversideCaliforniaUSA
- Center for Glial‐Neuronal InteractionsUniversity of California, RiversideRiversideCaliforniaUSA
| |
Collapse
|
34
|
Neumann C, Rosenbæk LL, Flygaard RK, Habeck M, Karlsen JL, Wang Y, Lindorff‐Larsen K, Gad HH, Hartmann R, Lyons JA, Fenton RA, Nissen P. Cryo-EM structure of the human NKCC1 transporter reveals mechanisms of ion coupling and specificity. EMBO J 2022; 41:e110169. [PMID: 36239040 PMCID: PMC9713717 DOI: 10.15252/embj.2021110169] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 12/03/2022] Open
Abstract
The sodium-potassium-chloride transporter NKCC1 of the SLC12 family performs Na+ -dependent Cl- - and K+ -ion uptake across plasma membranes. NKCC1 is important for regulating cell volume, hearing, blood pressure, and regulation of hyperpolarizing GABAergic and glycinergic signaling in the central nervous system. Here, we present a 2.6 Å resolution cryo-electron microscopy structure of human NKCC1 in the substrate-loaded (Na+ , K+ , and 2 Cl- ) and occluded, inward-facing state that has also been observed for the SLC6-type transporters MhsT and LeuT. Cl- binding at the Cl1 site together with the nearby K+ ion provides a crucial bridge between the LeuT-fold scaffold and bundle domains. Cl- -ion binding at the Cl2 site seems to undertake a structural role similar to conserved glutamate of SLC6 transporters and may allow for Cl- -sensitive regulation of transport. Supported by functional studies in mammalian cells and computational simulations, we describe a putative Na+ release pathway along transmembrane helix 5 coupled to the Cl2 site. The results provide insight into the structure-function relationship of NKCC1 with broader implications for other SLC12 family members.
Collapse
Affiliation(s)
- Caroline Neumann
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | | | - Rasmus Kock Flygaard
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Michael Habeck
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | | | - Yong Wang
- Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark,Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhouChina
| | - Kresten Lindorff‐Larsen
- Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Hans Henrik Gad
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Rune Hartmann
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Joseph Anthony Lyons
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark,Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhusDenmark
| | | | - Poul Nissen
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| |
Collapse
|
35
|
Gamba G. Arterial Blood Pressure, Neuronal Excitability, Mineral Metabolism and Cell Volume Regulation Mechanisms Revealed by Xenopus laevis oocytes. MEMBRANES 2022; 12:911. [PMID: 36295670 PMCID: PMC9612257 DOI: 10.3390/membranes12100911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/16/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
Xenopus laevis oocytes have been an invaluable tool to discover and explore the molecular mechanisms and characteristics of many proteins, in particular integral membrane proteins. The oocytes were fundamental in many projects designed to identify the cDNA encoding a diversity of membrane proteins including receptors, transporters, channels and pores. In addition to being a powerful tool for cloning, oocytes were later used to experiment with the functional characterization of many of the identified proteins. In this review I present an overview of my personal 30-year experience using Xenopus laevis oocytes and the impact this had on a variety of fields such as arterial blood pressure, neuronal excitability, mineral metabolism and cell volume regulation.
Collapse
Affiliation(s)
- Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 04510, Mexico
| |
Collapse
|
36
|
Del Alamo D, Meiler J, Mchaourab HS. Principles of Alternating Access in LeuT-fold Transporters: Commonalities and Divergences. J Mol Biol 2022; 434:167746. [PMID: 35843285 DOI: 10.1016/j.jmb.2022.167746] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/15/2022]
Abstract
Found in all domains of life, transporters belonging to the LeuT-fold class mediate the import and exchange of hydrophilic and charged compounds such as amino acids, metals, and sugar molecules. Nearly two decades of investigations on the eponymous bacterial transporter LeuT have yielded a library of high-resolution snapshots of its conformational cycle linked by solution-state experimental data obtained from multiple techniques. In parallel, its topology has been observed in symporters and antiporters characterized by a spectrum of substrate specificities and coupled to gradients of distinct ions. Here we review and compare mechanistic models of transport for LeuT, its well-studied homologs, as well as functionally distant members of the fold, emphasizing the commonalities and divergences in alternating access and the corresponding energy landscapes. Our integrated summary illustrates how fold conservation, a hallmark of the LeuT fold, coincides with divergent choreographies of alternating access that nevertheless capitalize on recurrent structural motifs. In addition, it highlights the knowledge gap that hinders the leveraging of the current body of research into detailed mechanisms of transport for this important class of membrane proteins.
Collapse
Affiliation(s)
- Diego Del Alamo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, USA. https://twitter.com/DdelAlamo
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA; Institute for Drug Discovery, Leipzig University, Leipzig, DE, USA. https://twitter.com/MeilerLab
| | - Hassane S Mchaourab
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
37
|
Zhao Y, Shen J, Wang Q, Ruiz Munevar MJ, Vidossich P, De Vivo M, Zhou M, Cao E. Structure of the human cation-chloride cotransport KCC1 in an outward-open state. Proc Natl Acad Sci U S A 2022; 119:e2109083119. [PMID: 35759661 PMCID: PMC9271165 DOI: 10.1073/pnas.2109083119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 04/25/2022] [Indexed: 12/30/2022] Open
Abstract
Cation-chloride cotransporters (CCCs) catalyze electroneutral symport of Cl- with Na+ and/or K+ across membranes. CCCs are fundamental in cell volume homeostasis, transepithelia ion movement, maintenance of intracellular Cl- concentration, and neuronal excitability. Here, we present a cryoelectron microscopy structure of human K+-Cl- cotransporter (KCC)1 bound with the VU0463271 inhibitor in an outward-open state. In contrast to many other amino acid-polyamine-organocation transporter cousins, our first outward-open CCC structure reveals that opening the KCC1 extracellular ion permeation path does not involve hinge-bending motions of the transmembrane (TM) 1 and TM6 half-helices. Instead, rocking of TM3 and TM8, together with displacements of TM4, TM9, and a conserved intracellular loop 1 helix, underlie alternate opening and closing of extracellular and cytoplasmic vestibules. We show that KCC1 intriguingly exists in one of two distinct dimeric states via different intersubunit interfaces. Our studies provide a blueprint for understanding the mechanisms of CCCs and their inhibition by small molecule compounds.
Collapse
Affiliation(s)
- Yongxiang Zhao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Jiemin Shen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Qinzhe Wang
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112
| | | | | | - Marco De Vivo
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Ming Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112
| |
Collapse
|
38
|
Mecawi AS, Varanda WA, da Silva MP. Osmoregulation and the Hypothalamic Supraoptic Nucleus: From Genes to Functions. Front Physiol 2022; 13:887779. [PMID: 35685279 PMCID: PMC9171026 DOI: 10.3389/fphys.2022.887779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Due to the relatively high permeability to water of the plasma membrane, water tends to equilibrate its chemical potential gradient between the intra and extracellular compartments. Because of this, changes in osmolality of the extracellular fluid are accompanied by changes in the cell volume. Therefore, osmoregulatory mechanisms have evolved to keep the tonicity of the extracellular compartment within strict limits. This review focuses on the following aspects of osmoregulation: 1) the general problems in adjusting the "milieu interieur" to challenges imposed by water imbalance, with emphasis on conceptual aspects of osmosis and cell volume regulation; 2) osmosensation and the hypothalamic supraoptic nucleus (SON), starting with analysis of the electrophysiological responses of the magnocellular neurosecretory cells (MNCs) involved in the osmoreception phenomenon; 3) transcriptomic plasticity of SON during sustained hyperosmolality, to pinpoint the genes coding membrane channels and transporters already shown to participate in the osmosensation and new candidates that may have their role further investigated in this process, with emphasis on those expressed in the MNCs, discussing the relationships of hydration state, gene expression, and MNCs electrical activity; and 4) somatodendritic release of neuropeptides in relation to osmoregulation. Finally, we expect that by stressing the relationship between gene expression and the electrical activity of MNCs, studies about the newly discovered plastic-regulated genes that code channels and transporters in the SON may emerge.
Collapse
Affiliation(s)
- André Souza Mecawi
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Wamberto Antonio Varanda
- Department of Physiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Melina Pires da Silva
- Laboratory of Cellular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
39
|
Li Y, Lin J, Wang P, Zhu F, Wu M, Luo Q, Zhang Y, Liu X. Tumor Microenvironment-Responsive Yolk-Shell NaCl@Virus-Inspired Tetrasulfide-Organosilica for Ion-Interference Therapy via Osmolarity Surge and Oxidative Stress Amplification. ACS NANO 2022; 16:7380-7397. [PMID: 35435672 DOI: 10.1021/acsnano.1c09496] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Ion-interference therapy, which utilizes ions to disturb intracellular biological processes, provides inspiration for tumor therapy. Artificially reversing osmotic pressure by transporting large amounts of physiological ions to tumor cells is a straightforward yet low-toxic strategy for ion-interference therapy. However, it is hard to achieve due to the serious limitations of single-ion delivery. Herein, we skillfully deliver NaCl nanocrystals to tumor sites and sequentially realize the explosive release of Na+/Cl- inside tumor cells by utilizing a virus-mimicking and glutathione (GSH)-responsive hollow mesoporous tetrasulfide-bridged organosilica (ssss-VHMS). Once the ssss-VHMS-wrapped NaCl nanocrystals (NaCl@ssss-VHMS) accumulate in the tumors, they would rapidly invade tumor cells via spike surface-assisted endocytosis, thus bypassing Na+/K+-ATPase transmembrane ion transporters. Afterward, the intracellular overproduced GSH of tumor cells would trigger the rapid degradation of ssss-VHMS via thiol-tetrasulfide exchange, which could not only remarkably deplete the GSH but also explosively release the Na+/Cl-, leading to the osmolarity surge accompanied by reactive oxygen species (ROS) generation. The cell swelling, ROS storm, and GSH exhaustion of NaCl@ssss-VHMS effectively eradicated tumor cells by caspase-1-dependent pyroptosis, caspase-3-dependent apoptosis, and GPX4-dependent ferroptosis, respectively, thus synergistically inhibiting tumor growth. We believe that NaCl@ssss-VHMS would be a potential cancer therapeutic agent, and this discovery could provide a perspective for exploring synergistic ion-interference therapy.
Collapse
Affiliation(s)
- Yang Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
- University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou 341000, People's Republic of China
| | - Jinyan Lin
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
| | - Peiyuan Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Fukai Zhu
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Qiang Luo
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Yun Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
- University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou 341000, People's Republic of China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
- University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou 341000, People's Republic of China
| |
Collapse
|
40
|
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
|
41
|
Duong PC, McCabe TC, Riley GF, Holmes HL, Piermarini PM, Romero MF, Gillen CM. Sequence analysis and function of mosquito aeCCC2 and Drosophila Ncc83 orthologs. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 143:103729. [PMID: 35150868 PMCID: PMC9012228 DOI: 10.1016/j.ibmb.2022.103729] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 05/08/2023]
Abstract
Dipteran insects have genes that code for two different Na+-dependent cation-chloride cotransporter (CCC) paralogs. Aedes aegypti aeNKCC1 is an ortholog of Drosophila melanogaster Ncc69, a bumetanide-sensitive Na+-K+-2Cl- cotransporter (NKCC). Aedes aegypti aeCCC2 and aeCCC3 are orthologs of Drosophila Ncc83. Prior work suggests that the transport properties of aeCCC2 differ from canonical NKCCs. In particular, Xenopus oocytes expressing aeCCC2 have increased Na+-dependent membrane currents compared to controls, whereas NKCCs are electroneutral. Here, we further evaluated the function and localization of aeCCC2 and Ncc83. In oocytes expressing aeCCC2 or Ncc83, membrane potential (Vm) hyperpolarized upon Na+ removal; following hypotonic exposure the change in Vm was greater than it was in controls. In voltage-clamp experiments, membrane currents were concentration dependent on Na+ with an apparent affinity (Km) of approximately 4.6 mM. In Malpighian tubules of larval and adult mosquitoes, aeCCC2 was localized along the basolateral aspect of principal cells. Sequence comparisons among transporters from Drosophila, Aedes, Anopheles, and Culex revealed 33 residues within the transmembrane domains (TMDs) that are fully conserved within paralogs but that differ between orthologs of NKCC1 and orthologs of aeCCC2/Ncc83. These residues are distributed across all 12 TMDs. Our results provide a foundation for further exploration of the structural basis for functional differences between insect Na+-dependent CCCs.
Collapse
Affiliation(s)
- Phu C Duong
- Department of Biology, Kenyon College, Gambier, OH, 43050, USA
| | - Tobias C McCabe
- Department of Biology, Kenyon College, Gambier, OH, 43050, USA
| | - Grace F Riley
- Department of Biology, Kenyon College, Gambier, OH, 43050, USA
| | - Heather L Holmes
- Physiology and Biomedical Engineering, Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, 55902, USA
| | - Peter M Piermarini
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, OH, 44691, USA
| | - Michael F Romero
- Physiology and Biomedical Engineering, Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, 55902, USA
| | | |
Collapse
|
42
|
Wang J, Liu W, Xu W, Yang B, Cui M, Li Z, Zhang H, Jin C, Xue H, Zhang J. Comprehensive Analysis of the Oncogenic, Genomic Alteration, and Immunological Landscape of Cation-Chloride Cotransporters in Pan-Cancer. Front Oncol 2022; 12:819688. [PMID: 35372048 PMCID: PMC8968682 DOI: 10.3389/fonc.2022.819688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background Assessing the phenotypic diversity underlying tumor progression requires the identification of variations in the respective molecular interaction in the tumor microenvironment (TME). Despite emerging studies focusing on the association between cation-chloride cotransporters (CCCs) and carcinogenesis, direct evidence that CCCs (KCC2 and NKCC1) mediate tumor progression in pan-cancer remains unclear. Methods We conducted a comprehensive assessment of the expression, DNA variation profiles, and prognostic and immunologic implications of CCCs based on a large-scale pan-cancer population, including 10,967 cancer patients from the Cancer Genome Atlas, 9,162 cancer patients from Genomics Expression Omnibus, 48,834 cancer patients from 188 independent studies, and 356 cancer patients from three real-world cohorts. Results In this study, we first found that CCCs were highly expressed in most tumors, and prominently associated with prognosis. Kaplan-Meier analysis and Cox regression analysis revealed that KCC2 and NKCC1 significantly predicted survival for patients with pan-cancer, suggesting that CCCs have inconsistent tumorigenesis regulatory mechanisms in cancers. Next, we examined the DNA variation landscape of KCC2 and NKCC1 and their prognostic implications in pan-cancer. The results demonstrated that UCEC patients with somatic copy number variation (CNV) of NKCC1 received significantly better outcomes (p < 0.05). Besides emphasizing the clinical implications of CNV of CCCs for cancer patients, we found that NKCC1MUT could prominently prolong progression-free survival (p = 2.59e-04), disease-specific survival (p = 0.019), and overall survival (p = 0.034) compared with NKCC1WT cancer patients possibly via regulation of cell proliferation and oncogenic stress pathways. Additionally, KCC2 positively correlated with the levels of tumor-infiltrating macrophages and CD4+ T cells, but NKCC1 showed a significantly widely negative association with tumor-infiltrated lymphocytes, suggesting an immune-excluded TME in cancers. Similarly, expression of KCC2, rather than NKCC1, was positively correlated with the immune checkpoint molecules, indicating its role as an immune regulator in a wide variety of cancers. Finally, to verify our hypothesis and altered expression of CCCs, we performed IHC analysis and revealed the staining distribution in tumor and adjacent normal tissues of glioma, clear cell renal cell carcinoma, papillary cell renal cell carcinoma, and hepatocellular and breast cancer from three real-world cohorts, and validated prominently prognostic implications of CCCs in patients with clear cell renal cell carcinoma. Conclusion This study first comprehensively investigated the molecular and clinical role of CCCs, and illustrated the significant association among KCC2/NKCC1 expression, DNA variation profiles prognosis, and TME of pan-cancer. The pan-cancer findings provided an in-depth understanding of potential oncogenic and immunologic of differential expression and DNA alteration of KCC2/NKCC1 cancers.
Collapse
Affiliation(s)
- Jie Wang
- Department of Anesthesiology and Perioperative Medicine, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Wangrui Liu
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Baofeng Yang
- Department of Anesthesiology and Perioperative Medicine, Affiliate Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingzhu Cui
- Department of Anesthesiology and Perioperative Medicine, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Zhen Li
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chuntao Jin
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Huanzhou Xue
- Department of Hepatobiliary Surgery, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
43
|
Serranilla M, Woodin MA. Striatal Chloride Dysregulation and Impaired GABAergic Signaling Due to Cation-Chloride Cotransporter Dysfunction in Huntington’s Disease. Front Cell Neurosci 2022; 15:817013. [PMID: 35095429 PMCID: PMC8795088 DOI: 10.3389/fncel.2021.817013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Intracellular chloride (Cl–) levels in mature neurons must be tightly regulated for the maintenance of fast synaptic inhibition. In the mature central nervous system (CNS), synaptic inhibition is primarily mediated by gamma-amino butyric acid (GABA), which binds to Cl– permeable GABAA receptors (GABAARs). The intracellular Cl– concentration is primarily maintained by the antagonistic actions of two cation-chloride cotransporters (CCCs): Cl–-importing Na+-K+-Cl– co-transporter-1 (NKCC1) and Cl– -exporting K+-Cl– co-transporter-2 (KCC2). In mature neurons in the healthy brain, KCC2 expression is higher than NKCC1, leading to lower levels of intracellular Cl–, and Cl– influx upon GABAAR activation. However, in neurons of the immature brain or in neurological disorders such as epilepsy and traumatic brain injury, impaired KCC2 function and/or enhanced NKCC1 expression lead to intracellular Cl– accumulation and GABA-mediated excitation. In Huntington’s disease (HD), KCC2- and NKCC1-mediated Cl–-regulation are also altered, which leads to GABA-mediated excitation and contributes to the development of cognitive and motor impairments. This review summarizes the role of Cl– (dys)regulation in the healthy and HD brain, with a focus on the basal ganglia (BG) circuitry and CCCs as potential therapeutic targets in the treatment of HD.
Collapse
|
44
|
Branchereau P, Cattaert D. Chloride Homeostasis in Developing Motoneurons. ADVANCES IN NEUROBIOLOGY 2022; 28:45-61. [PMID: 36066820 DOI: 10.1007/978-3-031-07167-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Maturation of GABA/Glycine chloride-mediated synaptic inhibitions is crucial for the establishment of a balance between excitation and inhibition. GABA and glycine are excitatory neurotransmitters on immature neurons that exhibit elevated [Cl-]i. Later in development [Cl-]i drops leading to the occurrence of inhibitory synaptic activity. This ontogenic change is closely correlated to a differential expression of two cation-chloride cotransporters that are the Cl- channel K+/Cl- co-transporter type 2 (KCC2) that extrudes Cl- ions and the Na+-K+-2Cl- cotransporter NKCC1 that accumulates Cl- ions. The classical scheme built from studies performed on cortical and hippocampal networks proposes that immature neurons display high [Cl-]i because NKCC1 is overexpressed compared to KCC2 and that the co-transporters ratio reverses in mature neurons, lowering [Cl-]i. In this chapter, we will see that this classical scheme is not true in motoneurons (MNs) and that an early alteration of the chloride homeostasis may be involved in pathological conditions.
Collapse
Affiliation(s)
- Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France.
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France
| |
Collapse
|
45
|
Tóth K, Lénárt N, Berki P, Fekete R, Szabadits E, Pósfai B, Cserép C, Alatshan A, Benkő S, Kiss D, Hübner CA, Gulyás A, Kaila K, Környei Z, Dénes Á. The NKCC1 ion transporter modulates microglial phenotype and inflammatory response to brain injury in a cell-autonomous manner. PLoS Biol 2022; 20:e3001526. [PMID: 35085235 PMCID: PMC8856735 DOI: 10.1371/journal.pbio.3001526] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/18/2022] [Accepted: 01/04/2022] [Indexed: 12/25/2022] Open
Abstract
The NKCC1 ion transporter contributes to the pathophysiology of common neurological disorders, but its function in microglia, the main inflammatory cells of the brain, has remained unclear to date. Therefore, we generated a novel transgenic mouse line in which microglial NKCC1 was deleted. We show that microglial NKCC1 shapes both baseline and reactive microglia morphology, process recruitment to the site of injury, and adaptation to changes in cellular volume in a cell-autonomous manner via regulating membrane conductance. In addition, microglial NKCC1 deficiency results in NLRP3 inflammasome priming and increased production of interleukin-1β (IL-1β), rendering microglia prone to exaggerated inflammatory responses. In line with this, central (intracortical) administration of the NKCC1 blocker, bumetanide, potentiated intracortical lipopolysaccharide (LPS)-induced cytokine levels. In contrast, systemic bumetanide application decreased inflammation in the brain. Microglial NKCC1 KO animals exposed to experimental stroke showed significantly increased brain injury, inflammation, cerebral edema and worse neurological outcome. Thus, NKCC1 emerges as an important player in controlling microglial ion homeostasis and inflammatory responses through which microglia modulate brain injury. The contribution of microglia to central NKCC1 actions is likely to be relevant for common neurological disorders.
Collapse
Affiliation(s)
- Krisztina Tóth
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Péter Berki
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
- Laboratory of Cerebral Cortex Research, Institute of Experimental Medicine, Budapest, Hungary
| | - Rebeka Fekete
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Szabadits
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ahmad Alatshan
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Benkő
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cellular and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dániel Kiss
- Software Engineering Institute, John von Neumann Faculty of Informatics, Óbuda University, Budapest, Hungary
| | | | - Attila Gulyás
- Laboratory of Cerebral Cortex Research, Institute of Experimental Medicine, Budapest, Hungary
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- * E-mail:
| |
Collapse
|
46
|
Whittamore JM, Hatch M. Oxalate Flux Across the Intestine: Contributions from Membrane Transporters. Compr Physiol 2021; 12:2835-2875. [PMID: 34964122 DOI: 10.1002/cphy.c210013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Epithelial oxalate transport is fundamental to the role occupied by the gastrointestinal (GI) tract in oxalate homeostasis. The absorption of dietary oxalate, together with its secretion into the intestine, and degradation by the gut microbiota, can all influence the excretion of this nonfunctional terminal metabolite in the urine. Knowledge of the transport mechanisms is relevant to understanding the pathophysiology of hyperoxaluria, a risk factor in kidney stone formation, for which the intestine also offers a potential means of treatment. The following discussion presents an expansive review of intestinal oxalate transport. We begin with an overview of the fate of oxalate, focusing on the sources, rates, and locations of absorption and secretion along the GI tract. We then consider the mechanisms and pathways of transport across the epithelial barrier, discussing the transcellular, and paracellular components. There is an emphasis on the membrane-bound anion transporters, in particular, those belonging to the large multifunctional Slc26 gene family, many of which are expressed throughout the GI tract, and we summarize what is currently known about their participation in oxalate transport. In the final section, we examine the physiological stimuli proposed to be involved in regulating some of these pathways, encompassing intestinal adaptations in response to chronic kidney disease, metabolic acid-base disorders, obesity, and following gastric bypass surgery. There is also an update on research into the probiotic, Oxalobacter formigenes, and the basis of its unique interaction with the gut epithelium. © 2021 American Physiological Society. Compr Physiol 11:1-41, 2021.
Collapse
Affiliation(s)
- Jonathan M Whittamore
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Marguerite Hatch
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
47
|
The Important Role of Ion Transport System in Cervical Cancer. Int J Mol Sci 2021; 23:ijms23010333. [PMID: 35008759 PMCID: PMC8745646 DOI: 10.3390/ijms23010333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Cervical cancer is a significant gynecological cancer and causes cancer-related deaths worldwide. Human papillomavirus (HPV) is implicated in the etiology of cervical malignancy. However, much evidence indicates that HPV infection is a necessary but not sufficient cause in cervical carcinogenesis. Therefore, the cellular pathophysiology of cervical cancer is worthy of study. This review summarizes the recent findings concerning the ion transport processes involved in cell volume regulation and intracellular Ca2+ homeostasis of epithelial cells and how these transport systems are themselves regulated by the tumor microenvironment. For cell volume regulation, we focused on the volume-sensitive Cl− channels and K+-Cl− cotransporter (KCC) family, important regulators for ionic and osmotic homeostasis of epithelial cells. Regarding intracellular Ca2+ homeostasis, the Ca2+ store sensor STIM molecules and plasma membrane Ca2+ channel Orai proteins, the predominant Ca2+ entry mechanism in epithelial cells, are discussed. Furthermore, we evaluate the potential of these membrane ion transport systems as diagnostic biomarkers and pharmacological interventions and highlight the challenges.
Collapse
|
48
|
Wang J, Zhou Y, Jiang L. Bio-inspired Track-Etched Polymeric Nanochannels: Steady-State Biosensors for Detection of Analytes. ACS NANO 2021; 15:18974-19013. [PMID: 34846138 DOI: 10.1021/acsnano.1c08582] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Bio-inspired polymeric nanochannel (also referred as nanopore)-based biosensors have attracted considerable attention on account of their controllable channel size and shape, multi-functional surface chemistry, unique ionic transport properties, and good robustness for applications. There are already very informative reviews on the latest developments in solid-state artificial nanochannel-based biosensors, however, which concentrated on the resistive-pulse sensing-based sensors for practical applications. The steady-state sensing-based nanochannel biosensors, in principle, have significant advantages over their counterparts in term of high sensitivity, fast response, target analytes with no size limit, and extensive suitable range. Furthermore, among the diverse materials, nanochannels based on polymeric materials perform outstandingly, due to flexible fabrication and wide application. This compressive Review summarizes the recent advances in bio-inspired polymeric nanochannels as sensing platforms for detection of important analytes in living organisms, to meet the high demand for high-performance biosensors for analysis of target analytes, and the potential for development of smart sensing devices. In the future, research efforts can be focused on transport mechanisms in the field of steady-state or resistive-pulse nanochannel-based sensors and on developing precisely size-controlled, robust, miniature and reusable, multi-functional, and high-throughput biosensors for practical applications. Future efforts should aim at a deeper understanding of the principles at the molecular level and incorporating these diverse pore architectures into homogeneous and defect-free multi-channel membrane systems. With the rapid advancement of nanoscience and biotechnology, we believe that many more achievements in nanochannel-based biosensors could be achieved in the near future, serving people in a better way.
Collapse
Affiliation(s)
- Jian Wang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, People's Republic of China
| | - Yahong Zhou
- Key Laboratory of Bio-inspired Materials and Interface Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, People's Republic of China
| | - Lei Jiang
- Key Laboratory of Bio-inspired Materials and Interface Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, People's Republic of China
| |
Collapse
|
49
|
Mucciolo S, Desiderato A, Salonna M, Mamos T, Prodocimo V, Di Domenico M, Mastrototaro F, Lana P, Gissi C, Calamita G. Finding Aquaporins in Annelids: An Evolutionary Analysis and a Case Study. Cells 2021; 10:3562. [PMID: 34944070 PMCID: PMC8700629 DOI: 10.3390/cells10123562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 01/26/2023] Open
Abstract
Aquaporins (AQPs) are a family of membrane channels facilitating diffusion of water and small solutes into and out of cells. Despite their biological relevance in osmoregulation and ubiquitous distribution throughout metazoans, the presence of AQPs in annelids has been poorly investigated. Here, we searched and annotated Aqp sequences in public genomes and transcriptomes of annelids, inferred their evolutionary relationships through phylogenetic analyses and discussed their putative physiological relevance. We identified a total of 401 Aqp sequences in 27 annelid species, including 367 sequences previously unrecognized as Aqps. Similar to vertebrates, phylogenetic tree reconstructions clustered these annelid Aqps in four clades: AQP1-like, AQP3-like, AQP8-like and AQP11-like. We found no clear indication of the existence of paralogs exclusive to annelids; however, several gene duplications seem to have occurred in the ancestors of some Sedentaria annelid families, mainly in the AQP1-like clade. Three of the six Aqps annotated in Alitta succinea, an estuarine annelid showing high salinity tolerance, were validated by RT-PCR sequencing, and their similarity to human AQPs was investigated at the level of "key" conserved residues and predicted three-dimensional structure. Our results suggest a diversification of the structures and functions of AQPs in Annelida comparable to that observed in other taxa.
Collapse
Affiliation(s)
- Serena Mucciolo
- Department of Invertebrate Zoology and Hydrobiology, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (A.D.); (T.M.)
- Centro de Estudos do Mar, Universidade Federal do Paraná, Av. Beira-Mar, s/n, Pontal do Sul, Pontal do Paraná 83255-976, PR, Brazil; (M.D.D.); (P.L.)
| | - Andrea Desiderato
- Department of Invertebrate Zoology and Hydrobiology, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (A.D.); (T.M.)
| | - Marika Salonna
- Institute of Medical Sciences, Foresterhill Health Campus, University of Aberdeen, Aberdeen AB25 2ZD, UK;
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari “A. Moro”, Via E. Orabona, 4, 70125 Bari, Italy; (C.G.); (G.C.)
| | - Tomasz Mamos
- Department of Invertebrate Zoology and Hydrobiology, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (A.D.); (T.M.)
| | - Viviane Prodocimo
- Laboratório de Fisiologia Comparativa da Osmorregulação, Departamento de Fisiologia, Setor de Ciências Biológicas, Campus Politécnico, Universidade Federal do Paraná, Av. Cel. Francisco H. dos Santos 100, Curitiba 81531-980, PR, Brazil;
| | - Maikon Di Domenico
- Centro de Estudos do Mar, Universidade Federal do Paraná, Av. Beira-Mar, s/n, Pontal do Sul, Pontal do Paraná 83255-976, PR, Brazil; (M.D.D.); (P.L.)
| | - Francesco Mastrototaro
- CoNISMa LRU, 70124 Bari, Italy;
- Dipartimento di Biologia, Università degli Studi di Bari “A. Moro”, 70124 Bari, Italy
| | - Paulo Lana
- Centro de Estudos do Mar, Universidade Federal do Paraná, Av. Beira-Mar, s/n, Pontal do Sul, Pontal do Paraná 83255-976, PR, Brazil; (M.D.D.); (P.L.)
| | - Carmela Gissi
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari “A. Moro”, Via E. Orabona, 4, 70125 Bari, Italy; (C.G.); (G.C.)
- CoNISMa LRU, 70124 Bari, Italy;
- IBIOM, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), CNR, Via Amendola 165/A, 70126 Bari, Italy
| | - Giuseppe Calamita
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università degli Studi di Bari “A. Moro”, Via E. Orabona, 4, 70125 Bari, Italy; (C.G.); (G.C.)
| |
Collapse
|
50
|
Kilb W. When Are Depolarizing GABAergic Responses Excitatory? Front Mol Neurosci 2021; 14:747835. [PMID: 34899178 PMCID: PMC8651619 DOI: 10.3389/fnmol.2021.747835] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Abstract
The membrane responses upon activation of GABA(A) receptors critically depend on the intracellular Cl− concentration ([Cl−]i), which is maintained by a set of transmembrane transporters for Cl−. During neuronal development, but also under several pathophysiological conditions, the prevailing expression of the Cl− loader NKCC1 and the low expression of the Cl− extruder KCC2 causes elevated [Cl−]i, which result in depolarizing GABAergic membrane responses. However, depolarizing GABAergic responses are not necessarily excitatory, as GABA(A) receptors also reduces the input resistance of neurons and thereby shunt excitatory inputs. To summarize our knowledge on the effect of depolarizing GABA responses on neuronal excitability, this review discusses theoretical considerations and experimental studies illustrating the relation between GABA conductances, GABA reversal potential and neuronal excitability. In addition, evidences for the complex spatiotemporal interaction between depolarizing GABAergic and glutamatergic inputs are described. Moreover, mechanisms that influence [Cl−]i beyond the expression of Cl− transporters are presented. And finally, several in vitro and in vivo studies that directly investigated whether GABA mediates excitation or inhibition during early developmental stages are summarized. In summary, these theoretical considerations and experimental evidences suggest that GABA can act as inhibitory neurotransmitter even under conditions that maintain substantial depolarizing membrane responses.
Collapse
Affiliation(s)
- Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|