Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Aerosol Med Pulm Drug Deliv ; 37(2): 64-76, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38354286

ABSTRACT

Background: Hyperosmolar aerosols appear to promote or suppress upper airway dysfunction caused by dehydration in a composition-dependent manner. We sought to explore this composition dependence experimentally, in an interventional human clinical study, and theoretically, by numerical analysis of upper airway ion and water transport. Methods: In a double-blinded, placebo-controlled clinical study, phonation threshold pressure (PTP) was measured prenasal and postnasal inhalation of hypertonic aerosols of NaCl, KCl, CaCl2, and MgCl2 in seven human subjects. Numerical analysis of water and solute exchanges in the upper airways following deposition of these same aerosols was performed using a mathematical model previously described in the literature. Results: PTP decreased by 9%-22% relative to baseline (p < 0.05) for all salts within the first 30 minutes postadministration, indicating effective laryngeal hydration. Only MgCl2 reduced PTP beyond 90 minutes (21% below baseline at 2 hours postadministration). By numerical analysis, we determined that, while airway water volume up to 15 minutes postdeposition is dictated by osmolarity, after 30 minutes, divalent cation salts, such as MgCl2, better retain airway surface liquid (ASL) volume by slow paracellular clearance of the divalent cation. Fall of CFTR chloride flux with rise in ASL height, a promoter of airway acidification, appears to be a signature of permeating cation (NaCl) and nonpermeating anion (mannitol) aerosol deposition. For hypertonic aerosols that lack permeating cation and include permeating anion (CaCl2 and MgCl2), this acid-trigger signature does not exist. Conclusions: Nonpermeating cation and permeating anion hypertonic aerosols appear to hydrate upper airways longer and, rather than provoke, may reduce laryngeal dysfunction such as cough and bronchoconstriction.


Subject(s)
Salts , Sodium Chloride , Humans , Administration, Inhalation , Aerosols , Anions , Calcium Chloride , Cations, Divalent , Hydrogen-Ion Concentration , Respiratory Aerosols and Droplets , Saline Solution, Hypertonic , Water
2.
Sci Rep ; 12(1): 4599, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35351914

ABSTRACT

Dehydration of the upper airways increases risks of respiratory diseases from COVID-19 to asthma and COPD. We find in human volunteer studies involving 464 human subjects in Germany, the US, and India that respiratory droplet generation increases by up to 4 orders of magnitude in dehydration-associated states of advanced age (n = 357), elevated BMI-age (n = 148), strenuous exercise (n = 20) and SARS-CoV-2 infection (n = 87), and falls with hydration of the nose, larynx and trachea by calcium-rich hypertonic salts. We also find in a protocol of exercise-induced airway dehydration that hydration of the airways by calcium-rich salts increases oxygenation relative to a non-treatment control (P < 0.05). In a random control study of COVID-19 positive subjects (n = 40), thrice-a-day delivery of the calcium-rich hypertonic salts (active) suppressed respiratory droplet generation by 51% ± 11% and increased oxygen saturation over three days of treatment by 48.08% ± 9.61% (P < 0.001), while no changes were observed in the nasal-saline control group. Self-reported symptoms significantly declined in the active group and did not decline in the control group. Hydration of the upper airways appears promising as a non-drug approach for reducing risks of respiratory diseases such as COVID-19.


Subject(s)
COVID-19 , Larynx , Exercise , Humans , SARS-CoV-2 , Trachea
3.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33563754

ABSTRACT

COVID-19 transmits by droplets generated from surfaces of airway mucus during processes of respiration within hosts infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. We studied respiratory droplet generation and exhalation in human and nonhuman primate subjects with and without COVID-19 infection to explore whether SARS-CoV-2 infection, and other changes in physiological state, translate into observable evolution of numbers and sizes of exhaled respiratory droplets in healthy and diseased subjects. In our observational cohort study of the exhaled breath particles of 194 healthy human subjects, and in our experimental infection study of eight nonhuman primates infected, by aerosol, with SARS-CoV-2, we found that exhaled aerosol particles vary between subjects by three orders of magnitude, with exhaled respiratory droplet number increasing with degree of COVID-19 infection and elevated BMI-years. We observed that 18% of human subjects (35) accounted for 80% of the exhaled bioaerosol of the group (194), reflecting a superspreader distribution of bioaerosol analogous to a classical 20:80 superspreader of infection distribution. These findings suggest that quantitative assessment and control of exhaled aerosol may be critical to slowing the airborne spread of COVID-19 in the absence of an effective and widely disseminated vaccine.


Subject(s)
COVID-19/physiopathology , COVID-19/transmission , Exhalation/physiology , Obesity/physiopathology , Aerosols , Age Factors , Animals , Body Mass Index , COVID-19/epidemiology , COVID-19/virology , Cohort Studies , Humans , Mucus/chemistry , Mucus/virology , Obesity/epidemiology , Obesity/virology , Particle Size , Primates , Respiratory System/metabolism , SARS-CoV-2/isolation & purification , Viral Load
4.
Nat Biomed Eng ; 5(3): 240-251, 2021 03.
Article in English | MEDLINE | ID: mdl-33257853

ABSTRACT

Low-cost non-invasive diagnostic tools for staging the progression of non-alcoholic chronic liver failure from fatty liver disease to steatohepatitis are unavailable. Here, we describe the development and performance of a portable single-sided magnetic-resonance sensor for grading liver steatosis and fibrosis using diffusion-weighted multicomponent T2 relaxometry. In a diet-induced mouse model of non-alcoholic fatty liver disease, the sensor achieved overall accuracies of 92% (Cohen's kappa, κ = 0.89) and 86% (κ = 0.78) in the ex vivo grading of steatosis and fibrosis, respectively. Localization of the measurements in living mice through frequency-dependent spatial encoding led to an overall accuracy of 87% (κ = 0.81) for the grading of steatosis. In human liver samples, the sensor graded steatosis with an overall accuracy of 93% (κ = 0.88). The use of T2 relaxometry as a sensitive measure in fully automated low-cost magnetic-resonance devices at the point of care would alleviate the accessibility and cost limits of magnetic-resonance imaging for diagnosing liver disease and assessing liver health before liver transplantation.


Subject(s)
Fibrosis/pathology , Liver Cirrhosis/pathology , Liver/pathology , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/methods , Animals , Humans , Liver Transplantation/instrumentation , Mice , Mice, Inbred C57BL , Mobile Applications , Non-alcoholic Fatty Liver Disease/pathology , Point-of-Care Systems
5.
Sci Transl Med ; 11(502)2019 07 24.
Article in English | MEDLINE | ID: mdl-31341060

ABSTRACT

Magnetic resonance imaging (MRI) is a powerful diagnostic tool, but its use is restricted to the scanner suite. Here, we demonstrate that a bedside nuclear magnetic resonance (NMR) sensor can assess fluid status changes in individuals at a fraction of the time and cost compared to MRI. Our study recruited patients with end-stage renal disease (ESRD) who were regularly receiving hemodialysis treatments with intradialytic fluid removal as a model of volume overload and healthy controls as a model of euvolemia. Quantitative T 2 measurements of the lower leg of patients with ESRD immediately before and after dialysis were compared to those of euvolemic healthy controls using both a 0.28-T bedside single-voxel NMR sensor and a 1.5-T clinical MRI scanner. In the MRI data, we found that the first sign of fluid overload was an expanded muscle extracellular fluid (ECF) space, a finding undetectable at this stage using physical exam. A decrease in muscle ECF upon fluid removal was similarly detectable with both the bedside sensor and MRI. Bioimpedance measurements performed comparably to the bedside NMR sensor but were generally worse than MRI. These findings suggest that bedside NMR may be a useful method to identify fluid overload early in patients with ESRD and potentially other hypervolemic patient populations.


Subject(s)
Renal Dialysis/methods , Adolescent , Adult , Extracellular Fluid , Humans , Kidney Failure, Chronic/therapy , Magnetic Resonance Imaging , Models, Theoretical , Point-of-Care Systems , Young Adult
6.
Curr Top Membr ; 83: 77-106, 2019.
Article in English | MEDLINE | ID: mdl-31196611

ABSTRACT

We have previously identified the interaction between mammalian V-ATPase a2-subunit isoform and cytohesin-2 (CTH2) and studied molecular details of binding between these proteins. In particular, we found that six peptides derived from the N-terminal cytosolic domain of a2 subunit (a2N1-402) are involved in interaction with CTH2 (Merkulova, Bakulina, Thaker, Grüber, & Marshansky, 2010). However, the actual 3D binding interface was not determined in that study due to the lack of high-resolution structural information about a-subunits of V-ATPase. Here, using a combination of homology modeling and NMR analysis, we generated the structural model of complete a2N1-402 and uncovered the CTH2-binding interface. First, using the crystal-structure of the bacterial M. rubber Icyt-subunit of A-ATPase as a template (Srinivasan, Vyas, Baker, & Quiocho, 2011), we built a homology model of mammalian a2N1-352 fragment. Next, we combined it with the determined NMR structures of peptides a2N368-395 and a2N386-402 of the C-terminal section of a2N1-402. The complete molecular model of a2N1-402 revealed that six CTH2 interacting peptides are clustered in the distal and proximal lobe sub-domains of a2N1-402. Our data indicate that the proximal lobe sub-domain is the major interacting site with the Sec7 domain of first CTH2 protein, while the distal lobe sub-domain of a2N1-402 interacts with the PH-domain of second CTH2. Indeed, using Sec7/Arf-GEF activity assay we experimentally confirmed our model. The interface formed by peptides a2N1-17 and a2N35-49 is involved in specific interaction with Sec7 domain and regulation of GEF activity. These data are critical for understanding of the cross-talk between V-ATPase and CTH2 as well as for the rational drug design to regulate their function.


Subject(s)
Drug Design , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Amino Acid Sequence , Animals , Bacteria , Binding Sites , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism
7.
J Acoust Soc Am ; 142(4): EL401, 2017 10.
Article in English | MEDLINE | ID: mdl-29092550

ABSTRACT

This pilot study used acoustic speech analysis to monitor patients with heart failure (HF), which is characterized by increased intracardiac filling pressures and peripheral edema. HF-related edema in the vocal folds and lungs is hypothesized to affect phonation and speech respiration. Acoustic measures of vocal perturbation and speech breathing characteristics were computed from sustained vowels and speech passages recorded daily from ten patients with HF undergoing inpatient diuretic treatment. After treatment, patients displayed a higher proportion of automatically identified creaky voice, increased fundamental frequency, and decreased cepstral peak prominence variation, suggesting that speech biomarkers can be early indicators of HF.


Subject(s)
Acoustics , Edema/diagnosis , Heart Failure/complications , Phonation , Speech Acoustics , Speech Production Measurement , Vocal Cords/physiopathology , Voice Disorders/diagnosis , Voice Quality , Aged , Aged, 80 and over , Diuretics/therapeutic use , Edema/drug therapy , Edema/etiology , Edema/physiopathology , Female , Heart Failure/diagnosis , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Lung/physiopathology , Male , Middle Aged , Phonation/drug effects , Pilot Projects , Predictive Value of Tests , Respiration , Treatment Outcome , Vocal Cords/drug effects , Voice Disorders/drug therapy , Voice Disorders/etiology , Voice Disorders/physiopathology , Voice Quality/drug effects
8.
Nat Rev Nephrol ; 12(12): 738-753, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27795549

ABSTRACT

Treatment and management of kidney disease currently presents an enormous global burden, and the application of nanotechnology principles to renal disease therapy, although still at an early stage, has profound transformative potential. The increasing translation of nanomedicines to the clinic, alongside research efforts in tissue regeneration and organ-on-a-chip investigations, are likely to provide novel solutions to treat kidney diseases. Our understanding of renal anatomy and of how the biological and physico-chemical properties of nanomedicines (the combination of a nanocarrier and a drug) influence their interactions with renal tissues has improved dramatically. Tailoring of nanomedicines in terms of kidney retention and binding to key membranes and cell populations associated with renal diseases is now possible and greatly enhances their localization, tolerability, and efficacy. This Review outlines nanomedicine characteristics central to improved targeting of renal cells and highlights the prospects, challenges, and opportunities of nanotechnology-mediated therapies for renal diseases.


Subject(s)
Kidney Diseases/drug therapy , Nanoparticles/therapeutic use , Carcinoma, Renal Cell/drug therapy , Forecasting , Humans , Kidney/cytology , Kidney/metabolism , Kidney Neoplasms/drug therapy , Nanomedicine/trends
9.
Big Data ; 3(3): 203-208, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26487991

ABSTRACT

The next frontier in medicine involves better quantifying human traits, known as "phenotypes." Biological markers have been directly associated with disease risks, but poor measurement of behaviors such as diet and exercise limits our understanding of preventive measures. By joining together an uncommonly wide range of disciplines and expertise, the Kavli HUMAN Project will advance measurement of behavioral phenotypes, as well as environmental factors that impact behavior. By following the same individuals over time, KHP will liberate new understanding of dynamic links between behavioral phenotypes, disease, and the broader environment. As KHP advances understanding of the bio-behavioral complex, it will seed new approaches to the diagnosis, prevention, and treatment of human disease.

11.
J Biol Chem ; 288(8): 5896-913, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23288846

ABSTRACT

Previously, we reported an acidification-dependent interaction of the endosomal vacuolar H(+)-ATPase (V-ATPase) with cytohesin-2, a GDP/GTP exchange factor (GEF), suggesting that it functions as a pH-sensing receptor. Here, we have studied the molecular mechanism of signaling between the V-ATPase, cytohesin-2, and Arf GTP-binding proteins. We found that part of the N-terminal cytosolic tail of the V-ATPase a2-subunit (a2N), corresponding to its first 17 amino acids (a2N(1-17)), potently modulates the enzymatic GDP/GTP exchange activity of cytohesin-2. Moreover, this peptide strongly inhibits GEF activity via direct interaction with the Sec7 domain of cytohesin-2. The structure of a2N(1-17) and its amino acids Phe(5), Met(10), and Gln(14) involved in interaction with Sec7 domain were determined by NMR spectroscopy analysis. In silico docking experiments revealed that part of the V-ATPase formed by its a2N(1-17) epitope competes with the switch 2 region of Arf1 and Arf6 for binding to the Sec7 domain of cytohesin-2. The amino acid sequence alignment and GEF activity studies also uncovered the conserved character of signaling between all four (a1-a4) a-subunit isoforms of mammalian V-ATPase and cytohesin-2. Moreover, the conserved character of this phenomenon was also confirmed in experiments showing binding of mammalian cytohesin-2 to the intact yeast V-ATPase holo-complex. Thus, here we have uncovered an evolutionarily conserved function of the V-ATPase as a novel cytohesin-signaling receptor.


Subject(s)
GTPase-Activating Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , ADP-Ribosylation Factors/metabolism , Amino Acid Sequence , Animals , Circular Dichroism , DNA, Complementary/metabolism , Epitopes/chemistry , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy/methods , Mice , Microscopy, Confocal/methods , Molecular Sequence Data , Peptides/chemistry , Protein Isoforms , Protein Structure, Secondary , Rats , Recombinant Proteins/chemistry , Signal Transduction , Tryptophan/chemistry
12.
Am J Physiol Renal Physiol ; 304(5): F553-64, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23235478

ABSTRACT

Vasopressin (VP) binds to the vasopressin type 2 receptor (V2R) to trigger physiological effects including body fluid homeostasis and blood pressure regulation. Signaling is terminated by receptor downregulation involving clathrin-mediated endocytosis and V2R degradation. We report here that both native and epitope-tagged V2R are internalized from the plasma membrane of LLC-PK1 kidney epithelial cells in the presence of another ligand, transferrin (Tf). The presence of iron-saturated Tf (holo-Tf; 4 h) reduced V2R binding sites at the cell surface by up to 33% while iron-free (apo-Tf) had no effect. However, no change in green fluorescent protein-tagged V2R distribution was observed in the presence of bovine serum albumin, atrial natriuretic peptide, or ANG II. Conversely, holo-Tf did not induce the internalization of another G protein-coupled receptor, the parathyroid hormone receptor. In contrast to the effect of VP, Tf did not increase intracellular cAMP or modify aquaporin-2 distribution in these cells, although addition of VP and Tf together augmented VP-induced V2R internalization. Tf receptor coimmunoprecipitated with V2R, suggesting that they interact closely, which may explain the additive effect of VP and Tf on V2R endocytosis. Furthermore, Tf-induced V2R internalization was abolished in cells expressing a dominant negative dynamin (K44A) mutant, indicating the involvement of clathrin-coated pits. We conclude that Tf can induce heterologous downregulation of the V2R and this might desensitize VP target cells without activating downstream V2R signaling events. It also provides new insights into urine-concentrating defects observed in rat models of hemochromatosis.


Subject(s)
Down-Regulation/drug effects , Epithelial Cells/drug effects , Kidney/drug effects , Receptors, Vasopressin/metabolism , Transferrin/pharmacology , Vasopressins/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Kidney/cytology , Kidney/metabolism , Receptors, Vasopressin/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Swine
13.
Endocrinology ; 152(10): 3893-904, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21828182

ABSTRACT

The vasopressin receptor type 2 (V2R) is the major target of vasopressin (VP) in renal epithelial cells. Although it is known that VP induces V2R internalization, accumulation in the perinuclear area, and degradation, the V2R intracellular trafficking pathways remain elusive. We visualized this process by developing a new fluorescent VP analog tagged by tetramethylrhodamine (TMR)-[Lys-(PEG)(2)-Suc-TMR(8)]VP or (VP(TMR)). This ligand is fully functional as revealed by its high binding affinity toward V2R [(K(d)) =157 ± 52 nM] and ability to increase intracellular cAMP 32-fold. VP(TMR) induced V2R internalization in LLC-PK1 cells expressing either a FLAG-tagged receptor (FLAG-V2R) or V2R C-terminally tagged with green fluorescent protein (GFP) (V2R-GFP). After internalization, VP(TMR) and V2R-GFP colocalized in the perinuclear area, suggesting that the hormone and receptor traffic along the same pathway. VP(TMR) and V2R colocalized initially with the early endosome markers EEA1 and Rab5, and later with the recycling and late endosome markers Rab11 and Rab25. Epifluorescence microscopy of LLC-PK1 cells expressing GFP-tagged microtubules (MT) showed that VP(TMR)-containing vesicles travel along the MT network, and even remain attached to MT during the metaphase and anaphase of mitosis. Colchicine, a MT-depolymerizing agent, abolished perinuclear accumulation of VP(TMR), and Western blot analysis showed that VP-induced V2R-GFP degradation is markedly retarded, but not abolished, by colchicine (10 µM). We conclude that the new VP(TMR) ligand is suitable for dissecting V2R and VP internalization and trafficking in cells, and that V2R trafficking and down-regulation is an MT-dependent mechanism.


Subject(s)
Fluorescent Dyes , Microtubules/physiology , Receptors, Vasopressin/metabolism , Animals , Cyclic AMP/metabolism , LLC-PK1 Cells , Ligands , Protein Transport , Swine , Vasopressins/metabolism
14.
Am J Physiol Cell Physiol ; 300(6): C1442-55, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21307348

ABSTRACT

Previously, we demonstrated that the vacuolar-type H(+)-ATPase (V-ATPase) a2-subunit functions as an endosomal pH sensor that interacts with the ADP-ribosylation factor (Arf) guanine nucleotide exchange factor, ARNO. In the present study, we showed that ARNO directly interacts not only with the a2-subunit but with all a-isoforms (a1-a4) of the V-ATPase, indicating a widespread regulatory interaction between V-ATPase and Arf GTPases. We then extended our search for other ARNO effectors that may modulate V-ATPase-dependent vesicular trafficking events and actin cytoskeleton remodeling. Pull-down experiments using cytosol of mouse proximal tubule cells (MTCs) showed that ARNO interacts with aldolase, but not with other enzymes of the glycolytic pathway. Direct interaction of aldolase with the pleckstrin homology domain of ARNO was revealed by pull-down assays using recombinant proteins, and surface plasmon resonance revealed their high avidity interaction with a dissociation constant: K(D) = 2.84 × 10(-10) M. MTC cell fractionation revealed that aldolase is also associated with membranes of early endosomes. Functionally, aldolase knockdown in HeLa cells produced striking morphological changes accompanied by long filamentous cell protrusions and acidic vesicle redistribution. However, the 50% knockdown we achieved did not modulate the acidification capacity of endosomal/lysosomal compartments. Finally, a combination of small interfering RNA knockdown and overexpression revealed that the expression of aldolase is inversely correlated with gelsolin levels in HeLa cells. In summary, we have shown that aldolase forms a complex with ARNO/Arf6 and the V-ATPase and that it may contribute to remodeling of the actin cytoskeleton and/or the trafficking and redistribution of V-ATPase-dependent acidic compartments via a combination of protein-protein interaction and gene expression mechanisms.


Subject(s)
Cell Shape , Cytoplasmic Vesicles/metabolism , Fructose-Bisphosphate Aldolase/metabolism , GTPase-Activating Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Animals , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Endosomes/metabolism , Fructose-Bisphosphate Aldolase/genetics , GTPase-Activating Proteins/genetics , Gelsolin/genetics , Gelsolin/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Hydrogen-Ion Concentration , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Vacuolar Proton-Translocating ATPases/chemistry , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
15.
Traffic ; 10(3): 275-84, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19170982

ABSTRACT

The kidney regulates body fluid, ion and acid/base homeostasis through the interaction of a host of channels, transporters and pumps within specific tubule segments, specific cell types and specific plasma membrane domains. Furthermore, renal epithelial cells have adapted to function in an often unique and challenging environment that includes high medullary osmolality, acidic pHs, variable blood flow and constantly changing apical and basolateral 'bathing' solutions. In this review, we focus on selected protein trafficking events by which kidney epithelial cells regulate body fluid, ion and acid-base homeostasis in response to changes in physiological conditions. We discuss aquaporin 2 and G-protein-coupled receptors in fluid and ion balance, the vacuolar H(+)-adenosine triphosphatase (V-ATPase) and intercalated cells in acid/base regulation and acidification events in the proximal tubule degradation pathway. Finally, in view of its direct role in vesicle trafficking that we outline in this study, we propose that the V-ATPase itself should, under some circumstances, be considered a fourth category of vesicle 'coat' protein (COP), alongside clathrin, caveolin and COPs.


Subject(s)
Cell Physiological Phenomena , Epithelial Cells/metabolism , Kidney/metabolism , Signal Transduction , Animals , Biological Transport , Cytoplasmic Vesicles/metabolism , Humans
16.
Am J Physiol Renal Physiol ; 296(1): F87-97, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945824

ABSTRACT

The primary cilium of renal epithelial cells is a nonmotile sensory organelle, implicated in mechanosensory transduction signals. Recent studies from our laboratory indicate that renal epithelial primary cilia display abundant channel activity; however, the presence and functional role of specific membrane receptors in this organelle are heretofore unknown. Here, we determined a functional signaling pathway associated with the type 2 vasopressin receptor (V2R) in primary cilia of renal epithelial cells. Besides their normal localization on basolateral membrane, V2R was expressed in primary cilia of LLC-PK(1) renal epithelial cells. The presence of V2R in primary cilia was determined by spontaneous fluorescence of a V2R-gfp chimera and confirmed by immunocytochemical analysis of wild-type LLC-PK(1) cells stained with anti-V2R antibodies and in LLC-PK(1) cells overexpressing the V2R-Flag, with anti-Flag antibody. Ciliary V2R colocalized with adenylyl cyclase (AC) type V/VI in all cell types tested. Functional coupling of the receptors with AC was confirmed by measurement of cAMP production in isolated cilia and by testing AVP-induced cation-selective channel activity either in reconstituted lipid bilayers or subjected to membrane-attached patch clamping. Addition of either 10 microM AVP (trans) or forskolin (cis) in the presence but not the absence of ATP (1 mM, cis) stimulated cation-selective channel activity in ciliary membranes. This channel activity was reduced by addition of the PKA inhibitor PKI. The data provide the first demonstration for the presence of V2R in primary cilia of renal epithelial cells, and a functional cAMP-signaling pathway, which targets ciliary channel function and may help control the sensory function of the primary cilium.


Subject(s)
Cilia/metabolism , Epithelial Cells/metabolism , Kidney/metabolism , Receptors, Vasopressin/metabolism , Signal Transduction/physiology , Adenosine Triphosphate/pharmacology , Adenylyl Cyclases/metabolism , Animals , Colforsin/pharmacology , Cyclic AMP/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Kidney/cytology , Kidney/drug effects , LLC-PK1 Cells , Patch-Clamp Techniques , Swine , Vasopressins/pharmacology
17.
Rio de Janeiro; Saunders; 23 ed; 2009. xlii,1766 p. ilus, tab, graf.
Monography in Portuguese | LILACS | ID: lil-711367

Subject(s)
Humans , Internal Medicine
18.
Rio de Janeiro; Saunders; 23 ed; 2009. xlii,1689 p. ilus, tab, graf.
Monography in Portuguese | LILACS | ID: lil-711368

Subject(s)
Humans , Internal Medicine
19.
Rio de Janeiro; Elsevier Editora; 23 ed; 2009. 1766 p.
Monography in Portuguese | Sec. Munic. Saúde SP, AHM-Acervo, TATUAPE-Acervo | ID: sms-1894

Subject(s)
Internal Medicine
20.
Rio de Janeiro; Elsevier Editora; 23 ed; 2009. 3458 p.
Monography in Portuguese | Sec. Munic. Saúde SP, AHM-Acervo, TATUAPE-Acervo | ID: sms-1895

Subject(s)
Internal Medicine
SELECTION OF CITATIONS
SEARCH DETAIL
...