1
|
Carretero VJ, Álvarez-Merz I, Hernández-Campano J, Kirov SA, Hernández-Guijo JM. Targeting harmful effects of non-excitatory amino acids as an alternative therapeutic strategy to reduce ischemic damage. Neural Regen Res 2025; 20:2454-2463. [PMID: 39314160 PMCID: PMC11801293 DOI: 10.4103/nrr.nrr-d-24-00536] [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: 05/11/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
The involvement of the excitatory amino acids glutamate and aspartate in cerebral ischemia and excitotoxicity is well-documented. Nevertheless, the role of non-excitatory amino acids in brain damage following a stroke or brain trauma remains largely understudied. The release of amino acids by necrotic cells in the ischemic core may contribute to the expansion of the penumbra. Our findings indicated that the reversible loss of field excitatory postsynaptic potentials caused by transient hypoxia became irreversible when exposed to a mixture of just four non-excitatory amino acids (L-alanine, glycine, L-glutamine, and L-serine) at their plasma concentrations. These amino acids induce swelling in the somas of neurons and astrocytes during hypoxia, along with permanent dendritic damage mediated by N-methyl-D-aspartate receptors. Blocking N-methyl-D-aspartate receptors prevented neuronal damage in the presence of these amino acids during hypoxia. It is likely that astroglial swelling caused by the accumulation of these amino acids via the alanine-serine-cysteine transporter 2 exchanger and system N transporters activates volume-regulated anion channels, leading to the release of excitotoxins and subsequent neuronal damage through N-methyl-D-aspartate receptor activation. Thus, previously unrecognized mechanisms involving non-excitatory amino acids may contribute to the progression and expansion of brain injury in neurological emergencies such as stroke and traumatic brain injury. Understanding these pathways could highlight new therapeutic targets to mitigate brain injury.
Collapse
Affiliation(s)
| | - Iris Álvarez-Merz
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jorge Hernández-Campano
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
| | - Sergei A. Kirov
- Department of Neuroscience and Regenerative Medicine & Department of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
| |
Collapse
|
2
|
Barroso IG, Ferreira C, Terra WR. Water Transport and Enzyme Recycling in Tenebrio molitor Midgut: Insights From Transcriptomics, Proteomics, and In Vivo Inhibition Assays. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2025; 118:e70059. [PMID: 40199745 DOI: 10.1002/arch.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025]
Abstract
The low excretory rates of secreted digestive enzymes, such as trypsins, in insect species with peritrophic membranes led to the hypothesis of ectoperitrophic countercurrent water fluxes causing enzyme recycling. The midgut water flux model of Tenebrio molitor (T. molitor) is revisited and supported by in vivo experiments. Sequences from proteins putatively involved in water transport were retrieved from the T. molitor transcriptome by Blast and analyzed using bioinformatics tools. Gene expression of selected proteins was determined in three midgut sections (anterior, AM; middle, MM; posterior, PM) by RNA-seq, and transporter proteins were verified in microvillar-membrane-enriched midgut samples by proteomics. Genes encoding three cation chloride cotransporters (CCC) and four aquaporins were expressed in the midgut. TmNaCCC2, TmPrip, and TmEglp1 showed higher expression in the front half, while TmKCC, TmNKCC1, TmDrip, and TmEglp2 were more highly expressed in the back half. However, only TmNaCCC2 was found by proteomics. Midgut water fluxes were quantified by feeding T. molitor larvae with nonabsorbable dye and measuring its concentration along the midgut. The results suggest water absorption in AM and secretion in MM and PM, potentially caused by TmNaCCC2 and TmPrip in AM, and TmKCC and TmDrip in PM, whereas MM serves as a transition region. Larvae fed on furosemide, an NKCC and KCC inhibitor, showed altered midgut water fluxes, resulting in higher trypsin excretion into the hindgut, thus reinforcing the hypothesis of a countercurrent water flux generated by CCCs powering enzyme recycling in insect midguts.
Collapse
Affiliation(s)
- Ignacio G Barroso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Clelia Ferreira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Walter R Terra
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Dienel GA, Lauritzen M. A budget for brain metabolic water production by glucose catabolism during rest, rises in activity and sleep. Fluids Barriers CNS 2025; 22:44. [PMID: 40329309 PMCID: PMC12057207 DOI: 10.1186/s12987-025-00647-8] [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: 12/15/2024] [Accepted: 03/27/2025] [Indexed: 05/08/2025] Open
Abstract
Maintaining brain fluid homeostasis is of critical importance for creating a stable environment conducive to optimal neuronal functioning, nutrient distribution, and waste product removal. In this study, we employed previously published data on brain oxygen and glucose consumption in awake rodents or humans to quantify the metabolic water production associated with distinct pathways of glucose metabolism. It is predicted that neuronal mitochondria are the primary source of metabolic water at rest, resulting in a continuous efflux into the cytosol, interstitial fluid, and cerebrospinal fluid. Net metabolic water production is predicted to be reduced by increases in activity due to a shift in metabolism from glucose oxidation to include glycolysis in neurons and ATP hydrolysis by the major cation pumps, which involves water consumption (ATP + H2O → ADP + Pi). In comparison, glycogenolysis, which occurs concurrently with the activation of astrocytes, potentially represents a major but previously unidentified contributor to metabolic water. Metabolic water production is dependent on the state of the brain, with a reduction of 30-40% occurring during deep sleep. Our estimates indicate that metabolic water functions as a conduit for interstitial fluid production within the brain, enabling flexible and efficient distribution of fluid that flows seamlessly from the parenchyma to the subarachnoid space and lymphatic vessels to facilitate the removal of brain waste, independent of the glymphatic system.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| | - Martin Lauritzen
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Cavallari E, Lorenzi E, Di Gregorio E, Ferrauto G, Aime S, Vallortigara G, Bifone A. In vivo assessment of the influence of general anesthetics on transmembrane water cycling in the brain. J Cereb Blood Flow Metab 2025; 45:977-988. [PMID: 39719068 PMCID: PMC11669147 DOI: 10.1177/0271678x241309783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/04/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024]
Abstract
This study presents the first in vivo measurement of transcytolemmal water exchange in the brain using a novel Magnetic Resonance technique. We extend previous applications of Chemical Exchange Saturation Transfer (CEST) to examine water exchange across cellular membranes in late-stage chicken embryo brains. The immature blood-brain barrier at this stage allows Gadolinium-Based Contrast Agents (GBCAs) to penetrate the brain's interstitial space, sensitizing the CEST effect to water exchange between intra- and extracellular environments. Exchange rates were measured in the awake brain and under different anaesthetic regimens, including isoflurane and ketamine/xylazine. Results show that brain water exchange is dominated by activity-dependent mechanisms, with anaesthesia reducing exchange rates by over an order of magnitude. These findings suggest that anaesthetics may impact neuronal and glial function by interfering with active transport mechanisms, potentially altering brain water homeostasis. This study highlights the utility of CEST MRI for studying dynamic biological processes in vivo.
Collapse
Affiliation(s)
- Eleonora Cavallari
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elena Lorenzi
- Center for Mind/Brain Sciences, University of Trento, Rovereto (TN), Italy
| | - Enza Di Gregorio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giuseppe Ferrauto
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Silvio Aime
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- IRCCS SDN SynLab, Napoli, Italy
| | | | - Angelo Bifone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
5
|
Selvam B, Paul A, Yu YC, Chen LQ, Shukla D. SWEET Family Transporters Act as Water-Conducting Carrier Proteins in Plants. J Chem Inf Model 2025; 65:3697-3705. [PMID: 40156514 DOI: 10.1021/acs.jcim.5c00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Dedicated water channels are involved in the facilitated diffusion of water molecules across cell membranes in plants. Transporter proteins are also known to transport water molecules along with substrates; however, the molecular mechanism of water permeation is not well understood in plant transporters. Here, we show that plant sugar transporters from the SWEET (sugar will eventually be exported transporter) family act as water-conducting carrier proteins via a variety of passive and active mechanisms that allow the diffusion of water molecules from one side of the membrane to the other. This study provides a molecular perspective on how plant membrane transporters act as water carrier proteins, a topic that has not been extensively explored in the literature. Water permeation in membrane transporters could occur via four distinct mechanisms, which form our hypothesis for water transport in SWEETs. These hypotheses are tested using molecular dynamics simulations of the outward-facing, occluded, and inward-facing states of AtSWEET1 to identify the water permeation pathways and the flux associated with them. The hydrophobic gates at the center of the transport tunnel act as barriers that restrict water permeation. We have performed in silico single and double mutations of the hydrophobic gate residues to examine the changes in water conductivity. Surprisingly, the double mutant allows water permeation to the intracellular half of the membrane and forms a continuous water channel. These computational results are validated by experimentally examining the transport of hydrogen peroxide molecules by the AtSWEET family of transporters. We have also shown that the transport of hydrogen peroxide follows a mechanism similar to that of water transport in AtSWEET1. Finally, we conclude that similar water-conduction states are also present in other SWEETs due to the high degree of sequence and structural conservation exhibited by this transporter family.
Collapse
Affiliation(s)
- Balaji Selvam
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Arnav Paul
- Department of Chemistry, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Ya-Chi Yu
- Department of Plant Biology, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Li-Qing Chen
- Department of Plant Biology, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
- Department of Chemistry, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
- Department of Plant Biology, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
6
|
Springer CS, Pike MM, Barbara TM. Metabolic Energy is Stored in a Homeostatic Trans-Membrane Water Barochemical Gradient. J Membr Biol 2025; 258:135-160. [PMID: 40009106 DOI: 10.1007/s00232-024-00332-1] [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: 08/29/2024] [Accepted: 12/13/2024] [Indexed: 02/27/2025]
Abstract
Trans-membrane water transport and co-transport is ubiquitous in cell biology. Integrated over all the cell's H2O transporters and co-transporters, the rate of homeostatic, bidirectional trans-cytolemmal water "exchange" is synchronized with the metabolic rate of the crucial Na+,K+-ATPase (NKA) enzyme: the active trans-membrane water cycling (AWC) phenomenon. Is AWC futile, or is it consequential? Conservatively representative literature metabolomic and proteinomic results enable comprehensive free energy (ΔG) calculations for the many transport reactions with known water stoichiometries. Including established intracellular pressure (Pi) magnitudes, these reveal an outward trans-membrane H2O barochemical ΔG gradient comparable to that of the well-known inward Na+ electrochemical ΔG gradient. For most co-influxers, these two gradients are finely balanced to maintain intracellular metabolite concentration values near their consuming enzyme Michaelis constants. Our analyses include glucose, glutamate-, gamma-aminobutyric acid (GABA), and lactate- transporters. 2%-4% Pi alterations can lead to disastrous metabolite concentrations. For the neurotransmitters glutamate- and GABA, very small astrocytic Pi changes can allow/disallow synaptic transmission. Unlike the Na+ and K+ electrochemical steady-states, the H2O barochemical steady-state is in (or near) chemical equilibrium. The analyses show why the presence of aquaporins (AQPs) does not dissipate trans-membrane pressure gradients. A feedback loop inherent in the opposing Na+ electrochemical and H2O barochemical gradients regulates AQP-catalyzed water flux as integral to AWC. A re-consideration of the underlying nature of Pi is also necessary. AWC is not a futile cycle but is inherent to the cell's "NKA system"-a new, fundamental aspect of biology. Metabolic energy is stored in the trans-membrane water barochemical gradient.
Collapse
Affiliation(s)
- Charles S Springer
- Advanced Imaging Research Center, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Road, L452, Portland, OR, 97239, USA.
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, USA.
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, USA.
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Martin M Pike
- Advanced Imaging Research Center, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Road, L452, Portland, OR, 97239, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Thomas M Barbara
- Advanced Imaging Research Center, Oregon Health and Science University, 3181 S. W. Sam Jackson Park Road, L452, Portland, OR, 97239, USA
| |
Collapse
|
7
|
Jensen DB, Toft‐Bertelsen TL, Barbuskaite D, Stubbe J, Nietzsche S, Capion T, Norager NH, Olsen MH, Sørensen AT, Dimke H, Hübner CA, Juhler M, MacAulay N. The Na +,K +,2Cl - Cotransporter, Not Aquaporin 1, Sustains Cerebrospinal Fluid Secretion While Controlling Brain K + Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409120. [PMID: 39692709 PMCID: PMC11809428 DOI: 10.1002/advs.202409120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/11/2024] [Indexed: 12/19/2024]
Abstract
Disturbances in the brain fluid balance can lead to life-threatening elevation in intracranial pressure (ICP), which represents a vast clinical challenge. Targeted and efficient pharmaceutical therapy of elevated ICP is not currently available, as the molecular mechanisms governing cerebrospinal fluid (CSF) secretion are largely unresolved. To resolve the quantitative contribution of key choroid plexus transport proteins, this study employs mice with genetic knockout and/or viral choroid plexus-specific knockdown of aquaporin 1 (AQP1) and the Na+, K+, 2Cl- cotransporter 1 (NKCC1) for in vivo determinations of CSF dynamics, ex vivo choroid plexus for transporter-mediated clearance of a CSF K+ load, and patient CSF for [K+] quantification. CSF secretion and ICP management occur independently of choroid plexus AQP1 expression, whereas both parameters are reduced by 40% upon choroid plexus NKCC1 knockdown. Elevation of [K+]CSF increases the choroid plexus Na+/K+-ATPase activity, and favors inwardly-directed net NKCC1 transport, which, together, promote CSF K+ clearance, while maintaining undisturbed CSF secretion rates. CSF from patients with post-hemorrhagic hydrocephalus does not display elevated [K+]CSF, suggesting that NKCC1 maintains net outward transport direction during post-hemorrhagic hydrocephalus formation. Direct or indirect therapeutic modulation of choroid plexus NKCC1 can thus be a potential promising pharmacological approach against brain pathologies associated with elevated ICP.
Collapse
Affiliation(s)
- Dennis Bo Jensen
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen N2200Denmark
| | | | - Dagne Barbuskaite
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen N2200Denmark
| | - Jane Stubbe
- Department of Molecular MedicineUniversity of Southern DenmarkCampusvej 55Odense5230Denmark
| | - Sandor Nietzsche
- Center for Electron MicroscopyJena University HospitalZiegelmühlenweg 107743JenaGermany
| | - Tenna Capion
- Department of NeurosurgeryUniversity Hospital of Copenhagen – RigshospitaletBlegdamsvej 9Copenhagen2100Denmark
| | - Nicolas H. Norager
- Department of NeurosurgeryUniversity Hospital of Copenhagen – RigshospitaletBlegdamsvej 9Copenhagen2100Denmark
| | - Markus H. Olsen
- Department of NeuroanaesthesiologyUniversity Hospital of Copenhagen – RigshospitaletBlegdamsvej 9Copenhagen2100Denmark
| | - Andreas T. Sørensen
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen N2200Denmark
| | - Henrik Dimke
- Department of NephrologyOdense University HospitalJ.B. Winsløws Vej 4Odense5000Denmark
| | - Christian A. Hübner
- HübnerInstitute of Human GeneticsJena University HospitalAm Klinikum 107747JenaGermany
| | - Marianne Juhler
- Department of NeurosurgeryUniversity Hospital of Copenhagen – RigshospitaletBlegdamsvej 9Copenhagen2100Denmark
- Department of Clinical MedicineUniversity of CopenhagenBlegdamsvej 3Copenhagen2200Denmark
| | - Nanna MacAulay
- Department of NeuroscienceUniversity of CopenhagenBlegdamsvej 3Copenhagen N2200Denmark
| |
Collapse
|
8
|
Gomez FD, Reppetti J, Alvarez RS, Girón Reyes DC, Sacerdoti F, Balestracci A, Damiano AE, Martínez NA, Di Giusto G, Amaral MM. Involvement of aquaporins in Shiga toxin-induced swelling and water transport dysfunction in human renal microvascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119866. [PMID: 39442808 DOI: 10.1016/j.bbamcr.2024.119866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
One of the hallmarks of Shiga toxin-producing Escherichia coli-associated hemolytic uremic syndrome (STEC-HUS) is kidney damage. Our previous research demonstrated that Shiga toxin type 2 (Stx2a) decreases cell viability and induces swelling of human glomerular endothelial cells (HGEC). However, Stx2a can disrupt net water transport across HGEC monolayers without affecting cell viability. This work aimed to elucidate the possible mechanisms involved in the water transport disruption caused by Stx2a across HGEC monolayers. We investigated paracellular and transcellular water transfer across HGEC by analyzing the passage of FITC-Dextran and the hydrostatic pressure (Phydr) and measuring the osmotic pressure (Posm), respectively. Stx2a selectively affected the transcellular pathway without impacting the paracellular route. Furthermore, Stx2a cell swelling was prevented by pretreatment with aquaporin inhibitors tetraethylammonium chloride (TEA), Mercury (II) chloride (HgCl2) or TGN-020, suggesting aquaporin involvement in this process. Confocal microscopy revealed that Stx2a increased HGEC total volume, which TEA and TGN-020 counteracted. Additionally, we identified in HGEC not only the expression of aquaporin-1 (AQP1) but also the expression of aquaporin-4 (AQP4). Surprisingly, we observed a decrease in the expression of both AQPs after Stx2a exposure. Our findings suggest that Stx2a may induce water movement into HGEC via AQP1 and AQP4, increasing total cell volume. Subsequently, decreased AQP1 and AQP4 expression could inhibit transcellular water transfer, potentially as a protective mechanism against excessive water entry and cell lysis.
Collapse
Affiliation(s)
- Fernando D Gomez
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Fisiopatogenia, Buenos Aires C1121ABG, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina
| | - Julieta Reppetti
- CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Médicas, Laboratorio de Biología de la Reproducción, Buenos Aires C1121ABG, Argentina
| | - Romina S Alvarez
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Fisiopatogenia, Buenos Aires C1121ABG, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina
| | - Daniel C Girón Reyes
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Fisiopatogenia, Buenos Aires C1121ABG, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina
| | - Flavia Sacerdoti
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Fisiopatogenia, Buenos Aires C1121ABG, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina
| | - Alejandro Balestracci
- Unidad de Nefrología, Hospital General de Niños Pedro de Elizalde, Buenos Aires C1270AAN, Argentina
| | - Alicia E Damiano
- CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Médicas, Laboratorio de Biología de la Reproducción, Buenos Aires C1121ABG, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Buenos Aires C1113AAD, Argentina
| | - Nora A Martínez
- CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Médicas, Laboratorio de Biología de la Reproducción, Buenos Aires C1121ABG, Argentina
| | - Gisela Di Giusto
- CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Buenos Aires C1121ABG, Argentina
| | - María M Amaral
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Ciencias Fisiológicas, Laboratorio de Fisiopatogenia, Buenos Aires C1121ABG, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Buenos Aires C1121ABG, Argentina.
| |
Collapse
|
9
|
Barroso IG, Nascimento BB, Ferreira C, Terra WR. Water fluxes and nutrient absorption along the midgut of three hemipterans, Mahanarva fimbriolata, Dysdercus peruvianus, and Rhodnius prolixus. Comp Biochem Physiol A Mol Integr Physiol 2025; 299:111773. [PMID: 39515658 DOI: 10.1016/j.cbpa.2024.111773] [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: 07/03/2024] [Revised: 09/16/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Hemiptera Order comprises insect species adapted to different diets regarding water and nutrient content and availability, thus suggesting different combinations of proteins to ensure their absorption. To find out whether hemipterans use the same or distinct set of proteins and whether these differences are related to the phylogeny or the diet, RNAseq analyses were conducted in gut sections of three hemipterans, M. fimbriolata, D. peruvianus, and R. prolixus, with remarkable distinct diet. Since only a few of the selected proteins were functionally characterized, the coded putative proteins were manually curated by bioinformatics to infer their physiological function. The results suggest a relationship between gene expression patterns and water and nutrient dietary content and availability. In contrast, putative gene expansions and deletions are related to phylogeny, corresponding to evolutionary adaptations of ancestral forms to feed on xylem, cotton seeds, and blood, resulting in more resemblances between D. peruvianus and R. prolixus than M. fimbriolata. M. fimbriolata absorbs water through aquaporins Drip and Prip in the filtration chamber by passive diffusion, with a higher contribution of water-selective Drip. D. peruvianus water absorption involves Drip and Prip, but Prip contribution appears to be higher, and they probably cooperate with water-ion cotransporters in the posterior midgut. R. prolixus absorbs water in the anterior midgut involving a sodium transporter and a putative water-urea Prip. Sugars, amino acids, and lipids might be absorbed along the midgut in the three species, with a higher contribution of the posterior midgut for amino acid and lipid absorption in M. fimbriolata and D. peruvianus and the middle midgut in R. prolixus.
Collapse
Affiliation(s)
- Ignacio G Barroso
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av.Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Bárbara B Nascimento
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av.Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Clelia Ferreira
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av.Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Walter R Terra
- Departamento de Bioquimica, Instituto de Quimica, Universidade de São Paulo, Av.Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil.
| |
Collapse
|
10
|
Ouellet V, Doyon N, Godin AG, Marquet P. Mathematical properties of pump-leak-cotransport models. J Math Biol 2024; 90:2. [PMID: 39625537 DOI: 10.1007/s00285-024-02163-z] [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: 10/23/2023] [Revised: 09/13/2024] [Accepted: 11/08/2024] [Indexed: 01/28/2025]
Abstract
Models of ordinary differential equations are often used to describe the electrical, ionic and volumetric responses of cells to external stimuli. Although these cellular models are often solved numerically, rigorous evidence regarding their steady state solutions is scarce. In this work, we provide a formalism defining the conditions ensuring the existence and uniqueness of a steady-state solution in a large class of models including leak channels, a pump and cotransporters. Our work generalizes previous results and provides explicit conditions that a model must satisfy to guarantee the existence and uniqueness of a steady state.
Collapse
Affiliation(s)
- Vincent Ouellet
- CERVO Brain Research Centre, Université Laval, rue de la Canardière, Québec, Québec, G1J 2G3, Canada.
- Département de psychiatrie et de neurosciences, Université Laval, Avenue de la Médecine, Québec, Québec, G1V 0A6, Canada.
| | - Nicolas Doyon
- CERVO Brain Research Centre, Université Laval, rue de la Canardière, Québec, Québec, G1J 2G3, Canada.
- Département de mathématiques et de statistique, Université Laval, Avenue de la Médecine, Québec, Québec, G1V 0A6, Canada.
| | - Antoine G Godin
- CERVO Brain Research Centre, Université Laval, rue de la Canardière, Québec, Québec, G1J 2G3, Canada
- Département de psychiatrie et de neurosciences, Université Laval, Avenue de la Médecine, Québec, Québec, G1V 0A6, Canada
| | - Pierre Marquet
- CERVO Brain Research Centre, Université Laval, rue de la Canardière, Québec, Québec, G1J 2G3, Canada
- Département de psychiatrie et de neurosciences, Université Laval, Avenue de la Médecine, Québec, Québec, G1V 0A6, Canada
| |
Collapse
|
11
|
Wu X, He Q, Yin Y, Tan S, Zhang B, Li W, Hsu YC, Xue R, Bai R. Relaxation-exchange magnetic resonance imaging (REXI): a non-invasive imaging method for evaluating trans-barrier water exchange in the choroid plexus. Fluids Barriers CNS 2024; 21:94. [PMID: 39593112 PMCID: PMC11590242 DOI: 10.1186/s12987-024-00589-7] [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: 07/30/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The choroid plexus (CP) plays a crucial role in cerebrospinal fluid (CSF) production and brain homeostasis. However, non-invasive imaging techniques to assess its function remain limited. This study was conducted to develop a novel, contrast-agent-free MRI technique, termed relaxation-exchange magnetic resonance imaging (REXI), for evaluating CP-CSF water transport, a potential biomarker of CP function. METHODS REXI utilizes the inherent and large difference in magnetic resonance transverse relaxation times (T2s) between CP tissue (e.g., blood vessels and epithelial cells) and CSF. It uses a filter block to remove most CP tissue magnetization (shorter T2), a mixing block for CP-CSF water exchange with mixing time tm, and a detection block with multi-echo acquisition to determine the CP/CSF component fraction after exchange. The REXI pulse sequence was implemented on a 9.4 T preclinical MRI scanner. For validation of REXI's ability to measure exchange, we conducted preliminary tests on urea-water proton-exchange phantoms with various pH levels. We measured the steady-state water efflux rate from CP to CSF in rats and tested the sensitivity of REXI in detecting CP dysfunction induced by the carbonic anhydrase inhibitor acetazolamide. RESULTS REXI pulse sequence successfully captured changes in the proton exchange rate (from short-T2 component to long-T2 component [i.e., ksl]) of urea-water phantoms at varying pH, demonstrating its sensitivity to exchange processes. In rat CP, REXI significantly suppressed the CP tissue signal, reducing the short-T2 fraction (fshort) from 0.44 to 0.23 (p < 0.0001), with significant recovery to 0.28 after a mixing time of 400 ms (p = 0.014). The changes in fshort at various mixing times can be accurately described by a two-site exchange model, yielding a steady-state water efflux rate from CP to CSF (i.e., kbc) of 0.49 s-1. A scan-rescan experiment demonstrated that REXI had excellent reproducibility in measuring kbc (intraclass correlation coefficient = 0.90). Notably, acetazolamide-induced CSF reduction resulted in a 66% decrease in kbc within rat CP. CONCLUSIONS This proof-of-concept study demonstrates the feasibility of REXI for measuring trans-barrier water exchange in the CP, offering a promising biomarker for future assessments of CP function.
Collapse
Affiliation(s)
- Xuetao Wu
- State Key Laboratory of Brain and Cognitive Science, Beijing MRI Center for Brain Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingping He
- School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Interdisciplinary Institute of Neuroscience and Technology and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Yin
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Shuyuan Tan
- Key Laboratory of Biomedical Engineering of Education Ministry, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Baogui Zhang
- State Key Laboratory of Brain and Cognitive Science, Beijing MRI Center for Brain Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Weiyun Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yi-Cheng Hsu
- MR Collaboration, Siemens Healthcare, Shanghai, China
| | - Rong Xue
- State Key Laboratory of Brain and Cognitive Science, Beijing MRI Center for Brain Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Ruiliang Bai
- Interdisciplinary Institute of Neuroscience and Technology and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
12
|
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
|
13
|
Selvam B, Paul A, Yu YC, Chen LQ, Shukla D. SWEET family transporters act as water conducting carrier proteins in plants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.23.600272. [PMID: 38979333 PMCID: PMC11230166 DOI: 10.1101/2024.06.23.600272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Dedicated water channels are involved in the facilitated diffusion of water molecules across the cell membrane in plants. Transporter proteins are also known to transport water molecules along with substrates, however the molecular mechanism of water permeation is not well understood in plant transporters. Here, we show plant sugar transporters from the SWEET (Sugar Will Eventually be Exported Transporter) family act as water-conducting carrier proteins via a variety of passive and active mechanisms that allow diffusion of water molecules from one side of the membrane to the other. This study provides a molecular perspective on how plant membrane transporters act as water carrier proteins, a topic that has not been extensively explored in literature. Water permeation in membrane transporters could occur via four distinct mechanisms which form our hypothesis for water transport in SWEETs. These hypothesis are tested using molecular dynamics simulations of the outward-facing, occluded, and inward-facing state of AtSWEET1 to identify the water permeation pathways and the flux associated with them. The hydrophobic gates at the center of the transport tunnel act as a barrier that restricts water permeation. We have performed in silico single and double mutations of the hydrophobic gate residues to examine the changes in the water conductivity. Surprisingly, the double mutant allows the water permeation to the intracellular half of the membrane and forms a continuous water channel. These computational results are validated by experimentally examining the transport of hydrogen peroxide molecules by the AtSWEET family of transporters. We have also shown that the transport of hydrogen peroxide follows the similar mechanism as water transport in AtSWEET1. Finally, we conclude that similar water-conduction states are also present in other SWEET transporters due to the high sequence and structure conservation exhibited by this transporter family.
Collapse
Affiliation(s)
- Balaji Selvam
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Arnav Paul
- Department of Chemistry, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Ya-Chi Yu
- Department of Plant Biology, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Li-Qing Chen
- Department of Plant Biology, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
- Department of Plant Biology, University of Illinois at Urbana-Champaign, Champaign, Illinois 61801, United States
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| |
Collapse
|
14
|
Springer CS, Pike MM, Barbara TM. A Futile Cycle?: Tissue Homeostatic Trans-Membrane Water Co-Transport: Kinetics, Thermodynamics, Metabolic Consequences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589812. [PMID: 38659823 PMCID: PMC11042311 DOI: 10.1101/2024.04.17.589812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The phenomenon of active trans-membrane water cycling (AWC) has emerged in little over a decade. Here, we consider H2O transport across cell membranes from the origins of its study. Historically, trans-membrane water transport processes were classified into: A) compensating bidirectional fluxes ("exchange"), and B) unidirectional flux ("net flow") categories. Recent literature molecular structure determinations and molecular dynamic (MD) simulations indicate probably all the many different hydrophilic substrate membrane co-transporters have membrane-spanning hydrophilic pathways and co-transport water along with their substrates, and that they individually catalyze category A and/or B water flux processes, although usually not simultaneously. The AWC name signifies that, integrated over the all the cell's co-transporters, the rate of homeostatic, bidirectional trans-cytolemmal water exchange (category A) is synchronized with the metabolic rate of the crucial Na+,K+-ATPase (NKA) enzyme. A literature survey indicates the stoichiometric (category B) water/substrate ratios of individual co-transporters are often very large. The MD simulations also suggest how different co-transporter reactions can be kinetically coupled molecularly. Is this (Na+,K+-ATPase rate-synchronized) cycling futile, or is it consequential? Conservatively representative literature metabolomic and proteinomic results enable comprehensive free energy analyses of the many transport reactions with known water stoichiometries. Free energy calculations, using literature intracellular pressure (Pi) values reveals there is an outward trans-membrane H2O barochemical gradient of magnitude comparable to that of the well-known inward Na+ electrochemical gradient. For most co-influxers, these gradients are finely balanced to maintain intracellular metabolite concentration values near their consuming enzyme Michaelis constants. The thermodynamic analyses include glucose, glutamate-, gamma-aminobutyric acid (GABA), and lactate- transporters. 2%-4% Pi alterations can lead to disastrous concentration levels. For the neurotransmitters glutamate- and GABA, very small astrocytic Pi changes can allow/disallow synaptic transmission. Unlike the Na+ and K+ electrochemical steady-states, the H2O barochemical steady-state is in (or near) chemical equilibrium. The analyses show why the presence of aquaporins (AQPs) does not dissipate the trans-membrane pressure gradient. A feedback loop inherent in the opposing Na+ electrochemical and H2O barochemical gradients regulates AQP-catalyzed water flux as an integral AWC aspect. These results also require a re-consideration of the underlying nature of Pi. Active trans-membrane water cycling is not futile, but is inherent to the cell's "NKA system" - a new, fundamental aspect of biology.
Collapse
Affiliation(s)
- Charles S Springer
- Advanced Imaging Research Center
- Department of Chemical Physiology and Biochemistry
- Department of Biomedical Engineering
- Brenden-Colson Center for Pancreatic Care
- Knight Cancer Institute, Oregon Health & Science University; Portland, Oregon
| | - Martin M Pike
- Advanced Imaging Research Center
- Department of Biomedical Engineering
- Knight Cancer Institute, Oregon Health & Science University; Portland, Oregon
| | | |
Collapse
|
15
|
Jensen JH. Diffusional kurtosis time dependence and the water exchange rate for the multi-compartment Kärger model. Magn Reson Med 2024; 91:1122-1135. [PMID: 37957820 PMCID: PMC11027117 DOI: 10.1002/mrm.29926] [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: 07/31/2023] [Revised: 10/02/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023]
Abstract
PURPOSE To demonstrate an analytic formula giving the time dependence of the diffusional kurtosis for the Kärger model (KM) with an arbitrary number of exchanging compartments and its application in estimating the mean KM water exchange rate. THEORY AND METHODS The general formula for the kurtosis is derived from a power series solution for the multi-compartment KM. A lower bound on the exchange rate is established from the observation that the kurtosis is always a logarithmically convex function of time. Both the kurtosis time dependence and the lower bound are illustrated with numerical calculations. The lower bound is also applied to previously published data for the time dependence of the kurtosis in both brain and tumors. RESULTS The kurtosis for the multi-compartment KM is given by a sum in which each term is associated with an eigenvector of the exchange rate matrix. The lower bound is determined from the most negative value for the logarithmic derivative of the kurtosis with respect to time. In the cerebral cortex, the lower bound is found to vary from 15 to 76 s-1 , depending on the experimental details, while for the tumors considered, it varies from 2 to 4 s-1 . CONCLUSION The time dependence of the kurtosis for the multi-compartment KM has a simple analytic solution that allows a lower bound for the mean KM water exchange rate to be determined directly from experiment. This may be useful in tissues with complex microstructure that is difficult to model explicitly.
Collapse
Affiliation(s)
- Jens H. Jensen
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, South Carolina
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
16
|
Di Gregorio E, Papi C, Conti L, Di Lorenzo A, Cavallari E, Salvatore M, Cavaliere C, Ferrauto G, Aime S. A Magnetic Resonance Imaging-Chemical Exchange Saturation Transfer (MRI-CEST) Method for the Detection of Water Cycling across Cellular Membranes. Angew Chem Int Ed Engl 2024; 63:e202313485. [PMID: 37905585 DOI: 10.1002/anie.202313485] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/02/2023]
Abstract
Water cycling across the membrane transporters is considered a hallmark of cellular metabolism and it could be of high diagnostic relevance in the characterization of tumors and other diseases. The method relies on the response of intracellular proton exchanging molecules to the presence of extracellular Gd-based contrast agents (GBCAs). Paramagnetic GBCAs enhances the relaxation rate of water molecules in the extracellular compartment and, through membrane exchange, the relaxation enhancement is transferred to intracellular molecules. The effect is detected at the MRI-CEST (Magnetic Resonance Imaging - Chemical Exchange Saturation Transfer) signal of intracellular proton exchanging molecules. The magnitude of the change in the CEST response reports on water cycling across the membrane. The method has been tested on Red Blood Cells and on orthotopic murine models of breast cancer with different degree of malignancy (4T1, TS/A and 168FARN). The distribution of voxels reporting on membrane permeability fits well with the cells' aggressiveness and acts as an early reporter to monitor therapeutic treatments.
Collapse
Affiliation(s)
- Enza Di Gregorio
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Chiara Papi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Laura Conti
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Antonino Di Lorenzo
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Eleonora Cavallari
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Marco Salvatore
- IRCCS SDN SynLab, Via E. Gianturco 113, 80143, Napoli, Italy
| | - Carlo Cavaliere
- IRCCS SDN SynLab, Via E. Gianturco 113, 80143, Napoli, Italy
| | - Giuseppe Ferrauto
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Silvio Aime
- IRCCS SDN SynLab, Via E. Gianturco 113, 80143, Napoli, Italy
| |
Collapse
|
17
|
Wei Y, Wei H, Tian C, Wu Q, Li D, Huang C, Zhang G, Chen R, Wang N, Li Y, Li B, Chu XM. The Transcriptome Analysis of Circular RNAs Between the Doxorubicin- Induced Cardiomyocytes and Bone Marrow Mesenchymal Stem Cells- Derived Exosomes Treated Ones. Comb Chem High Throughput Screen 2024; 27:1056-1070. [PMID: 38305398 DOI: 10.2174/0113862073261891231115072310] [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: 07/19/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 02/03/2024]
Abstract
AIM To analyze the sequencing results of circular RNAs (circRNAs) in cardiomyocytes between the doxorubicin (DOX)-injured group and exosomes treatment group. Moreover, to offer potential circRNAs possibly secreted by exosomes mediating the therapeutic effect on DOX-induced cardiotoxicity for further study. METHODS The DOX-injured group (DOX group) of cardiomyocytes was treated with DOX, while an exosomes-treated group of injured cardiomyocytes were cocultured with bone marrow mesenchymal stem cells (BMSC)-derived exosomes (BEC group). The high-throughput sequencing of circRNAs was conducted after the extraction of RNA from cardiomyocytes. The differential expression of circRNA was analyzed after identifying the number, expression, and conservative of circRNAs. Then, the target genes of differentially expressed circRNAs were predicted based on the targetscan and Miranda database. Next, the GO and KEGG enrichment analyses of target genes of circRNAs were performed. The crucial signaling pathways participating in the therapeutic process were identified. Finally, a real-time quantitative polymerase chain reaction experiment was conducted to verify the results obtained by sequencing. RESULTS Thirty-two circRNAs are differentially expressed between the two groups, of which twenty-three circRNAs were elevated in the exosomes-treated group (BEC group). The GO analysis shows that target genes of differentially expressed circRNAs are mainly enriched in the intracellular signalactivity, regulation of nucleic acid-templated transcription, Golgi-related activity, and GTPase activator activity. The KEGG analysis displays that they were involved in the autophagy biological process and NOD-like receptor signaling pathway. The verification experiment suggested that mmu_circ_0000425 (ID: 116324210) was both decreased in the DOX group and elevated in BEC group, which was consistent with the result of sequencing. CONCLUSION mmu_circ_0000425 in exosomes derived from bone marrow mesenchymal stem cells (BMSC) may have a therapeutic role in alleviating doxorubicin-induced cardiotoxicity (DIC).
Collapse
Affiliation(s)
- Yanhuan Wei
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Emergency Medicine, Rizhao People's Hospital, Rizhao, China
| | - Haixia Wei
- Qingdao Chengyang People's Hospital, Qingdao, China
| | - Chao Tian
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Qinchao Wu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Huang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guoliang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruolan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ni Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yonghong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Li
- Department of Genetics, Basic Medicine School, Qingdao University, Qingdao, China
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
18
|
MacAulay N, Toft-Bertelsen TL. Dual function of the choroid plexus: Cerebrospinal fluid production and control of brain ion homeostasis. Cell Calcium 2023; 116:102797. [PMID: 37801806 DOI: 10.1016/j.ceca.2023.102797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/08/2023]
Abstract
The choroid plexus is a small monolayered epithelium located in the brain ventricles and serves to secrete the cerebrospinal fluid (CSF) that envelops the brain and fills the central ventricles. The CSF secretion is sustained with a concerted effort of a range of membrane transporters located in a polarized fashion in this tissue. Prominent amongst these are the Na+/K+-ATPase, the Na+,K+,2Cl- cotransporter (NKCC1), and several HCO3- transporters, which together support the net transepithelial transport of the major electrolytes, Na+ and Cl-, and thus drive the CSF secretion. The choroid plexus, in addition, serves an important role in keeping the CSF K+ concentration at a level compatible with normal brain function. The choroid plexus Na+/K+-ATPase represents a key factor in the barrier-mediated control of the CSF K+ homeostasis, as it increases its K+ uptake activity when faced with elevated extracellular K+ ([K+]o). In certain developmental or pathological conditions, the NKCC1 may revert its net transport direction to contribute to CSF K+ homeostasis. The choroid plexus ion transport machinery thus serves dual, yet interconnected, functions with its contribution to electrolyte and fluid secretion in combination with its control of brain K+ levels.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen 2200, Denmark.
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen 2200, Denmark
| |
Collapse
|
19
|
Zhu C, Nie X, Lu Q, Bai Y, Jiang Z. Roles and regulation of Aquaporin-3 in maintaining the gut health: an updated review. Front Physiol 2023; 14:1264570. [PMID: 38089478 PMCID: PMC10714013 DOI: 10.3389/fphys.2023.1264570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2025] Open
Abstract
Aquaporin-3 (AQP3) is a predominant water channel protein expressed in the intestine, and plays important roles in the gut physiology and pathophysiology due to its permeability to water, glycerol and hydrogen peroxide. In this review, we systematically summarized the current understanding of the expression of AQP3 in the intestine of different species, and focused on the potential roles of AQP3 in water transport, different types of diarrhea and constipation, intestinal inflammation, intestinal barrier function, oxidative stress, and autophagy. These updated findings have supported that AQP3 may function as an important target in maintaining gut health of human and animals.
Collapse
Affiliation(s)
- Cui Zhu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Xiaoyan Nie
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Qi Lu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yinshan Bai
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
20
|
Israelsen IME, Kamp-Jensen C, Westgate CSJ, Styrishave B, Jensen RH, Eftekhari S. Cycle-dependent sex differences in expression of membrane proteins involved in cerebrospinal fluid secretion at rat choroid plexus. BMC Neurosci 2023; 24:60. [PMID: 37946101 PMCID: PMC10633912 DOI: 10.1186/s12868-023-00829-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: 06/15/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Female sex is a known risk factor of brain disorders with raised intracranial pressure (ICP) and sex hormones have been suggested to alter cerebrospinal fluid (CSF) dynamics, thus impairing ICP regulation in CSF disorders such as idiopathic intracranial hypertension (IIH). The choroid plexus (CP) is the tissue producing CSF and it has been hypothesized that altered hormonal composition could affect the activity of transporters involved in CSF secretion, thus affecting ICP. Therefore, we aimed to investigate if expression of various transporters involved in CSF secretion at CP were different between males and females and between females in different estrous cycle states. Steroid levels in serum was also investigated. METHODS Female and male rats were used to determine sex-differences in the genes encoding for the transporters Aqp1 and 4, NKCC1, NBCe2, NCBE; carbonic anhydrase enzymes II and III (CA), subunits of the Na+/K+-ATPase including Atp1a1, Atp1b1 and Fxyd1 at CP. The estrous cycle stage metestrus (MET) and estrous (ES) were determined before euthanasia. Serum and CP were collected and subjected to RT-qPCR analysis and western blots. Serum was used to measure steroid levels using liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS Significant differences in gene expression and steroid levels between males and ES females were found, while no differences were found between male and MET females. During ES, expression of Aqp1 was lower (p < 0.01) and NKCC1 was higher in females compared to males. CAII was lower while CAIII was higher in ES females (p < 0.0001). Gene expression of Atp1a1 was lower in ES compared to male (p = 0.0008). Several of these choroidal genes were also significantly different in MET compared to females in ES. Differences in gene expression during the estrus cycle were correlated to serum level of steroid hormones. Protein expression of AQP1 (p = 0.008) and CAII (p = 0.035) was reduced in ES females compared to males. CONCLUSIONS This study demonstrates for the first time that expression at CP is sex-dependent and markedly affected by the estrous cycle in female rats. Further, expression was related to hormone levels in serum. This opens a completely new avenue for steroid regulation of the expression of CSF transporters and the close link to the understanding of CSF disorders such as IIH.
Collapse
Affiliation(s)
- Ida Marchen Egerod Israelsen
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Christina Kamp-Jensen
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Connar Stanley James Westgate
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Bjarne Styrishave
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rigmor H Jensen
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Sajedeh Eftekhari
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark.
| |
Collapse
|
21
|
Sever M, Merzel F. Collective Domain Motion Facilitates Water Transport in SGLT1. Int J Mol Sci 2023; 24:10528. [PMID: 37445706 DOI: 10.3390/ijms241310528] [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: 05/05/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The human sodium-glucose cotransporter protein (SGLT1) is an important representative of the sodium solute symporters belonging to the secondary active transporters that are critical to the homeostasis of sugar, sodium, and water in the cell. The underlying transport mechanism of SGLT1 is based on switching between inward- and outward-facing conformations, known as the alternating access model, which is crucial for substrate transport, and has also been postulated for water permeation. However, the nature of water transport remains unclear and is disputed along the passive and active transport, with the latter postulating the presence of the pumping effect. To better examine the water transport in SGLT1, we performed a series of equilibrium all-atom molecular dynamics simulations, totaling over 6 μs of sample representative conformational states of SGLT1 and its complexes, with the natural substrates, ions, and inhibitors. In addition to elucidating the basic physical factors influencing water permeation, such as channel openings and energetics, we focus on dynamic flexibility and its relationship with domain motion. Our results clearly demonstrate a dependence of instantaneous water flux on the channel opening and local water diffusion in the channel, strongly supporting the existence of a passive water transport in SGLT1. In addition, a strong correlation found between the local water diffusion and protein domain motion, resembling the "rocking-bundle" motion, reveals its facilitating role in the water transport.
Collapse
Affiliation(s)
- Marko Sever
- Theory Departnemt, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Franci Merzel
- Theory Departnemt, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| |
Collapse
|
22
|
Blazer-Yost BL. Consideration of Kinase Inhibitors for the Treatment of Hydrocephalus. Int J Mol Sci 2023; 24:ijms24076673. [PMID: 37047646 PMCID: PMC10094860 DOI: 10.3390/ijms24076673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
Hydrocephalus is a devastating condition characterized by excess cerebrospinal fluid (CSF) in the brain. Currently, the only effective treatment is surgical intervention, usually involving shunt placement, a procedure prone to malfunction, blockage, and infection that requires additional, often repetitive, surgeries. There are no long-term pharmaceutical treatments for hydrocephalus. To initiate an intelligent drug design, it is necessary to understand the biochemical changes underlying the pathology of this chronic condition. One potential commonality in the various forms of hydrocephalus is an imbalance in fluid–electrolyte homeostasis. The choroid plexus, a complex tissue found in the brain ventricles, is one of the most secretory tissues in the body, producing approximately 500 mL of CSF per day in an adult human. In this manuscript, two key transport proteins of the choroid plexus epithelial cells, transient receptor potential vanilloid 4 and sodium, potassium, 2 chloride co-transporter 1, will be considered. Both appear to play key roles in CSF production, and their inhibition or genetic manipulation has been shown to affect CSF volume. As with most transporters, these proteins are regulated by kinases. Therefore, specific kinase inhibitors are also potential targets for the development of pharmaceuticals to treat hydrocephalus.
Collapse
Affiliation(s)
- Bonnie L. Blazer-Yost
- Biology Department, Indiana University—Purdue University, 723 West Michigan Street, Indianapolis, IN 46202, USA
| |
Collapse
|
23
|
Stoeckelhuber M, Grill FD, Wolff KD, Kesting MR, Wolff CT, Fichter AM, Loeffelbein DJ, Schmitz C, Ritschl LM. Infantile human labial glands: Distribution of aquaporins and claudins in the context of paracellular and transcellular pathways. Tissue Cell 2023; 82:102052. [PMID: 36905859 DOI: 10.1016/j.tice.2023.102052] [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: 12/13/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023]
Abstract
Human labial glands consist of saliva-secreting cells which are formed by serous and predominantly mucous glandular cells. The following excretory duct system converts the isotonic saliva into a hypotonic fluid. Liquids are transported across the membrane of epithelial cells by paracellular or transcellular mode of action. We studied aquaporins (AQP) and tight junction proteins in the endpieces and duct system of human labial glands of 3-5-month-old infants for the first time. AQP1, AQP3, and AQP5 represent the transcellular transport; tight junction proteins like claudin-1, - 3, - 4, and - 7 regulate the permeability of the paracellular pathway. Specimens of 28 infants were included in this study and analyzed histologically. AQP1 was present in myoepithelial cells and in endothelial cells of small blood vessels. AQP3 showed basolateral plasmamembrane localization in glandular endpieces. AQP5 was localized at the apical cytomembrane in serous and mucous glandular cells and at the lateral membrane in serous cells. Ducts remained unstained with the antibody to AQP1, AQP3, and AQP5. Claudin-1, - 3, - 4, and - 7 were expressed mainly in the lateral plasmamembrane of serous glandular cells. In the ducts, claudin-1, - 4, and - 7 were detected at the basal cell layer, claudin-7 also at the lateral cytomembrane. Our findings provide new insights into the localization of epithelial barrier components necessary for regulating saliva-modification in infantile labial glands.
Collapse
Affiliation(s)
- Mechthild Stoeckelhuber
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
| | - Florian D Grill
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Klaus-Dietrich Wolff
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marco R Kesting
- Department of Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuernberg, Erlangen, Germany
| | - Constantin T Wolff
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Andreas M Fichter
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Denys J Loeffelbein
- Department of Oral and Maxillofacial Surgery, Helios Hospital Munich West, Teaching Hospital of Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Christoph Schmitz
- Department of Anatomy II, Faculty of Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Lucas M Ritschl
- Department of Oral and Maxillofacial Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
24
|
Role of NKCC1 and KCC2 during hypoxia-induced neuronal swelling in the neonatal neocortex. Neurobiol Dis 2023; 178:106013. [PMID: 36706928 PMCID: PMC9945323 DOI: 10.1016/j.nbd.2023.106013] [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: 11/07/2022] [Revised: 12/21/2022] [Accepted: 01/22/2023] [Indexed: 01/26/2023] Open
Abstract
Neonatal hypoxia causes cytotoxic neuronal swelling by the entry of ions and water. Multiple water pathways have been implicated in neurons because these cells lack water channels, and their membrane has a low water permeability. NKCC1 and KCC2 are cation-chloride cotransporters (CCCs) involved in water movement in various cell types. However, the role of CCCs in water movement in neonatal neurons during hypoxia is unknown. We studied the effects of modulating CCCs pharmacologically on neuronal swelling in the neocortex (layer IV/V) of neonatal mice (post-natal day 8-13) during prolonged and brief hypoxia. We used acute brain slices from Clomeleon mice which express a ratiometric fluorophore sensitive to Cl- and exposed them to oxygen-glucose deprivation (OGD) while imaging neuronal size and [Cl-]i by multiphoton microscopy. Neurons were identified using a convolutional neural network algorithm, and changes in the somatic area and [Cl-]i were evaluated using a linear mixed model for repeated measures. We found that (1) neuronal swelling and Cl- accumulation began after OGD, worsened during 20 min of OGD, or returned to baseline during reoxygenation if the exposure to OGD was brief (10 min). (2) Neuronal swelling did not occur when the extracellular Cl- concentration was low. (3) Enhancing KCC2 activity did not alter OGD-induced neuronal swelling but prevented Cl- accumulation; (4) blocking KCC2 led to an increase in Cl- accumulation during prolonged OGD and aggravated neuronal swelling during reoxygenation; (5) blocking NKCC1 reduced neuronal swelling during early but not prolonged OGD and aggravated Cl- accumulation during prolonged OGD; and (6) treatment with the "broad" CCC blocker furosemide reduced both swelling and Cl- accumulation during prolonged and brief OGD, whereas simultaneous NKCC1 and KCC2 inhibition using specific pharmacological blockers aggravated neuronal swelling during prolonged OGD. We conclude that CCCs, and other non-CCCs, contribute to water movement in neocortical neurons during OGD in the neonatal period.
Collapse
|
25
|
Uchida Y, Kan H, Sakurai K, Oishi K, Matsukawa N. Contributions of blood-brain barrier imaging to neurovascular unit pathophysiology of Alzheimer's disease and related dementias. Front Aging Neurosci 2023; 15:1111448. [PMID: 36861122 PMCID: PMC9969807 DOI: 10.3389/fnagi.2023.1111448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
The blood-brain barrier (BBB) plays important roles in the maintenance of brain homeostasis. Its main role includes three kinds of functions: (1) to protect the central nervous system from blood-borne toxins and pathogens; (2) to regulate the exchange of substances between the brain parenchyma and capillaries; and (3) to clear metabolic waste and other neurotoxic compounds from the central nervous system into meningeal lymphatics and systemic circulation. Physiologically, the BBB belongs to the glymphatic system and the intramural periarterial drainage pathway, both of which are involved in clearing interstitial solutes such as β-amyloid proteins. Thus, the BBB is believed to contribute to preventing the onset and progression for Alzheimer's disease. Measurements of BBB function are essential toward a better understanding of Alzheimer's pathophysiology to establish novel imaging biomarkers and open new avenues of interventions for Alzheimer's disease and related dementias. The visualization techniques for capillary, cerebrospinal, and interstitial fluid dynamics around the neurovascular unit in living human brains have been enthusiastically developed. The purpose of this review is to summarize recent BBB imaging developments using advanced magnetic resonance imaging technologies in relation to Alzheimer's disease and related dementias. First, we give an overview of the relationship between Alzheimer's pathophysiology and BBB dysfunction. Second, we provide a brief description about the principles of non-contrast agent-based and contrast agent-based BBB imaging methodologies. Third, we summarize previous studies that have reported the findings of each BBB imaging method in individuals with the Alzheimer's disease continuum. Fourth, we introduce a wide range of Alzheimer's pathophysiology in relation to BBB imaging technologies to advance our understanding of the fluid dynamics around the BBB in both clinical and preclinical settings. Finally, we discuss the challenges of BBB imaging techniques and suggest future directions toward clinically useful imaging biomarkers for Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Yuto Uchida
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Ōbu, Aichi, Japan
| | - Kenichi Oishi
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Noriyuki Matsukawa
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| |
Collapse
|
26
|
Yu J, Wang G, Chen Z, Wan L, Zhou J, Cai J, Liu X, Wang Y. Deficit of PKHD1L1 in the dentate gyrus increases seizure susceptibility in mice. Hum Mol Genet 2023; 32:506-519. [PMID: 36067019 DOI: 10.1093/hmg/ddac220] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 01/24/2023] Open
Abstract
Epilepsy is a chronic neurological disorder featuring recurrent, unprovoked seizures, which affect more than 65 million people worldwide. Here, we discover that the PKHD1L1, which is encoded by polycystic kidney and hepatic disease1-like 1 (Pkhd1l1), wildly distributes in neurons in the central nervous system (CNS) of mice. Disruption of PKHD1L1 in the dentate gyrus region of the hippocampus leads to increased susceptibility to pentylenetetrazol-induced seizures in mice. The disturbance of PKHD1L1 leads to the overactivation of the mitogen-activated protein kinase (MAPK)/extracellular regulated kinase (ERK)-Calpain pathway, which is accompanied by remarkable degradation of cytoplasmic potassium chloride co-transporter 2 (KCC2) level together with the impaired expression and function of membrane KCC2. However, the reduction of membrane KCC2 is associated with the damaged inhibitory ability of the vital GABA receptors, which ultimately leads to the significantly increased susceptibility to epileptic seizures. Our data, thus, indicate for the first time that Pkhd1l1, a newly discovered polycystic kidney disease (PKD) association gene, is required in neurons to maintain neuronal excitability by regulation of KCC2 expression in CNS. A new mechanism of the clinical association between genetic PKD and seizures has been built, which could be a potential therapeutic target for treating PKD-related seizures.
Collapse
Affiliation(s)
- Jiangning Yu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guoxiang Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhiyun Chen
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Wan
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Rehabilitation Center, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Rehabilitation Center, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Jingyi Cai
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xu Liu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
27
|
Tran TL, Hamann S, Heegaard S. Aquaporins in Eye. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:203-209. [PMID: 36717496 DOI: 10.1007/978-981-19-7415-1_14] [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: 02/01/2023]
Abstract
The major part of the eye consists of water. Continuous movement of water and ions between the ocular compartments and to the systemic circulation is pivotal for many physiological functions in the eye. The movement of water facilitates removal of the many metabolic products of corneal-, ciliary body-, lens-, and retinal metabolism, while maintaining transparency in the optical compartments. Transport across the corneal epithelium and endothelium maintains the corneal transparency. Also, aqueous humor is continuously secreted by the epithelia of the ciliary body and maintains the intraocular pressure. In the retina, water is transported into the vitreous body and across the retinal pigment epithelium to regulate the extracellular environment and the hydration of the retina. Aquaporins are a major contributor in the water transport throughout the eye.
Collapse
Affiliation(s)
- Thuy Linh Tran
- Department of Ophthalmology, Rigshospitalet - Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Hamann
- Department of Ophthalmology, Rigshospitalet - Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet - Glostrup, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Huang B, Wang H, Yang B. Non-Aquaporin Water Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:331-342. [PMID: 36717505 DOI: 10.1007/978-981-19-7415-1_23] [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: 02/01/2023]
Abstract
Water transport through membrane is so intricate that there are still some debates. AQPs are entirely accepted to allow water transmembrane movement depending on osmotic gradient. Cotransporters and uniporters, however, are also concerned in water homeostasis. UT-B has a single-channel water permeability that is similar to AQP1. CFTR was initially thought as a water channel but now not believed to transport water directly. By cotransporters, such as KCC4, NKCC1, SGLT1, GAT1, EAAT1, and MCT1, water is transported by water osmosis coupling with substrates, which explains how water is transported across the isolated small intestine. This chapter provides information about water transport mediated by other membrane proteins except AQPs.
Collapse
Affiliation(s)
- Boyue Huang
- Laboratory of Neuroscience and Tissue Engineering, Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Hongkai Wang
- Northwestern University Interdepartmental Neuroscience Program, Chicago, IL, USA
- Laboratory of Regenerative Rehabilitation and Department of Physical Medicine and Rehabilitation, Shirley Ryan AbilityLab and Northwestern University Feinberg School of Medicine and Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Baoxue Yang
- School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
29
|
Li Y, Sadiq A, Wang Z. Arterial Spin Labelling-Based Blood-Brain Barrier Assessment and Its Applications. INVESTIGATIVE MAGNETIC RESONANCE IMAGING 2022; 26:229-236. [PMID: 36687769 PMCID: PMC9851084 DOI: 10.13104/imri.2022.26.4.229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 01/12/2023]
Abstract
The brain relies on the blood-brain barrier (BBB) for the selective absorption of nutrients and the exclusion of other big molecules from the circulating blood. Therefore, the integrity of BBB is critical to brain health, and assessing BBB condition is of great clinical importance. BBB is often examined using exogenous tracers that can travel across the BBB, but the tracers might cause severe side effects. To avoid the use of external tracers, researchers have used magnetically labeled arterial blood as the endogenous tracer to assess the water permeability of BBB as a surrogate index of BBB. This paper reviews the three major types of Arterial Spin Labelling (ASL) based BBB water permeability assessment techniques and their applications in brain diseases such as Alzheimer's Disease.
Collapse
Affiliation(s)
- Yiran Li
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alishba Sadiq
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ze Wang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
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: 22] [Impact Index Per Article: 7.3] [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
|
31
|
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: 3.0] [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
|
32
|
Oernbo EK, Steffensen AB, Razzaghi Khamesi P, Toft-Bertelsen TL, Barbuskaite D, Vilhardt F, Gerkau NJ, Tritsaris K, Simonsen AH, Lolansen SD, Andreassen SN, Hasselbalch SG, Zeuthen T, Rose CR, Kurtcuoglu V, MacAulay N. Membrane transporters control cerebrospinal fluid formation independently of conventional osmosis to modulate intracranial pressure. Fluids Barriers CNS 2022; 19:65. [PMID: 36038945 PMCID: PMC9422132 DOI: 10.1186/s12987-022-00358-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022] Open
Abstract
Background Disturbances in the brain fluid balance can lead to life-threatening elevation in the intracranial pressure (ICP), which represents a vast clinical challenge. Nevertheless, the details underlying the molecular mechanisms governing cerebrospinal fluid (CSF) secretion are largely unresolved, thus preventing targeted and efficient pharmaceutical therapy of cerebral pathologies involving elevated ICP. Methods Experimental rats were employed for in vivo determinations of CSF secretion rates, ICP, blood pressure and ex vivo excised choroid plexus for morphological analysis and quantification of expression and activity of various transport proteins. CSF and blood extractions from rats, pigs, and humans were employed for osmolality determinations and a mathematical model employed to determine a contribution from potential local gradients at the surface of choroid plexus. Results We demonstrate that CSF secretion can occur independently of conventional osmosis and that local osmotic gradients do not suffice to support CSF secretion. Instead, the CSF secretion across the luminal membrane of choroid plexus relies approximately equally on the Na+/K+/2Cl− cotransporter NKCC1, the Na+/HCO3− cotransporter NBCe2, and the Na+/K+-ATPase, but not on the Na+/H+ exchanger NHE1. We demonstrate that pharmacological modulation of CSF secretion directly affects the ICP. Conclusions CSF secretion appears to not rely on conventional osmosis, but rather occur by a concerted effort of different choroidal transporters, possibly via a molecular mode of water transport inherent in the proteins themselves. Therapeutic modulation of the rate of CSF secretion may be employed as a strategy to modulate ICP. These insights identify new promising therapeutic targets against brain pathologies associated with elevated ICP. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00358-4.
Collapse
Affiliation(s)
- Eva K Oernbo
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Annette B Steffensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Pooya Razzaghi Khamesi
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Dagne Barbuskaite
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Niklas J Gerkau
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Katerina Tritsaris
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Anja H Simonsen
- Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sara D Lolansen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Søren N Andreassen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Steen G Hasselbalch
- Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Zeuthen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Vartan Kurtcuoglu
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
33
|
Targeting choroid plexus epithelium as a novel therapeutic strategy for hydrocephalus. J Neuroinflammation 2022; 19:156. [PMID: 35715859 PMCID: PMC9205094 DOI: 10.1186/s12974-022-02500-3] [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: 11/27/2021] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
The choroid plexus is a tissue located in the lateral ventricles of the brain and is composed mainly of choroid plexus epithelium cells. The main function is currently thought to be the secretion of cerebrospinal fluid and the regulation of its pH, and more functions are gradually being demonstrated. Assistance in the removal of metabolic waste and participation in the apoptotic pathway are also the functions of choroid plexus. Besides, it helps to repair the brain by regulating the secretion of neuropeptides and the delivery of drugs. It is involved in the immune response to assist in the clearance of infections in the central nervous system. It is now believed that the choroid plexus is in an inflammatory state after damage to the brain. This state, along with changes in the cilia, is thought to be an abnormal physiological state of the choroid plexus, which in turn leads to abnormal conditions in cerebrospinal fluid and triggers hydrocephalus. This review describes the pathophysiological mechanism of hydrocephalus following choroid plexus epithelium cell abnormalities based on the normal physiological functions of choroid plexus epithelium cells, and analyzes the attempts and future developments of using choroid plexus epithelium cells as a therapeutic target for hydrocephalus.
Collapse
|
34
|
Abstract
The brain harbors a unique ability to, figuratively speaking, shift its gears. During wakefulness, the brain is geared fully toward processing information and behaving, while homeostatic functions predominate during sleep. The blood-brain barrier establishes a stable environment that is optimal for neuronal function, yet the barrier imposes a physiological problem; transcapillary filtration that forms extracellular fluid in other organs is reduced to a minimum in brain. Consequently, the brain depends on a special fluid [the cerebrospinal fluid (CSF)] that is flushed into brain along the unique perivascular spaces created by astrocytic vascular endfeet. We describe this pathway, coined the term glymphatic system, based on its dependency on astrocytic vascular endfeet and their adluminal expression of aquaporin-4 water channels facing toward CSF-filled perivascular spaces. Glymphatic clearance of potentially harmful metabolic or protein waste products, such as amyloid-β, is primarily active during sleep, when its physiological drivers, the cardiac cycle, respiration, and slow vasomotion, together efficiently propel CSF inflow along periarterial spaces. The brain's extracellular space contains an abundance of proteoglycans and hyaluronan, which provide a low-resistance hydraulic conduit that rapidly can expand and shrink during the sleep-wake cycle. We describe this unique fluid system of the brain, which meets the brain's requisites to maintain homeostasis similar to peripheral organs, considering the blood-brain-barrier and the paths for formation and egress of the CSF.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
35
|
MacAulay N, Keep RF, Zeuthen T. Cerebrospinal fluid production by the choroid plexus: a century of barrier research revisited. Fluids Barriers CNS 2022; 19:26. [PMID: 35317823 PMCID: PMC8941821 DOI: 10.1186/s12987-022-00323-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Cerebrospinal fluid (CSF) envelops the brain and fills the central ventricles. This fluid is continuously replenished by net fluid extraction from the vasculature by the secretory action of the choroid plexus epithelium residing in each of the four ventricles. We have known about these processes for more than a century, and yet the molecular mechanisms supporting this fluid secretion remain unresolved. The choroid plexus epithelium secretes its fluid in the absence of a trans-epithelial osmotic gradient, and, in addition, has an inherent ability to secrete CSF against an osmotic gradient. This paradoxical feature is shared with other 'leaky' epithelia. The assumptions underlying the classical standing gradient hypothesis await experimental support and appear to not suffice as an explanation of CSF secretion. Here, we suggest that the elusive local hyperosmotic compartment resides within the membrane transport proteins themselves. In this manner, the battery of plasma membrane transporters expressed in choroid plexus are proposed to sustain the choroidal CSF secretion independently of the prevailing bulk osmotic gradient.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Thomas Zeuthen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| |
Collapse
|
36
|
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: 0.3] [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
|
37
|
Okamoto H, Kitamura S, Masaki N. Activation of the root xylem proton pump by hydraulic signals from leaves under suppressed transpiration. JOURNAL OF PLANT RESEARCH 2022; 135:311-322. [PMID: 35075591 DOI: 10.1007/s10265-022-01368-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
Long term field observations have revealed that the inhibition of transpiration by heavy rainfall promotes immediate positive shift in the trans-root electric potential (TRP), indicating activation of the xylem proton pump in the tree root system presumably participating in acropetal water transport. This phenomenon is indicative of signal transmission from the aerial part to the root system via change in the xylem hydraulic pressure. To test this hypothesis, we constructed a new device that enables the simultaneous recording of artificially applied xylem hydraulic pressure and the change in the TRP of tree saplings. With the application of artificial pressure to the xylem vessels (20-62 kPa), TRP shifted towards positive potential by 20-80 mV, which indicates the activation of the proton pump in the root xylem. The reaction was observed in 11 tree species, six deciduous and five evergreen, although only during the resting phase of the xylem proton pump (May to October) when the transpiration rates were high. Contrastingly the application of tension (negative pressure) produced no reaction. Simultaneous determination of the two components of the TRP, i.e. Vps (electric membrane potential difference across root surface cell membrane) and Vpx (electric membrane potential difference between root symplast and xylem vessel), are performed using the intra-cellular micro-electrode technique throughout the four seasons. Application of excess xylem hydraulic pressure had no significant effect on Vps, while it brought about hyper-polarisation of Vpx except during the winter season, most significantly during summer when transpiration is vigorous and the xylem pump is in a resting state. Such effect of excess xylem pressure was, however, not observed under anoxia.
Collapse
Affiliation(s)
- Hisashi Okamoto
- Mori Laboratory of Plant Physiology, 443-5 Enden Mori, Shizuoka, 437-0221, Japan.
| | - Sayaka Kitamura
- Mori Laboratory of Plant Physiology, 443-5 Enden Mori, Shizuoka, 437-0221, Japan
| | - Nobuyuki Masaki
- Masaki Laboratory of Arboriculture, 729-2 Higashihara Iwata, Shizuoka, 438-0802, Japan
| |
Collapse
|
38
|
Funai Y, Takemura M, Inoue K, Shirasaka Y. Effect of Ingested Fluid Volume and Solution Osmolality on Intestinal Drug Absorption: Impact on Drug Interaction with Beverages. Eur J Pharm Sci 2022; 172:106136. [PMID: 35121020 DOI: 10.1016/j.ejps.2022.106136] [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: 06/07/2021] [Revised: 01/17/2022] [Accepted: 01/31/2022] [Indexed: 11/03/2022]
Abstract
It was recently shown that osmolality-dependent fluid movement is a significant factor causing the clinically observed apple juice (AJ)-atenolol interaction. Here we examined whether osmolality-dependent fluid movement may also explain the AJ volume dependence of the AJ-atenolol interaction. In Wistar rats, the luminal fluid volume after administration of different volumes of purified water (0.5 and 1.0 mL) gradually reduced to a similar steady-state level, while that after administration of different volumes of AJ (0.5 and 1.0 mL) increased and attained different apparent steady-state levels. It was hypothesized that osmolality-dependent fluid secretion would account for the volume dependence of the apparent steady-state. Indeed, the luminal concentration of FD-4, a non-permeable compound, after administration in AJ was attenuated depending upon the ingested volume, whereas that after administration in purified water was independent of the ingested fluid volume. An in vivo pharmacokinetic study in rats showed that co-administration of AJ and hyperosmotic solution (adjusted to the osmolality of AJ) with atenolol volume-dependently reduced the AUC and Cmax of atenolol significantly. These results show that osmolality-dependent variations in luminal fluid volume may indirectly influence the absorption characteristics of drugs, and can account for the observed volume dependence of beverage-drug interactions.
Collapse
Affiliation(s)
- Yuta Funai
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.; School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| | - Miyuki Takemura
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.; School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Katsuhisa Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.; School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| |
Collapse
|
39
|
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
|
40
|
Boursiac Y, Protto V, Rishmawi L, Maurel C. Experimental and conceptual approaches to root water transport. PLANT AND SOIL 2022; 478:349-370. [PMID: 36277078 PMCID: PMC9579117 DOI: 10.1007/s11104-022-05427-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/03/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Root water transport, which critically contributes to the plant water status and thereby plant productivity, has been the object of extensive experimental and theoretical studies. However, root systems represent an intricate assembly of cells in complex architectures, including many tissues at distinct developmental stages. Our comprehension of where and how molecular actors integrate their function in order to provide the root with its hydraulic properties is therefore still limited. SCOPE Based on current literature and prospective discussions, this review addresses how root water transport can be experimentally measured, what is known about the underlying molecular actors, and how elementary water transport processes are scaled up in numerical/mathematical models. CONCLUSIONS The theoretical framework and experimental procedures on root water transport that are in use today have been established a few decades ago. However, recent years have seen the appearance of new techniques and models with enhanced resolution, down to a portion of root or to the tissue level. These advances pave the way for a better comprehension of the dynamics of water uptake by roots in the soil.
Collapse
Affiliation(s)
- Yann Boursiac
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut Agro, 34060 Montpellier, France
| | - Virginia Protto
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut Agro, 34060 Montpellier, France
| | - Louai Rishmawi
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut Agro, 34060 Montpellier, France
| | - Christophe Maurel
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut Agro, 34060 Montpellier, France
| |
Collapse
|
41
|
Studying cell volume beyond cell volume. CURRENT TOPICS IN MEMBRANES 2021; 88:165-188. [PMID: 34862025 DOI: 10.1016/bs.ctm.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The first part of the paper describes two simple microscopic techniques that we use in our laboratory. One measures cell volumes in adherent cultures and the other measures cell dry mass; both measurements are done on the same instrument (a standard bright-field transmission microscope with only one or two narrow-band color filters added) and on the same cells. The reason for combining cell volume with dry mass is that the ratio of the two-dry mass concentration (MC)-is an important and insufficiently utilized biological parameter. We then describe a few applications of MC. The available experimental data strongly suggest its critical role in biological processes, including cell volume regulation. For example, most eukaryotic cells have surprisingly similar values of MC. Moreover, MC (and not cell volume) is tightly controlled in growing cell cultures at highly variable external osmolarities. We review the results showing that elevation of MC is a direct cause of shrinkage-induced apoptosis. Also, by focusing on MC, one can study heterogenous processes, such as necrotic swelling, or discriminate between apoptotic dehydration and the loss of cell fragments.
Collapse
|
42
|
Barany A, Shaughnessy CA, Pelis RM, Fuentes J, Mancera JM, McCormick SD. Tissue and salinity specific Na +/Cl - cotransporter (NCC) orthologues involved in the adaptive osmoregulation of sea lamprey (Petromyzon marinus). Sci Rep 2021; 11:22698. [PMID: 34811419 PMCID: PMC8608846 DOI: 10.1038/s41598-021-02125-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Two orthologues of the gene encoding the Na+-Cl− cotransporter (NCC), termed ncca and nccb, were found in the sea lamprey genome. No gene encoding the Na+-K+-2Cl− cotransporter 2 (nkcc2) was identified. In a phylogenetic comparison among other vertebrate NCC and NKCC sequences, the sea lamprey NCCs occupied basal positions within the NCC clades. In freshwater, ncca mRNA was found only in the gill and nccb only in the intestine, whereas both were found in the kidney. Intestinal nccb mRNA levels increased during late metamorphosis coincident with salinity tolerance. Acclimation to seawater increased nccb mRNA levels in the intestine and kidney. Electrophysiological analysis of intestinal tissue ex vivo showed this tissue was anion absorptive. After seawater acclimation, the proximal intestine became less anion absorptive, whereas the distal intestine remained unchanged. Luminal application of indapamide (an NCC inhibitor) resulted in 73% and 30% inhibition of short-circuit current (Isc) in the proximal and distal intestine, respectively. Luminal application of bumetanide (an NKCC inhibitor) did not affect intestinal Isc. Indapamide also inhibited intestinal water absorption. Our results indicate that NCCb is likely the key ion cotransport protein for ion uptake by the lamprey intestine that facilitates water absorption in seawater. As such, the preparatory increases in intestinal nccb mRNA levels during metamorphosis of sea lamprey are likely critical to development of whole animal salinity tolerance.
Collapse
Affiliation(s)
- A Barany
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, Puerto Real, 11519, Cádiz, Spain. .,Centre of Marine Sciences (CCMar), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal. .,Conte Anadromous Fish Research Laboratory, Eastern Ecological Science Center, U.S. Geological Survey, Turners Falls, MA, 01376, USA.
| | - C A Shaughnessy
- Conte Anadromous Fish Research Laboratory, Eastern Ecological Science Center, U.S. Geological Survey, Turners Falls, MA, 01376, USA.,Graduate Program in Organismic and Evolutionary Biology, University of Massachusetts, Amherst, MA, 01003, USA
| | - R M Pelis
- Department of Pharmaceutical Sciences, Binghamton University, State University of New York, Johnson City, NY, 13790, USA
| | - J Fuentes
- Centre of Marine Sciences (CCMar), University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal
| | - J M Mancera
- Department of Biology, Faculty of Marine and Environmental Sciences, Campus de Excelencia Internacional del Mar (CEI-MAR), University of Cádiz, Puerto Real, 11519, Cádiz, Spain
| | - S D McCormick
- Conte Anadromous Fish Research Laboratory, Eastern Ecological Science Center, U.S. Geological Survey, Turners Falls, MA, 01376, USA.,Graduate Program in Organismic and Evolutionary Biology, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
43
|
Lee S, Kang HG, Ryou C, Cheon YP. Spatiotemporal expression of aquaporin 9 is critical for the antral growth of mouse ovarian follicles†. Biol Reprod 2021; 103:828-839. [PMID: 32577722 DOI: 10.1093/biolre/ioaa108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 09/21/2019] [Accepted: 06/18/2020] [Indexed: 12/21/2022] Open
Abstract
Although a few aquaporins (AQPs) expressed in granulosa cells have been postulated to mediate fluid passage into the antrum, the specific expression of AQPs in different follicle cell types and stages and their roles have not been evaluated extensively. The spatiotemporal expression of aquaporin (Aqp) 7, 8, and 9 and the functional roles of Aqp9 in antral growth and ovulation were examined using a superovulation model and 3-dimensional follicle culture. Aqp9 was expressed at a high level in the rapid growth phase (24-48 h post equine chorionic gonadotropin (eCG) for superovulation induction) compared to Aqp7 (after human chorionic gonadotropin (hCG)) and Aqp8 (8-24 h post eCG and 24 h post hCG). A dramatic increase in the expression and localization of Aqp9 mRNA in theca cells was observed, as evaluated using quantitative reverse transcription-polymerase (RT-PCR) coupled with laser capture microdissection and immunohistochemistry. AQP9 was located primarily on the theca cells of the tertiary and preovulatory follicles but not on the ovulated follicles. In phloretin-treated mice, the diameter of the preovulatory follicles and the number of ovulated oocytes decreased. Consistent with these findings, knocking down Aqp9 expression with an Aqp9 siRNA inhibited follicle growth (0.28:1 = siRNA:control) and decreased the number of ovulated follicles (0.36:1 = siRNA:control) during in vitro growth and ovulation induction. Based on these results, the expression of AQPs is under the control of the physiological status, and AQP9 expression in theca during folliculogenesis is required for antral growth and ovulation in a tissue-specific and stage-dependent manner.
Collapse
Affiliation(s)
- Sungeun Lee
- Department of Biotechnology, Sungshin University, Seoul, Korea
| | - Hee-Gyoo Kang
- Department of Biomedical Engineering and Institute of Pharmaceutical Science and Technology, Eulji University, Seongnam-Si, Gyeonggi-Do, Korea
| | - Chongsuk Ryou
- Department of Pharmacy, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do, Korea
| | - Yong-Pil Cheon
- Department of Biotechnology, Sungshin University, Seoul, Korea
| |
Collapse
|
44
|
Chew TA, Zhang J, Feng L. High-Resolution Views and Transport Mechanisms of the NKCC1 and KCC Transporters. J Mol Biol 2021; 433:167056. [PMID: 34022207 PMCID: PMC9722358 DOI: 10.1016/j.jmb.2021.167056] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/08/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022]
Abstract
Cation-chloride cotransporters (CCCs) are responsible for the coupled co-transport of Cl- with K+ and/or Na+ in an electroneutral manner. They play important roles in myriad fundamental physiological processes--from cell volume regulation to transepithelial solute transport and intracellular ion homeostasis--and are targeted by medicines commonly prescribed to treat hypertension and edema. After several decades of studies into the functions and pharmacology of these transporters, there have been several breakthroughs in the structural determination of CCC transporters. The insights provided by these new structures for the Na+/K+/Cl- cotransporter NKCC1 and the K+/Cl- cotransporters KCC1, KCC2, KCC3 and KCC4 have deepened our understanding of their molecular basis and transport function. This focused review discusses recent advances in the structural and mechanistic understanding of CCC transporters, including architecture, dimerization, functional roles of regulatory domains, ion binding sites, and coupled ion transport.
Collapse
Affiliation(s)
- Thomas A Chew
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jinru Zhang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liang Feng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
45
|
Tagge IJ, Anderson VC, Springer CS, Sammi MK, Bourdette DN, Spain RI, Rooney WD. Gray matter blood-brain barrier water exchange dynamics are reduced in progressive multiple sclerosis. J Neuroimaging 2021; 31:1111-1118. [PMID: 34355458 PMCID: PMC9291753 DOI: 10.1111/jon.12912] [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: 05/26/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose To compare transcapillary wall water exchange, a putative marker of cerebral metabolic health, in brain T2 white matter (WM) lesions and normal appearing white and gray matter (NAWM and NAGM, respectively) in individuals with progressive multiple sclerosis (PMS) and healthy controls (HC). Methods Dynamic‐contrast‐enhanced 7T MRI data were obtained from 19 HC and 23 PMS participants. High‐resolution pharmacokinetic parametric maps representing tissue microvascular and microstructural properties were created by shutter‐speed (SS) paradigm modeling to obtain estimates of blood volume fraction (vb), water molecule capillary efflux rate constant (kpo), and the water capillary wall permeability surface area product (PwS ≡ vb*kpo). Linear regression models were used to investigate differences in (i) kpo and PwS between groups in NAWM and NAGM, and (ii) between WM lesions and NAWM in PMS. Results High‐resolution parametric maps were produced to visualize tissue classes and resolve individual WM lesions. Normal‐appearing gray matter kpo and PwS were significantly decreased in PMS compared to HC (p ≤ .01). Twenty‐one T2 WM lesions were analyzed in 10 participants with PMS. kpo was significantly decreased in WM lesions compared to PMS NAWM (p < .0001). Conclusions Transcapillary water exchange is reduced in PMS NAGM compared to HC and is further reduced in PMS WM lesions, suggesting pathologically impaired brain metabolism. kpo provides a sensitive measure of cerebral metabolic activity and/or coupling, and can be mapped at higher spatial resolution than conventional imaging techniques assessing metabolic activity.
Collapse
Affiliation(s)
- Ian J Tagge
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Valerie C Anderson
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Charles S Springer
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Manoj K Sammi
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Dennis N Bourdette
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Rebecca I Spain
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA.,Neurology Division, Veterans Affairs Portland Health Care System, Portland, Oregon, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
46
|
Ritter M, Bresgen N, Kerschbaum HH. From Pinocytosis to Methuosis-Fluid Consumption as a Risk Factor for Cell Death. Front Cell Dev Biol 2021; 9:651982. [PMID: 34249909 PMCID: PMC8261248 DOI: 10.3389/fcell.2021.651982] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
The volumes of a cell [cell volume (CV)] and its organelles are adjusted by osmoregulatory processes. During pinocytosis, extracellular fluid volume equivalent to its CV is incorporated within an hour and membrane area equivalent to the cell's surface within 30 min. Since neither fluid uptake nor membrane consumption leads to swelling or shrinkage, cells must be equipped with potent volume regulatory mechanisms. Normally, cells respond to outwardly or inwardly directed osmotic gradients by a volume decrease and increase, respectively, i.e., they shrink or swell but then try to recover their CV. However, when a cell death (CD) pathway is triggered, CV persistently decreases in isotonic conditions in apoptosis and it increases in necrosis. One type of CD associated with cell swelling is due to a dysfunctional pinocytosis. Methuosis, a non-apoptotic CD phenotype, occurs when cells accumulate too much fluid by macropinocytosis. In contrast to functional pinocytosis, in methuosis, macropinosomes neither recycle nor fuse with lysosomes but with each other to form giant vacuoles, which finally cause rupture of the plasma membrane (PM). Understanding methuosis longs for the understanding of the ionic mechanisms of cell volume regulation (CVR) and vesicular volume regulation (VVR). In nascent macropinosomes, ion channels and transporters are derived from the PM. Along trafficking from the PM to the perinuclear area, the equipment of channels and transporters of the vesicle membrane changes by retrieval, addition, and recycling from and back to the PM, causing profound changes in vesicular ion concentrations, acidification, and-most importantly-shrinkage of the macropinosome, which is indispensable for its proper targeting and cargo processing. In this review, we discuss ion and water transport mechanisms with respect to CVR and VVR and with special emphasis on pinocytosis and methuosis. We describe various aspects of the complex mutual interplay between extracellular and intracellular ions and ion gradients, the PM and vesicular membrane, phosphoinositides, monomeric G proteins and their targets, as well as the submembranous cytoskeleton. Our aim is to highlight important cellular mechanisms, components, and processes that may lead to methuotic CD upon their derangement.
Collapse
Affiliation(s)
- Markus Ritter
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
- Institute for Physiology and Pathophysiology, Paracelsus Medical University, Nuremberg, Germany
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis und Rehabilitation, Salzburg, Austria
- Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | - Nikolaus Bresgen
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | | |
Collapse
|
47
|
Kalia M, Meijer HGE, van Gils SA, van Putten MJAM, Rose CR. Ion dynamics at the energy-deprived tripartite synapse. PLoS Comput Biol 2021; 17:e1009019. [PMID: 34143772 PMCID: PMC8244923 DOI: 10.1371/journal.pcbi.1009019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 06/30/2021] [Accepted: 04/28/2021] [Indexed: 01/09/2023] Open
Abstract
The anatomical and functional organization of neurons and astrocytes at 'tripartite synapses' is essential for reliable neurotransmission, which critically depends on ATP. In low energy conditions, synaptic transmission fails, accompanied by a breakdown of ion gradients, changes in membrane potentials and cell swelling. The resulting cellular damage and cell death are causal to the often devastating consequences of an ischemic stroke. The severity of ischemic damage depends on the age and the brain region in which a stroke occurs, but the reasons for this differential vulnerability are far from understood. In the present study, we address this question by developing a comprehensive biophysical model of a glutamatergic synapse to identify key determinants of synaptic failure during energy deprivation. Our model is based on fundamental biophysical principles, includes dynamics of the most relevant ions, i.e., Na+, K+, Ca2+, Cl- and glutamate, and is calibrated with experimental data. It confirms the critical role of the Na+/K+-ATPase in maintaining ion gradients, membrane potentials and cell volumes. Our simulations demonstrate that the system exhibits two stable states, one physiological and one pathological. During energy deprivation, the physiological state may disappear, forcing a transit to the pathological state, which can be reverted when blocking voltage-gated Na+ and K+ channels. Our model predicts that the transition to the pathological state is favoured if the extracellular space fraction is small. A reduction in the extracellular space volume fraction, as, e.g. observed with ageing, will thus promote the brain's susceptibility to ischemic damage. Our work provides new insights into the brain's ability to recover from energy deprivation, with translational relevance for diagnosis and treatment of ischemic strokes.
Collapse
Affiliation(s)
- Manu Kalia
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, The Netherlands
- * E-mail:
| | - Hil G. E. Meijer
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, The Netherlands
| | - Stephan A. van Gils
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, The Netherlands
| | | | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
48
|
Abstract
Our brains consist of 80% water, which is continuously shifted between different compartments and cell types during physiological and pathophysiological processes. Disturbances in brain water homeostasis occur with pathologies such as brain oedema and hydrocephalus, in which fluid accumulation leads to elevated intracranial pressure. Targeted pharmacological treatments do not exist for these conditions owing to our incomplete understanding of the molecular mechanisms governing brain water transport. Historically, the transmembrane movement of brain water was assumed to occur as passive movement of water along the osmotic gradient, greatly accelerated by water channels termed aquaporins. Although aquaporins govern the majority of fluid handling in the kidney, they do not suffice to explain the overall brain water movement: either they are not present in the membranes across which water flows or they appear not to be required for the observed flow of water. Notably, brain fluid can be secreted against an osmotic gradient, suggesting that conventional osmotic water flow may not describe all transmembrane fluid transport in the brain. The cotransport of water is an unconventional molecular mechanism that is introduced in this Review as a missing link to bridge the gap in our understanding of cellular and barrier brain water transport.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
49
|
van Putten MJ, Fahlke C, Kafitz KW, Hofmeijer J, Rose CR. Dysregulation of Astrocyte Ion Homeostasis and Its Relevance for Stroke-Induced Brain Damage. Int J Mol Sci 2021; 22:5679. [PMID: 34073593 PMCID: PMC8198632 DOI: 10.3390/ijms22115679] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of mortality and chronic disability. Either recovery or progression towards irreversible failure of neurons and astrocytes occurs within minutes to days, depending on remaining perfusion levels. Initial damage arises from energy depletion resulting in a failure to maintain homeostasis and ion gradients between extra- and intracellular spaces. Astrocytes play a key role in these processes and are thus central players in the dynamics towards recovery or progression of stroke-induced brain damage. Here, we present a synopsis of the pivotal functions of astrocytes at the tripartite synapse, which form the basis of physiological brain functioning. We summarize the evidence of astrocytic failure and its consequences under ischemic conditions. Special emphasis is put on the homeostasis and stroke-induced dysregulation of the major monovalent ions, namely Na+, K+, H+, and Cl-, and their involvement in maintenance of cellular volume and generation of cerebral edema.
Collapse
Affiliation(s)
- Michel J.A.M. van Putten
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christoph Fahlke
- Institut für Biologische Informationsprozesse, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, 52425 Jülich, Germany;
| | - Karl W. Kafitz
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Jeannette Hofmeijer
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
50
|
Vitali V, Sutka M, Ojeda L, Aroca R, Amodeo G. Root hydraulics adjustment is governed by a dominant cell-to-cell pathway in Beta vulgaris seedlings exposed to salt stress. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2021; 306:110873. [PMID: 33775369 DOI: 10.1016/j.plantsci.2021.110873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 06/12/2023]
Abstract
Soil salinity reduces root hydraulic conductivity (Lpr) of several plant species. However, how cellular signaling and root hydraulic properties are linked in plants that can cope with water restriction remains unclear. In this work, we exposed the halotolerant species red beet (Beta vulgaris) to increasing concentrations of NaCl to determine the components that might be critical to sustaining the capacity to adjust root hydraulics. Our strategy was to use both hydraulic and cellular approaches in hydroponically grown seedlings during the first osmotic phase of salt stress. Interestingly, Lpr presented a bimodal profile response apart from the magnitude of the imposed salt stress. As well as Lpr, the PIP2-aquaporin profile follows an unphosphorylated/phosphorylated pattern when increasing NaCl concentration while PIP1 aquaporins remain constant. Lpr also shows high sensitivity to cycloheximide. In low NaCl concentrations, Lpr was high and 70 % of its capacity could be attributed to the CHX-inhibited cell-to-cell pathway. More interestingly, roots can maintain a constant spontaneous exudated flow that is independent of the applied NaCl concentration. In conclusion, Beta vulgaris root hydraulic adjustment completely lies in a dominant cell-to-cell pathway that contributes to satisfying plant water demands.
Collapse
Affiliation(s)
- Victoria Vitali
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales & Instituto de Biodiversidad, Biología Experimental y Aplicada, Universidad de Buenos Aires and Consejo Nacional de Investigaciones Científicas y Técnicas, C1428EGA, Buenos Aires, Argentina
| | - Moira Sutka
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales & Instituto de Biodiversidad, Biología Experimental y Aplicada, Universidad de Buenos Aires and Consejo Nacional de Investigaciones Científicas y Técnicas, C1428EGA, Buenos Aires, Argentina
| | - Lucas Ojeda
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales & Instituto de Biodiversidad, Biología Experimental y Aplicada, Universidad de Buenos Aires and Consejo Nacional de Investigaciones Científicas y Técnicas, C1428EGA, Buenos Aires, Argentina
| | - Ricardo Aroca
- Departamento de Microbiología del Suelo y Sistemas Simbióticos, Estación Experimental del Zaidín (EEZ-CSIC), Profesor Albareda 1, 18008, Granada, Spain
| | - Gabriela Amodeo
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales & Instituto de Biodiversidad, Biología Experimental y Aplicada, Universidad de Buenos Aires and Consejo Nacional de Investigaciones Científicas y Técnicas, C1428EGA, Buenos Aires, Argentina.
| |
Collapse
|