Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dis Model Mech ; 16(6)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37283037

RESUMEN

Lower urinary tract symptoms (LUTS) refer to various urological diseases, and incomplete bladder emptying is common among affected patients. The etiology of LUTS is largely unknown, and investigations of LUTS suggest that bladder fibrosis contributes to pathogenesis of LUTS. MicroRNAs (miRNAs) are short (∼22 nucleotides), non-coding RNAs that repress target gene expression by a combination of mRNA degradation and translation inhibition. The miR-29 family is best known for its anti-fibrotic role in various organs. miR-29 was decreased in bladders of patients with outlet obstruction and a rat model of bladder outlet obstruction, suggesting that miR-29 may contribute to impaired bladder function subsequent to tissue fibrosis. We characterized bladder function in male mice lacking expression of Mir29a and Mir29b-1 (miR-29a/b1). Lack of miR-29a/b1 resulted in severe urinary retention, increased voiding duration and reduced flow rate, and these mice failed to void or voided irregularly during anesthetized cytometry. Collagens and elastin were increased in bladders of mice lacking miR-29a/b1. These findings reveal an important role for miR-29 in bladder homeostasis and suggest the therapeutic potential of miR-29 to improve symptoms in patients with LUTS.


Asunto(s)
MicroARNs , Vejiga Urinaria , Ratones , Masculino , Ratas , Animales , Vejiga Urinaria/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fibrosis , Colágeno
2.
Chem Sci ; 11(5): 1307-1315, 2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-33209250

RESUMEN

The use of hyperosmolar agents (osmotherapy) has been a major treatment for intracranial hypertension, which occurs frequently in brain diseases or trauma. However, side-effects of osmotherapy on the brain, especially on the blood-brain barrier (BBB) are still not fully understood. Hyperosmolar conditions, termed hyperosmolality here, are known to transiently disrupt the tight junctions (TJs) at the endothelium of the BBB resulting in loss of BBB function. Present techniques for evaluation of BBB transport typically reveal aggregated responses from the entirety of BBB transport components, with little or no opportunity to evaluate heterogeneity present in the system. In this study, we utilized potentiometric-scanning ion conductance microscopy (P-SICM) to acquire nanometer-scale conductance maps of Madin-Darby Canine Kidney strain II (MDCKII) cells under hyperosmolality, from which two types of TJs, bicellular tight junctions (bTJs) and tricellular tight junctions (tTJs), can be visualized and differentiated. We discovered that hyperosmolality leads to increased conductance at tTJs without significant alteration in conductance at bTJs. To quantify this effect, an automated computer vision algorithm was designed to extract and calculate conductance components at both tTJs and bTJs. Additionally, lowering Ca2+ concentration in the bath facilitates tTJ disruption under hyperosmolality. Strengthening tTJ structure by overexpressing immunoglobulin-like domain-containing receptor 1 (ILDR1) protein abrogates the effect of hyperosmolality. We posit that osmotic stress physically disrupts tTJ structure, as evidenced by super-resolution microscopy. Findings from this study not only provide a high-resolution view of TJ structure and function, but also can inform current osmotherapy and drug delivery strategies for brain diseases.

3.
mBio ; 11(1)2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32047126

RESUMEN

Venezuelan and western equine encephalitis viruses (VEEV and WEEV, respectively) invade the central nervous system (CNS) early during infection, via neuronal and hematogenous routes. While viral replication mediates host shutoff, including expression of type I interferons (IFN), few studies have addressed how alphaviruses gain access to the CNS during established infection or the mechanisms of viral crossing at the blood-brain barrier (BBB). Here, we show that hematogenous dissemination of VEEV and WEEV into the CNS occurs via caveolin-1 (Cav-1)-mediated transcytosis (Cav-MT) across an intact BBB, which is impeded by IFN and inhibitors of RhoA GTPase. Use of reporter and nonreplicative strains also demonstrates that IFN signaling mediates viral restriction within cells comprising the neurovascular unit (NVU), differentially rendering brain endothelial cells, pericytes, and astrocytes permissive to viral replication. Transmission and immunoelectron microscopy revealed early events in virus internalization and Cav-1 association within brain endothelial cells. Cav-1-deficient mice exhibit diminished CNS VEEV and WEEV titers during early infection, whereas viral burdens in peripheral tissues remained unchanged. Our findings show that alphaviruses exploit Cav-MT to enter the CNS and that IFN differentially restricts this process at the BBB.IMPORTANCE VEEV, WEEV, and eastern equine encephalitis virus (EEEV) are emerging infectious diseases in the Americas, and they have caused several major outbreaks in the human and horse population during the past few decades. Shortly after infection, these viruses can infect the CNS, resulting in severe long-term neurological deficits or death. Neuroinvasion has been associated with virus entry into the CNS directly from the bloodstream; however, the underlying molecular mechanisms have remained largely unknown. Here, we demonstrate that following peripheral infection alphavirus augments vesicular formation/trafficking at the BBB and utilizes Cav-MT to cross an intact BBB, a process regulated by activators of Rho GTPases within brain endothelium. In vivo examination of early viral entry in Cav-1-deficient mice revealed significantly lower viral burdens in the brain than in similarly infected wild-type animals. These studies identify a potentially targetable pathway to limit neuroinvasion by alphaviruses.


Asunto(s)
Barrera Hematoencefálica/virología , Caveolas/virología , Virus de la Encefalitis Equina Venezolana/fisiología , Virus de la Encefalitis Equina del Oeste/fisiología , Transcitosis , Internalización del Virus , Animales , Caveolina 1/genética , Línea Celular , Sistema Nervioso Central/virología , Células Endoteliales/virología , Masculino , Ratones Endogámicos C57BL , Replicación Viral
4.
Proc Natl Acad Sci U S A ; 116(38): 19176-19186, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31488724

RESUMEN

Familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC) was previously considered to be a paracellular channelopathy caused by mutations in the claudin-16 and claudin-19 genes. Here, we provide evidence that a missense FHHNC mutation c.908C>G (p.T303R) in the claudin-16 gene interferes with the phosphorylation in the claudin-16 protein. The claudin-16 protein carrying phosphorylation at residue T303 is localized in the distal convoluted tubule (DCT) but not in the thick ascending limb (TAL) of the mouse kidney. The phosphomimetic claudin-16 protein carrying the T303E mutation but not the wildtype claudin-16 or the T303R mutant protein increases the Trpv5 channel conductance and membrane abundance in human kidney cells. Phosphorylated claudin-16 and Trpv5 are colocalized in the luminal membrane of the mouse DCT tubule; phosphomimetic claudin-16 and Trpv5 interact in the yeast and mammalian cell membranes. Knockdown of claudin-16 gene expression in transgenic mouse kidney delocalizes Trpv5 from the luminal membrane in the DCT. Unlike wildtype claudin-16, phosphomimetic claudin-16 is delocalized from the tight junction but relocated to the apical membrane in renal epithelial cells because of diminished binding affinity to ZO-1. High-Ca2+ diet reduces the phosphorylation of claudin-16 protein at T303 in the DCT of mouse kidney via the PTH signaling cascade. Knockout of the PTH receptor, PTH1R, from the mouse kidney abrogates the claudin-16 phosphorylation at T303. Together, these results suggest a pathogenic mechanism for FHHNC involving transcellular Ca2+ pathway in the DCT and identify a molecular component in renal Ca2+ homeostasis under direct regulation of PTH.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Claudinas/metabolismo , Túbulos Renales Distales/metabolismo , Canales Catiónicos TRPV/metabolismo , Uniones Estrechas/metabolismo , Transcitosis , Animales , Canales de Calcio/genética , Permeabilidad de la Membrana Celular , Claudinas/antagonistas & inhibidores , Claudinas/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Fosforilación , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética
5.
J Cell Sci ; 132(4)2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30209136

RESUMEN

Tight junctions have been hypothesized to act as molecular fences in the plasma membrane of epithelial cells, helping to form differentiated apical and basolateral domains. While this fence function is believed to arise from the interaction of four-pass transmembrane claudins, the complexity of tight junctions has made direct evidence of their role as a putative diffusion barrier difficult to obtain. Here, we address this challenge by reconstituting claudin-4 into giant unilamellar vesicles using microfluidic jetting. We find that reconstituted claudin-4 alone can form adhesive membrane interfaces without the accessory proteins that are present in vivo By controlling the molecular composition of the inner and outer leaflets of jetted vesicle membranes, we show that claudin-4-mediated interfaces can drive partitioning of extracellular membrane proteins with ectodomains as small as 5 nm but not of inner or outer leaflet lipids. Our findings indicate that homotypic interactions of claudins and their small size can contribute to the polarization of epithelial cells.


Asunto(s)
Membrana Celular/metabolismo , Claudina-4/metabolismo , Proteolípidos/metabolismo , Uniones Estrechas/metabolismo , Claudina-4/genética , Células Epiteliales/metabolismo , Humanos , Liposomas Unilamelares/metabolismo
6.
Small ; 14(18): e1702945, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29226633

RESUMEN

Integration of dual-barrel membrane patch-ion channel probes (MP-ICPs) to scanning ion conductance microscopy (SICM) holds promise of providing a revolutionized approach of spatially resolved chemical sensing. A series of experiments are performed to further the understanding of the system and to answer some fundamental questions, in preparation for future developments of this approach. First, MP-ICPs are constructed that contain different types of ion channels including transient receptor potential vanilloid 1 and large conductance Ca2+ -activated K+ channels to establish the generalizability of the methods. Next, the capability of the MP-ICP platforms in single ion channel activity measurements is proved. In addition, the interplay between the SICM barrel and the ICP barrel is studied. For ion channels gated by uncharged ligands, channel activity at the ICP barrel is unaffected by the SICM barrel potential; whereas for ion channels that are gated by charged ligands, enhanced channel activity can be obtained by biasing the SICM barrel at potentials with opposite polarity to the charge of the ligand molecules. Finally, a proof-of-principle experiment is performed and site-specific molecular/ionic flux sensing is demonstrated at single-ion-channel level, which show that the MP-ICP platform can be used to quantify local molecular/ionic concentrations.


Asunto(s)
Canales Iónicos/química , Microscopía/métodos , Transporte Iónico , Porosidad
7.
Anal Chem ; 89(24): 13603-13609, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29164870

RESUMEN

Understanding ion transport properties at various interfaces, especially at small length scales, is critical in advancing our knowledge of membrane materials and cell biology. Recently, we described potentiometric-scanning ion conductance microscopy (P-SICM) for ion-conductance measurement in polymer membranes and epithelial cell monolayers at discrete points in a sample. Here, we combine hopping mode techniques with P-SICM to allow simultaneous nanometer-scale conductance and topography mapping. First validated with standard synthetic membranes and then demonstrated in living epithelial cell monolayers under physiological conditions, this new method allows direct visualization of heterogeneous ion transport of biological samples for the first time. These advances provide a noncontact local probe, require no labeling, and present a new tool for quantifying intrinsic transport properties of a variety of biological samples.


Asunto(s)
Transporte Iónico , Células de Riñón Canino Madin Darby/química , Polímeros/química , Animales , Células Cultivadas , Espectroscopía Dieléctrica , Perros , Conductividad Eléctrica , Células de Riñón Canino Madin Darby/metabolismo , Microscopía Confocal , Nanoporos , Potenciometría
8.
Proc Natl Acad Sci U S A ; 114(20): 5271-5276, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28461473

RESUMEN

Whether the tight junction is permeable to water remains highly controversial. Here, we provide evidence that the tricellular tight junction is important for paracellular water permeation and that Ig-like domain containing receptor 1 (ILDR1) regulates its permeability. In the mouse kidney, ILDR1 is localized to tricellular tight junctions of the distal tubules. Genetic knockout of Ildr1 in the mouse kidney causes polyuria and polydipsia due to renal concentrating defects. Microperfusion of live renal distal tubules reveals that they are impermeable to water in normal animals but become highly permeable to water in Ildr1 knockout animals whereas paracellular ionic permeabilities in the Ildr1 knockout mouse renal tubules are not affected. Vasopressin cannot correct paracellular water loss in Ildr1 knockout animals despite normal effects on the transcellular aquaporin-2-dependent pathway. In cultured renal epithelial cells normally lacking the expression of Ildr1, overexpression of Ildr1 significantly reduces the paracellular water permeability. Together, our study provides a mechanism of how cells transport water and shows how such a mechanism may be exploited as a therapeutic approach to maintain water homeostasis.


Asunto(s)
Acuaporinas/fisiología , Capacidad de Concentración Renal/fisiología , Receptores de Superficie Celular/fisiología , Animales , Acuaporina 2/metabolismo , Acuaporinas/metabolismo , Transporte Biológico , Permeabilidad de la Membrana Celular/fisiología , Células Epiteliales/metabolismo , Riñón/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales Distales/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptores de Superficie Celular/metabolismo , Uniones Estrechas/metabolismo , Uniones Estrechas/fisiología , Vasopresinas/metabolismo
9.
J Neuroimmunol ; 308: 118-130, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28501330

RESUMEN

Viral infections of the central nervous system (CNS) are often associated with blood-brain barrier (BBB) disruption, yet the impact of virus replication and immune cell recruitment on BBB integrity are incompletely understood. Using two-photon microscopy, we demonstrate that Venezuelan equine encephalitis virus (VEEV) strain TC83-GFP, a GFP expressing, attenuated strain with a G3A mutation within the 5' UTR that is associated with increased sensitivity to type I interferons (IFNs), does not directly impact BBB permeability. Following intranasal infection of both wild-type and IFN-induced protein with tetratricopeptide repeats 1 (IFIT1)-deficient mice, which fail to block TC83-specific RNA translation, virus spreads to the olfactory bulb and cortex via migration along axonal tracts of neurons originating from the olfactory neuroepithelium. Global dissemination of virus in the CNS by 2days post-infection (dpi) was associated with increased BBB permeability in the olfactory bulb, but not in the cortex or hindbrain, where permeability only increased after the recruitment of CX3CR1+ and CCR2+ mononuclear cells on 6 dpi, which corresponded with tight junction loss and claudin 5 redistribution. Importantly, despite higher levels of viral replication, similar results were obtained in IFIT1-deficient mice. These findings indicate that TC83 gains CNS access via anterograde axonal migration without directly altering BBB function and that mononuclear and endothelial cell interactions may underlie BBB disruption during alphavirus encephalitis.


Asunto(s)
Infecciones por Alphavirus/patología , Barrera Hematoencefálica/fisiopatología , Encéfalo/metabolismo , Encéfalo/virología , Replicación Viral/fisiología , Complejo 1 de Proteína Adaptadora/genética , Complejo 1 de Proteína Adaptadora/metabolismo , Infecciones por Alphavirus/genética , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/ultraestructura , Barrera Hematoencefálica/virología , Receptor 1 de Quimiocinas CX3C , Permeabilidad Capilar/fisiología , Células Cultivadas , Corteza Cerebral/citología , Cricetinae , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/fisiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Células Epiteliales/ultraestructura , Células Epiteliales/virología , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Internalización del Virus
10.
Proc Natl Acad Sci U S A ; 114(16): E3344-E3353, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373577

RESUMEN

Renal Ca2+ reabsorption is essential for maintaining systemic Ca2+ homeostasis and is tightly regulated through the parathyroid hormone (PTH)/PTHrP receptor (PTH1R) signaling pathway. We investigated the role of PTH1R in the kidney by generating a mouse model with targeted deletion of PTH1R in the thick ascending limb of Henle (TAL) and in distal convoluted tubules (DCTs): Ksp-cre;Pth1rfl/fl Mutant mice exhibited hypercalciuria and had lower serum calcium and markedly increased serum PTH levels. Unexpectedly, proteins involved in transcellular Ca2+ reabsorption in DCTs were not decreased. However, claudin14 (Cldn14), an inhibitory factor of the paracellular Ca2+ transport in the TAL, was significantly increased. Analyses by flow cytometry as well as the use of Cldn14-lacZ knock-in reporter mice confirmed increased Cldn14 expression and promoter activity in the TAL of Ksp-cre;Pth1rfl/fl mice. Moreover, PTH treatment of HEK293 cells stably transfected with CLDN14-GFP, together with PTH1R, induced cytosolic translocation of CLDN14 from the tight junction. Furthermore, mice with high serum PTH levels, regardless of high or low serum calcium, demonstrated that PTH/PTH1R signaling exerts a suppressive effect on Cldn14. We therefore conclude that PTH1R signaling directly and indirectly regulates the paracellular Ca2+ transport pathway by modulating Cldn14 expression in the TAL. Finally, systemic deletion of Cldn14 completely rescued the hypercalciuric and lower serum calcium phenotype in Ksp-cre;Pth1rfl/fl mice, emphasizing the importance of PTH in inhibiting Cldn14. Consequently, suppressing CLDN14 could provide a potential treatment to correct urinary Ca2+ loss, particularly in patients with hypoparathyroidism.


Asunto(s)
Calcio/metabolismo , Claudinas/fisiología , Extremidades/fisiología , Regulación de la Expresión Génica , Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Uniones Estrechas/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Transducción de Señal
11.
Proc Natl Acad Sci U S A ; 114(2): E219-E227, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28028216

RESUMEN

The thick ascending limb (TAL) of Henle's loop drives paracellular Na+, Ca2+, and Mg2+ reabsorption via the tight junction (TJ). The TJ is composed of claudins that consist of four transmembrane segments, two extracellular segments (ECS1 and -2), and one intracellular loop. Claudins interact within the same (cis) and opposing (trans) plasma membranes. The claudins Cldn10b, -16, and -19 facilitate cation reabsorption in the TAL, and their absence leads to a severe disturbance of renal ion homeostasis. We combined electrophysiological measurements on microperfused mouse TAL segments with subsequent analysis of claudin expression by immunostaining and confocal microscopy. Claudin interaction properties were examined using heterologous expression in the TJ-free cell line HEK 293, live-cell imaging, and Förster/FRET. To reveal determinants of interaction properties, a set of TAL claudin protein chimeras was created and analyzed. Our main findings are that (i) TAL TJs show a mosaic expression pattern of either cldn10b or cldn3/cldn16/cldn19 in a complex; (ii) TJs dominated by cldn10b prefer Na+ over Mg2+, whereas TJs dominated by cldn16 favor Mg2+ over Na+; (iii) cldn10b does not interact with other TAL claudins, whereas cldn3 and cldn16 can interact with cldn19 to form joint strands; and (iv) further claudin segments in addition to ECS2 are crucial for trans interaction. We suggest the existence of at least two spatially distinct types of paracellular channels in TAL: a cldn10b-based channel for monovalent cations such as Na+ and a spatially distinct site for reabsorption of divalent cations such as Ca2+ and Mg2.


Asunto(s)
Claudinas/metabolismo , Asa de la Nefrona/metabolismo , Magnesio/metabolismo , Sodio/metabolismo , Animales , Claudinas/genética , Células HEK293 , Humanos , Asa de la Nefrona/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas Sprague-Dawley , Uniones Estrechas/metabolismo
12.
Pflugers Arch ; 469(1): 105-113, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27878608

RESUMEN

Claudins are discovered to be key players in renal epithelial physiology. They are involved in developmental, physiological, and pathophysiological differentiation. In the glomerular podocytes, claudin-1 is an important determinant of cell junction fate. In the proximal tubule, claudin-2 plays important roles in paracellular salt reabsorption. In the thick ascending limb, claudin-14, -16, and -19 regulate the paracellular reabsorption of calcium and magnesium. Recessive mutations in claudin-16 or -19 cause an inherited calcium and magnesium losing disease. Synonymous variants in claudin-14 have been associated with hypercalciuric nephrolithiasis by genome-wide association studies (GWASs). More importantly, claudin-14 gene expression can be regulated by extracellular calcium levels via the calcium sensing receptor. In the distal tubules, claudin-4 and -8 form paracellular chloride pathway to facilitate electrogenic sodium reabsorption. Aldosterone, WNK4, Cap1, and KLHL3 are powerful regulators of claudin and the paracellular chloride permeability. The lessons learned on claudins from the kidney will have a broader impact on tight junction biology in other epithelia and endothelia.


Asunto(s)
Claudinas/metabolismo , Túbulos Renales Proximales/metabolismo , Animales , Claudinas/genética , Expresión Génica/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Uniones Estrechas/metabolismo
13.
J Am Soc Nephrol ; 28(1): 106-117, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27151920

RESUMEN

The tight junction (TJ) has a key role in regulating paracellular permeability to water and solutes in the kidney. However, the functional role of the TJ in the glomerular podocyte is unclear. In diabetic nephropathy, the gene expression of claudins, in particular claudin-1, is markedly upregulated in the podocyte, accompanied by a tighter filtration slit and the appearance of TJ-like structures between the foot processes. However, there is no definitive evidence to show slit diaphragm (SD) to TJ transition in vivo Here, we report the generation of a claudin-1 transgenic mouse model with doxycycline-inducible transgene expression specifically in the glomerular podocyte. We found that induction of claudin-1 gene expression in mature podocytes caused profound proteinuria, and with deep-etching freeze-fracture electron microscopy, we resolved the ultrastructural change in the claudin-1-induced SD-TJ transition. Notably, immunolabeling of kidney proteins revealed that claudin-1 induction destabilized the SD protein complex in podocytes, with significantly reduced expression and altered localization of nephrin and podocin proteins. Mechanistically, claudin-1 interacted with both nephrin and podocin through cis- and trans-associations in cultured cells. Furthermore, the rat puromycin aminonucleoside nephrosis model, previously suspected of undergoing SD-TJ transition, exhibited upregulated expression levels of claudin-1 mRNA and protein in podocytes. Together, our data attest to the novel concept that claudins and the TJ have essential roles in podocyte pathophysiology and that claudin interactions with SD components may facilitate SD-TJ transition that appears to be common to many nephrotic conditions.


Asunto(s)
Claudina-1/biosíntesis , Podocitos/metabolismo , Podocitos/ultraestructura , Proteinuria/etiología , Uniones Estrechas/patología , Animales , Glomérulos Renales/citología , Masculino , Ratas , Ratas Sprague-Dawley
14.
Anal Chem ; 88(19): 9630-9637, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27618532

RESUMEN

Tight junctions (TJs) are barrier forming structures of epithelia and can be described as tightly sealed intercellular spaces. Transport properties have been extensively studied for bicellular TJs (bTJs). Knowledge of the barrier functions of tricellular junctions (tTJs) are less well understood, due largely to a lack of proper techniques to locally measure discrete tTJ properties within a much larger area of epithelium. In this study, we use a nanoscale pipet to precisely locate tTJs within epithelia and measure the apparent local conductance of tTJs with a technique termed potentiometric scanning ion conductance microscopy (P-SICM). P-SICM shows the ability to differentiate transport through tTJs and bTJs, which was not possible with previous techniques and assays. We describe P-SICM investigations of both wild type and tricellulin overexpression Madin-Darby Canine Kidney (strain II, MDCKII) cells.


Asunto(s)
Epitelio/química , Proteína 2 con Dominio MARVEL/análisis , Potenciometría , Animales , Perros , Células de Riñón Canino Madin Darby , Microscopía Electroquímica de Rastreo , Tamaño de la Partícula
15.
Curr Opin Nephrol Hypertens ; 25(5): 424-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27490784

RESUMEN

PURPOSE OF REVIEW: The paracellular pathway through the tight junction provides an important route for chloride reabsorption in the collecting duct of the kidney. This review describes recent findings of how defects in paracellular chloride permeation pathway may cause kidney diseases and how such a pathway may be regulated to maintain normal chloride homeostasis. RECENT FINDINGS: The tight junction in the collecting duct expresses two important claudin genes - claudin-4 and claudin-8. Transgenic knockout of either claudin gene causes hypotension, hypochloremia, and metabolic alkalosis in experimental animals. The claudin-4 mediated chloride permeability can be regulated by a protease endogenously expressed by the collecting duct cell - channel-activating protease 1. Channel-activating protease 1 regulates the intercellular interaction of claudin-4 and its membrane stability. Kelch-like 3, previously identified as a causal gene for Gordon's syndrome, also known as pseudohypoaldosteronism II, directly interacts with claudin-8 and regulates its ubiquitination and degradation. The dominant pseudohypoaldosteronism-II mutation (R528H) in Kelch-like 3 abolishes claudin-8 binding, ubiquitination, and degradation. SUMMARY: The paracellular chloride permeation pathway in the kidney is an important but understudied area in nephrology. It plays vital roles in renal salt handling and regulation of extracellular fluid volume and blood pressure. Two claudin proteins, claudin-4 and claudin-8, contribute to the function of this paracellular pathway. Deletion of either claudin protein from the collecting duct causes renal chloride reabsorption defects and low blood pressure. Claudins can be regulated on posttranslational levels by several mechanisms involving protease and ubiquitin ligase. Deregulation of claudins may cause human hypertension as exemplified in the Gordon's syndrome.


Asunto(s)
Cloruros/metabolismo , Claudina-4/metabolismo , Enfermedades Renales/metabolismo , Túbulos Renales Colectores/metabolismo , Uniones Estrechas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Presión Sanguínea , Proteínas Portadoras , Claudina-4/genética , Claudinas/genética , Humanos , Enfermedades Renales/etiología , Túbulos Renales Colectores/enzimología , Proteínas de Microfilamentos , Permeabilidad , Seudohipoaldosteronismo/metabolismo , Reabsorción Renal , Serina Endopeptidasas/metabolismo
16.
Tissue Barriers ; 4(1): e1142492, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27141421

RESUMEN

Brain endothelial cells form a unique cellular structure known as the tight junction to regulate the exchanges between the blood and the parenchyma by limiting the paracellular diffusion of blood-borne substance. Together with the restricted pathway of transcytosis, the tight junction in the brain endothelial cells provides the central nervous system (CNS) with effective protection against both the foreign pathogens and the host immune cells, which is also termed the "blood-brain barrier." The blood-brain barrier is particularly important for defending against neurotropic viral infections that have become a major source of diseases worldwide. Many neurotropic viruses are able to cross the BBB and infect the CNS through very poorly understood processes. This review focuses upon the structural and functional changes of the brain endothelial tight junction in response to viral infections in the CNS and how the tight junction changes may be studied with advanced imaging and recording approaches to reveal novel processes used by the viruses to cross the barrier system. Additional emphasis is placed upon new countermeasures that can act directly upon the tight junction to improve the pathogen clearance and minimize the inflammatory damage.


Asunto(s)
Barrera Hematoencefálica/virología , Enfermedades Virales del Sistema Nervioso Central/metabolismo , Interacciones Huésped-Patógeno , Animales , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Humanos , Uniones Estrechas/metabolismo , Uniones Estrechas/virología
17.
Curr Opin Nephrol Hypertens ; 25(4): 308-13, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27191348

RESUMEN

PURPOSE OF REVIEW: The tight junction conductance made of the claudin-based paracellular channel is important in the regulation of calcium and magnesium reabsorption in the kidney. This review describes recent findings of the structure, the function, and the physiologic regulation of claudin-14, claudin-16, and claudin-19 channels that through protein interactions confer calcium and magnesium permeability to the tight junction. RECENT FINDINGS: Mutations in two tight junction genes - claudin-16 and claudin-19 - cause the inherited renal disorder familial hypomagnesemia with hypercalciuria and nephrocalcinosis. A recent genome-wide association study has identified claudin-14 as a major risk gene of hypercalciuric nephrolithiasis. The crystal structure of claudin-19 has recently been resolved allowing the reconstruction of a claudin assembly model from cis-dimers made of claudin-16 and claudin-19 interaction. MicroRNAs have been identified as novel regulators of the claudin-14 gene. The microRNA-claudin-14 operon is directly regulated by the Ca sensing receptor gene in response to hypercalcemia. SUMMARY: The paracellular pathway in the kidney is particularly important for mineral metabolism. Three claudin proteins - claudin-14, claudin-16, and claudin-19 - contribute to the structure and function of this paracellular pathway. Genetic mutations and gene expression changes in these claudins may lead to alteration of the paracellular permeability to calcium and magnesium, ultimately affecting renal mineral metabolism.


Asunto(s)
Calcio/metabolismo , Claudinas/metabolismo , Magnesio/metabolismo , Uniones Estrechas/metabolismo , Claudinas/genética , Estudio de Asociación del Genoma Completo , Humanos , Hipercalciuria/genética , Hipercalciuria/metabolismo , Riñón/metabolismo , MicroARNs/metabolismo , Nefrocalcinosis/genética , Nefrocalcinosis/metabolismo , Nefrolitiasis/genética , Nefrolitiasis/metabolismo , Receptores Sensibles al Calcio/metabolismo , Defectos Congénitos del Transporte Tubular Renal/genética , Defectos Congénitos del Transporte Tubular Renal/metabolismo
18.
J Bone Miner Res ; 31(3): 498-513, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26426912

RESUMEN

Claudin-16 protein (CLDN16) is a component of tight junctions (TJ) with a restrictive distribution so far demonstrated mainly in the kidney. Here, we demonstrate the expression of CLDN16 also in the tooth germ and show that claudin-16 gene (CLDN16) mutations result in amelogenesis imperfecta (AI) in the 5 studied patients with familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC). To investigate the role of CLDN16 in tooth formation, we studied a murine model of FHHNC and showed that CLDN16 deficiency led to altered secretory ameloblast TJ structure, lowering of extracellular pH in the forming enamel matrix, and abnormal enamel matrix protein processing, resulting in an enamel phenotype closely resembling human AI. This study unravels an association of FHHNC owing to CLDN16 mutations with AI, which is directly related to the loss of function of CLDN16 during amelogenesis. Overall, this study indicates for the first time the importance of a TJ protein in tooth formation and underlines the need to establish a specific dental follow-up for these patients.


Asunto(s)
Ameloblastos/metabolismo , Claudinas/deficiencia , Esmalte Dental/anomalías , Esmalte Dental/metabolismo , Uniones Estrechas/metabolismo , Adulto , Ameloblastos/patología , Amelogénesis Imperfecta/metabolismo , Amelogénesis Imperfecta/patología , Animales , Niño , Claudinas/genética , Esmalte Dental/patología , Femenino , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones , Persona de Mediana Edad , Mutación/genética , Fenotipo , Síndrome , Adulto Joven
19.
Pflugers Arch ; 468(2): 293-303, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26497703

RESUMEN

The thick ascending limb of Henle's loop (TAL) drives an important part of the reabsorption of divalent cations. This reabsorption occurs via the paracellular pathway formed by the tight junction (TJ), which in the TAL shows cation selectivity. Claudins, a family of TJ proteins, determine the permeability and selectivity of this pathway. Mice were fed with normal or high-Ca(2+) diet, and effects on the reabsorptive properties of cortical and medullary TAL segments were analysed by tubule microdissection and microperfusion. Claudin expression was investigated by immunostaining and quantitative PCR. We show that the TAL adapted to high Ca(2+) load in a sub-segment-specific manner. In medullary TAL, transcellular NaCl transport was attenuated. The transepithelial voltage decreased from 10.9 ± 0.6 mV at control diet to 8.3 ± 0.5 mV at high Ca(2+) load, thereby reducing the driving force for Ca(2+) and Mg(2+) uptake. Cortical TAL showed a reduction in paracellular Ca(2+) and Mg(2+) permeabilities from 8.2 ± 0.7 to 6.2 ± 0.5 ∙ 10(-4) cm/s and from 4.8 ± 0.5 to 3.0 ± 0.2 · 10(-4) cm/s at control and high-Ca(2+) diet, respectively. Expression, localisation and regulation of claudins 10, 14, 16 and 19 differed along the corticomedullary axis: Towards the cortex, the main site of divalent cation reabsorption in TAL, high-Ca(2+) intake led to a strong upregulation of claudin-14 within TAL TJs while claudin-16 and -19 were unaltered. Towards the inner medulla, only claudin-10 was present in TAL TJ strands. In summary, high-Ca(2+) diet induced a reduction of divalent cation reabsorption via a diminution of NaCl transport and driving force in mTAL and via decreased paracellular permeabilities in cTAL. We reveal an important regulatory pattern along the corticomedullary axis and improve the understanding how the kidney disposes of detrimental excess Ca(2+).


Asunto(s)
Calcio de la Dieta/farmacología , Asa de la Nefrona/metabolismo , Reabsorción Renal , Uniones Estrechas/metabolismo , Potenciales de Acción , Animales , Calcio/metabolismo , Claudinas/genética , Claudinas/metabolismo , Femenino , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Médula Renal/efectos de los fármacos , Médula Renal/metabolismo , Asa de la Nefrona/efectos de los fármacos , Magnesio/metabolismo , Ratones , Ratones Endogámicos C57BL , Cloruro de Sodio/metabolismo , Uniones Estrechas/efectos de los fármacos , Regulación hacia Arriba
20.
Tissue Barriers ; 3(4): e1105907, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26716077

RESUMEN

Direct recording of tight junction permeability is of pivotal importance to many biologic fields. Previous approaches bear an intrinsic disadvantage due to the difficulty of separating tight junction conductance from nearby membrane conductance. Here, we propose the design of Double whole-cell Voltage Clamp - Ion Conductance Microscopy (DVC-ICM) based on previously demonstrated potentiometric scanning of local conductive pathways. As proposed, DVC-ICM utilizes two coordinated whole-cell patch-clamps to neutralize the apical membrane current during potentiometric scanning, which in models described here will profoundly enhance the specificity of tight junction recording. Several potential pitfalls are considered, evaluated and addressed with alternative countermeasures.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...