Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Neurobiol Dis ; 149: 105226, 2021 02.
Article in English | MEDLINE | ID: mdl-33347975

ABSTRACT

Abnormal excitability in cortical networks has been reported in patients and animal models of Alzheimer's disease (AD), and other neurodegenerative conditions. Whether hyperexcitability is a core feature of alpha(α)-synucleinopathies, including dementia with Lewy bodies (DLB) is unclear. To assess this, we used two murine models of DLB that express either human mutant α-synuclein (α-syn) the hA30P, or human wild-type α-syn (hWT-α-syn) mice. We observed network hyperexcitability in vitro in young (2-5 months), pre-symptomatic transgenic α-syn mice. Interictal discharges (IIDs) were seen in the extracellular local field potential (LFP) in the hippocampus in hA30P and hWT-α-syn mice following kainate application, while only gamma frequency oscillations occurred in control mice. In addition, the concentration of the GABAA receptor antagonist (gabazine) needed to evoke IIDs was lower in slices from hA30P mice compared to control mice. hA30P mice also showed increased locomotor activity in the open field test compared to control mice. Intracellular recordings from CA3 pyramidal cells showed a more depolarised resting membrane potential in hA30P mice. Quadruple immunohistochemistry for human α-syn, and the mitochondrial markers, porin and the complex IV enzyme cytochrome c oxidase subunit 1 (COX1) in parvalbumin (PV+)-expressing interneurons showed that 25% of PV+ cells contained human α-syn in hA30P mice. While there was no change in PV expression, COX1 expression was significantly increased in PV+ cells in hA30P mice, perhaps reflecting a compensatory change to support PV+ interneuron activity. Our findings suggest that hippocampal network hyperexcitability may be an important early consequence of α-syn-mediated impairment of neuronal/synaptic function, which occurs without any overt loss of PV interneurons. The therapeutic benefit of targeting network excitability early in the disease stage should be explored with respect to α-synucleinopathies such as DLB.


Subject(s)
Gamma Rhythm/physiology , Hippocampus/metabolism , Mutation/physiology , Nerve Net/metabolism , alpha-Synuclein/biosynthesis , Age Factors , Animals , Dose-Response Relationship, Drug , Female , Gamma Rhythm/drug effects , Gene Expression , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/drug effects , Nerve Net/physiopathology , Organ Culture Techniques , alpha-Synuclein/genetics
2.
Am J Physiol Renal Physiol ; 318(5): F1138-F1146, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32174144

ABSTRACT

The proximal renal tubule (PT) is characterized by a highly conductive paracellular pathway, which contributes to a significant amount of solute and water reabsorption by the kidney. Claudins are tight junction proteins that, in part, determine the paracellular permeability of epithelia. In the present study, we determined the expression pattern of the major PT claudins. We found that claudin-2 and claudin-10 are coexpressed throughout the PT, whereas claudin-3 is coexpressed with claudin-2 predominantly in the proximal straight tubule. Additionally, claudin-2 and claudin-3 are expressed separately within mutually exclusive populations of descending thin limbs. We developed a novel double-inducible Madin-Darby canine kidney I cell model to characterize in vitro the functional effect of coexpression of PT claudins. In keeping with previous studies, we found that claudin-2 alone primarily increased cation (Na+ and Ca2+) permeability, whereas claudin-10a alone increased anion (Cl-) permeability. Coexpression of claudin-2 and claudin-10a together led to a weak physical interaction between the isoforms and the formation of a monolayer with high conductance but neutral charge selectivity. Claudin-3 expression had a negligible effect on all measures of cell permeability, whether expressed alone or together with claudin-2. In cells coexpressing a claudin-2 mutant, S68C, together with claudin-10a, inhibition of cation permeability through the claudin-2 pore with a thiol-reactive pore blocker did not block anion permeation through claudin-10a. We conclude that claudin-2 and claudin-10a form independent paracellular cation- and anion-selective channels that function in parallel.


Subject(s)
Claudins/metabolism , Kidney Tubules, Proximal/metabolism , Tight Junctions/metabolism , Animals , Claudins/genetics , Dogs , Electric Conductivity , Ion Transport , Kidney Tubules, Proximal/cytology , Madin Darby Canine Kidney Cells , Membrane Potentials , Mice , Permeability , Signal Transduction
3.
Am J Physiol Renal Physiol ; 316(5): F966-F969, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30838875

ABSTRACT

The proximal tubule (PT) is responsible for the majority of calcium reabsorption by the kidney. Most PT calcium transport appears to be passive, although the molecular facilitators have not been well established. Emerging evidence supports a major role for PT calcium transport in idiopathic hypercalciuria and the development of kidney stones. This review will cover recent developments in our understanding of PT calcium transport and the role of the PT in kidney stone formation.


Subject(s)
Calcium/metabolism , Hypercalciuria/complications , Kidney Calculi/etiology , Kidney Tubules, Proximal/metabolism , Membrane Transport Proteins/metabolism , Nephrocalcinosis/etiology , Renal Reabsorption , Animals , Claudins/metabolism , Humans , Hypercalciuria/metabolism , Hypercalciuria/physiopathology , Ion Transport , Kidney Calculi/metabolism , Kidney Calculi/physiopathology , Kidney Tubules, Proximal/physiopathology , Nephrocalcinosis/metabolism , Nephrocalcinosis/physiopathology
4.
Sensors (Basel) ; 19(8)2019 Apr 25.
Article in English | MEDLINE | ID: mdl-31027204

ABSTRACT

The Internet of Things (IoT) is a constantly-evolving area of research and touches almost every aspect of life in the modern world. As technology moves forward, it is becoming increasingly important for these IoT devices for environmental sensing to become self-powered to enable long-term operation. This paper provides an outlook on the current state-of-the-art in terms of energy harvesting for these low-power devices. An analytical approach is taken, first defining types of environments in which energy-harvesters operate, before exploring both well-known and novel energy harvesting techniques and their uses in modern-day sensing.

6.
J Am Soc Nephrol ; 28(12): 3504-3517, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28774999

ABSTRACT

Compared with males, females have lower BP before age 60, blunted hypertensive response to angiotensin II, and a leftward shift in pressure natriuresis. This study tested the concept that this female advantage associates with a distinct sexual dimorphic pattern of transporters along the nephron. We applied quantitative immunoblotting to generate profiles of transporters, channels, claudins, and selected regulators in both sexes and assessed the physiologic consequences of the differences. In rats, females excreted a saline load more rapidly than males did. Compared with the proximal tubule of males, the proximal tubule of females had greater phosphorylation of Na+/H+ exchanger isoform 3 (NHE3), distribution of NHE3 at the base of the microvilli, and less abundant expression of Na+/Pi cotransporter 2, claudin-2, and aquaporin 1. These changes associated with less bicarbonate reabsorption and higher lithium clearance in females. The distal nephrons of females had a higher abundance of total and phosphorylated Na+/Cl- cotransporter (NCC), claudin-7, and cleaved forms of epithelial Na+ channel (ENaC) α and γ subunits, which associated with a lower baseline plasma K+ concentration. A K+-rich meal increased the urinary K+ concentration and decreased the level of renal phosphorylated NCC in females. Notably, we observed similar abundance profiles in female versus male C57BL/6 mice. These results define sexual dimorphic phenotypes along the nephron and suggest that lower proximal reabsorption in female rats expedites excretion of a saline load and enhances NCC and ENaC abundance and activation, which may facilitate K+ secretion and set plasma K+ at a lower level.


Subject(s)
Electrolytes/metabolism , Kidney Tubules/metabolism , Kidney/metabolism , Membrane Transport Proteins/metabolism , Sex Characteristics , Animals , Biological Transport , Blood Pressure , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Microvilli/metabolism , Nephrons/metabolism , Phosphorylation , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Sodium/metabolism
7.
Am J Physiol Renal Physiol ; 305(12): F1687-96, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24133122

ABSTRACT

The cation cotransporters Na(+)-K(+)-2Cl(-) cotransporter 1 and 2 (NKCC1 and NKCC2) and Na(+)-Cl cotransporter (NCC) are phosphorylated and activated by the kinases Ste20-related proline alanine-rich kinase (SPAK) and oxidative stress-responsive kinase (OSR1), and their targeted disruption in mice causes phenotypes resembling the human disorders Bartter syndrome and Gitelman syndrome, reflecting reduced NKCC2 and NCC activity, respectively. We previously cloned a kinase-inactive kidney-specific SPAK isoform, kidney-specific (KS)-SPAK, which lacks the majority of the kinase domain present in full-length SPAK. Another putative inactive SPAK isoform, SPAK2, which only lacks the initial portion of the kinase domain, is also highly expressed in kidney. The functional relevance of inactive SPAK isoforms is unclear. Here, we tested whether KS-SPAK and SPAK2 differentially affect cation cotransporter activity. While KS-SPAK and SPAK2 both strongly inhibited NKCC1 activity, SPAK2 was a much weaker inhibitor of NKCC2 activity. Removal of the catalytic loop from SPAK2 resulted in an inhibitory effect on NKCC2 similar to that of KS-SPAK. Full-length SPAK is phosphorylated and activated by members of the with-no-lysine[K] (WNK) kinase family. Mutation of a WNK phosphorylation in KS-SPAK did not alter its ability to inhibit NKCC2 activity. In contrast, we found that residues involved in KS-SPAK interactions with cation cotransporters are required for it to inhibit cotransporter activity. Finally, both KS-SPAK and SPAK2 associated with NKCC2, as demonstrated by coimmunoprecipitation. Together, these data identify the structural basis for the differential effects of KS-SPAK and SPAK2 on cation cotransporter activity that may be physiologically important.


Subject(s)
Kidney/metabolism , Protein Serine-Threonine Kinases/metabolism , Solute Carrier Family 12, Member 1/metabolism , Animals , Female , Humans , Isoenzymes , Oocytes/cytology , Oocytes/metabolism , Solute Carrier Family 12, Member 2/metabolism , Xenopus laevis
8.
Micromachines (Basel) ; 14(6)2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37374787

ABSTRACT

With the Internet of Things expanding to more locations across our planet, power becomes the main factor affecting device longevity. There is a need for more novel energy harvesting systems that are able to power remote devices for sustained periods. This publication presents one such device. Based on a novel actuator that utilises off-the-shelf gas mixtures to generate a variable force from temperature change, this publication presents a device capable of generating up to 150mJ per diurnal temperature cycle; this is enough electrical energy to send up to three LoRaWAN transmissions per day using slowly changing environmental temperatures.

9.
Micromachines (Basel) ; 13(11)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36422402

ABSTRACT

As the internet of things expands to more remote locations, solutions are required for long-term remote powering of environmental sensing devices. In this publication, a device is presented which utilises the slow-moving diurnal temperature change present in many natural environments to produce electrical energy. This device utilises a novel actuator which harnesses temperature-dependent phase change to provide a variable force output, and this is combined with energy storage and release apparatus to convert the output force into electrical energy. Appropriate modelling is utilised to identify parameters for system tuning, and a final proof-of-concept solution is constructed and demonstrated to generate up to 10 mJ per 24 h period.

10.
J Clin Invest ; 130(4): 1948-1960, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32149733

ABSTRACT

The major risk factor for kidney stone disease is idiopathic hypercalciuria. Recent evidence implicates a role for defective calcium reabsorption in the renal proximal tubule. We hypothesized that claudin-2, a paracellular cation channel protein, mediates proximal tubule calcium reabsorption. We found that claudin-2-null mice have hypercalciuria due to a primary defect in renal tubule calcium transport and papillary nephrocalcinosis that resembles the intratubular plugs in kidney stone formers. Our findings suggest that a proximal tubule defect in calcium reabsorption predisposes to papillary calcification, providing support for the vas washdown hypothesis. Claudin-2-null mice were also found to have increased net intestinal calcium absorption, but reduced paracellular calcium permeability in the colon, suggesting that this was due to reduced intestinal calcium secretion. Common genetic variants in the claudin-2 gene were associated with decreased tissue expression of claudin-2 and increased risk of kidney stones in 2 large population-based studies. Finally, we describe a family in which males with a rare missense variant in claudin-2 have marked hypercalciuria and kidney stone disease. Our findings indicate that claudin-2 is a key regulator of calcium excretion and a potential target for therapies to prevent kidney stones.


Subject(s)
Claudins , Gene Expression Regulation , Genetic Variation , Hypercalciuria , Kidney Calculi , Kidney Tubules, Proximal , Animals , Calcium/urine , Claudins/deficiency , Claudins/metabolism , Hypercalciuria/genetics , Hypercalciuria/pathology , Hypercalciuria/urine , Kidney Calculi/genetics , Kidney Calculi/pathology , Kidney Calculi/urine , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice , Mice, Knockout
11.
Adv Chronic Kidney Dis ; 25(3): 236-243, 2018 05.
Article in English | MEDLINE | ID: mdl-29793662

ABSTRACT

Magnesium is a divalent cation that fills essential roles as regulator and cofactor in a variety of biological pathways, and maintenance of magnesium balance is vital to human health. The kidney, in concert with the intestine, has an important role in maintaining magnesium homeostasis. Although micropuncture and microperfusion studies in the mammalian nephron have shone a light on magnesium handling in the various nephron segments, much of what we know about the protein mediators of magnesium handling in the kidney have come from more recent genetic studies. In the proximal tubule and thick ascending limb, magnesium reabsorption is believed to occur primarily through the paracellular shunt pathway, which ultimately depends on the electrochemical gradient setup by active sodium reabsorption. In the distal convoluted tubule, magnesium transport is transcellular, although magnesium reabsorption also appears to be related to active sodium reabsorption in this segment. In addition, evidence suggests that magnesium transport is highly regulated, although a specific hormonal regulator of extracellular magnesium has yet to be identified.


Subject(s)
Homeostasis , Kidney/metabolism , Magnesium/metabolism , Humans
12.
J Clin Invest ; 126(7): 2509-18, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27214555

ABSTRACT

Efficient oxygen utilization in the kidney may be supported by paracellular epithelial transport, a form of passive diffusion that is driven by preexisting transepithelial electrochemical gradients. Claudins are tight-junction transmembrane proteins that act as paracellular ion channels in epithelial cells. In the proximal tubule (PT) of the kidney, claudin-2 mediates paracellular sodium reabsorption. Here, we used murine models to investigate the role of claudin-2 in maintaining energy efficiency in the kidney. We found that claudin-2-null mice conserve sodium to the same extent as WT mice, even during profound dietary sodium depletion, as a result of the upregulation of transcellular Na-K-2Cl transport activity in the thick ascending limb of Henle. We hypothesized that shifting sodium transport to transcellular pathways would lead to increased whole-kidney oxygen consumption. Indeed, compared with control animals, oxygen consumption in the kidneys of claudin-2-null mice was markedly increased, resulting in medullary hypoxia. Furthermore, tubular injury in kidneys subjected to bilateral renal ischemia-reperfusion injury was more severe in the absence of claudin-2. Our results indicate that paracellular transport in the PT is required for efficient utilization of oxygen in the service of sodium transport. We speculate that paracellular permeability may have evolved as a general strategy in epithelial tissues to maximize energy efficiency.


Subject(s)
Claudin-2/metabolism , Gene Expression Regulation , Kidney/metabolism , Oxygen/metabolism , Sodium/metabolism , Animals , Diet , Epithelial Cells/metabolism , Female , Ion Transport , Kidney Tubules, Proximal/metabolism , Loop of Henle/metabolism , Magnesium/metabolism , Male , Malondialdehyde/urine , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption , Permeability , Reperfusion Injury/metabolism , Tight Junctions/metabolism
13.
Hypertension ; 58(5): 888-94, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21896937

ABSTRACT

The sodium chloride cotransporter (NCC) is the primary target of thiazides diuretics, drugs used commonly for long-term hypertension therapy. Thiazides also completely reverse the signs of familial hyperkalemic hypertension (FHHt), suggesting that the primary defect in FHHt is increased NCC activity. To test whether increased NCC abundance alone is sufficient to generate the FHHt phenotype, we generated NCC transgenic mice; surprisingly, these mice did not display an FHHt-like phenotype. Systolic blood pressures of NCC transgenic mice did not differ from those of wild-type mice, even after dietary salt loading. NCC transgenic mice also did not display hyperkalemia or hypercalciuria, even when challenged with dietary electrolyte manipulation. Administration of fludrocortisone to NCC transgenic mice, to stimulate NCC, resulted in an increase in systolic blood pressure equivalent to that of wild-type mice (approximately 20 mm Hg). Although total NCC abundance was increased in the transgenic animals, phosphorylated (activated) NCC was not, suggesting that the defect in FHHt involves either activation of ion transport pathways other than NCC, or else direct activation of NCC, in addition to an increase in NCC abundance.


Subject(s)
Phosphorylation/physiology , Pseudohypoaldosteronism/physiopathology , Sodium Chloride Symporters/metabolism , Up-Regulation/physiology , Animals , Blotting, Western , Disease Models, Animal , Fludrocortisone/pharmacology , Gene Expression Regulation , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pseudohypoaldosteronism/drug therapy , Random Allocation , Reference Values , Risk Factors , Sensitivity and Specificity , Signal Transduction/drug effects , Sodium Chloride Symporters/genetics , Up-Regulation/genetics
14.
Cell Metab ; 14(3): 352-64, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21907141

ABSTRACT

The renal thick ascending limb (TAL) and distal convoluted tubule (DCT) play central roles in salt homeostasis and blood pressure regulation. An emerging model suggests that bumetanide- and thiazide-sensitive NaCl transporters (NKCC2 and NCC) along these segments are phosphorylated and activated by WNK kinases, via SPAK and OSR1. Here, we show that a kidney-specific SPAK isoform, which lacks the kinase domain, inhibits phosphorylation of NCC and NKCC2 by full-length SPAK in vitro. Kidney-specific SPAK is highly expressed along the TAL, whereas full-length SPAK is more highly expressed along the DCT. As predicted from the differential expression, SPAK knockout in animals has divergent effects along TAL and DCT, with increased phosphorylated NKCC2 along TAL and decreased phosphorylated NCC along DCT. In mice, extracellular fluid volume depletion shifts SPAK isoform abundance to favor NaCl retention along both segments, indicating that a SPAK isoform switch modulates sodium avidity along the distal nephron.


Subject(s)
Blood Pressure/physiology , Extracellular Fluid/metabolism , Isoenzymes , Kidney Tubules, Distal/metabolism , Loop of Henle/metabolism , Protein Serine-Threonine Kinases , Sodium Chloride/metabolism , Water-Electrolyte Balance/physiology , Animals , Gene Expression Regulation , Homeostasis , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/pharmacology , Kidney Tubules, Distal/cytology , Loop of Henle/cytology , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/pharmacology , Protein Structure, Tertiary , Receptors, Drug/genetics , Receptors, Drug/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Salts/metabolism , Signal Transduction , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3 , Symporters/genetics , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL