Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
J Neurosci Methods ; 325: 108326, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31265869

ABSTRACT

BACKGROUND: Parameterization of neuronal membrane conductance models relies on data acquired from current clamp (CC) or voltage clamp (VC) recordings. Although the CC approach provides key information on a neuron's firing properties, it is often difficult to disentangle the influence of multiple conductances that contribute to the excitation properties of a real neuron. Isolation of a single conductance using pharmacological agents or heterologous expression simplifies analysis but requires extensive VC evaluation to explore the complete state behavior of the channel of interest. NEW METHOD: We present an improved parameterization approach that uses data derived from dynamic action potential clamp (DAPC) recordings to extract conductance equation parameters. We demonstrate the utility of the approach by applying it to the standard Hodgkin-Huxley conductance model although other conductance models could be easily incorporated as well. RESULTS: Using a fully simulated setup we show that, with as few as five action potentials previously recorded in DAPC mode, sodium conductance equation parameters can be determined with average parameter errors of less than 4% while action potential firing accuracy approaches 100%. In real DAPC experiments, we show that by "training" our model with five or fewer action potentials, subsequent firing lasting for several seconds could be predicted with ˜96% mean firing rate accuracy and 94% temporal overlap accuracy. COMPARISON WITH EXISTING METHODS: Our DAPC-based approach surpasses the accuracy of VC-based approaches for extracting conductance equation parameters with a significantly reduced temporal overhead. CONCLUSION: DAPC-based approach will facilitate the rapid and systematic characterization of neuronal channelopathies.


Subject(s)
Electrophysiological Phenomena/physiology , Models, Biological , Neurons/physiology , Patch-Clamp Techniques/methods , Action Potentials/physiology , Animals , Humans
2.
Allergy ; 72(11): 1686-1693, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28376268

ABSTRACT

BACKGROUND: Aryl hydrocarbon receptor (AhR), an important regulator of immune responses, is activated by UVB irradiation in the skin. Langerhans cells (LC) in the epidermis of patients with atopic dermatitis (AD) carry the high-affinity receptor for IgE, FcεRI, and are crucially involved in the pathogenesis of AD by inducing inflammatory responses and regulating tolerogenic processes. OBJECTIVES: We investigated AhR and AhR repressor (AhRR) expression and functional consequences of AhR activation in human ex vivo skin cells and in in vitro-generated LC. METHODS: Epidermal cells from healthy skin were analyzed for their expression of AhR and AhRR. LC generated from CD34+ hematopoietic stem cells (CD34LC) were treated with the UV photoproduct and AhR ligand 6-formylindolo[3,2-b]carbazole (FICZ). Cell surface receptors, transcription factors, and the tolerogenic tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO) were analyzed using flow cytometry and quantitative PCR. RESULTS: Epidermal LC and CD34LC express AhR and AhRR. AhR was also found in keratinocytes, which lack AhRR. AhR activation of LC by FICZ caused downregulation of FcεRI in CD34LC without affecting their maturation. AhR-mediated regulation of FcεRI did not involve any known transcription factors related to this receptor. Furthermore, we could show upregulation of IDO mediated by AhR engagement. CONCLUSIONS: Our study shows that AhR activation by FICZ reduces FcεRI and upregulates IDO expression in LC. This AhR-mediated anti-inflammatory feedback mechanism may dampen the allergen-induced inflammation in AD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Dermatitis, Atopic/immunology , Feedback, Physiological/physiology , Inflammation/immunology , Langerhans Cells/metabolism , Receptors, Aryl Hydrocarbon/physiology , Adult , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Middle Aged , Receptors, IgE/metabolism , Repressor Proteins/metabolism
3.
Pflugers Arch ; 458(1): 39-52, 2009 May.
Article in English | MEDLINE | ID: mdl-18758808

ABSTRACT

Homeostasis of inorganic phosphate (P(i)) is primarily an affair of the kidneys. Reabsorption of the bulk of filtered P(i) occurs along the renal proximal tubule and is initiated by apically localized Na(+)-dependent P(i) cotransporters. Tubular P(i) reabsorption and therefore renal excretion of P(i) is controlled by a number of hormones, including phosphatonins, and metabolic factors. In most cases, regulation of P(i) reabsorption is achieved by changing the apical abundance of Na(+)/Pi cotransporters. The regulatory mechanisms involve various signaling pathways and a number of proteins that interact with Na(+)/P(i) cotransporters.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins/physiology , Acidosis/physiopathology , Animals , Cholecalciferol/physiology , Circadian Rhythm , Diet , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/physiology , Gene Expression Regulation , Glucuronidase/physiology , Gonadal Steroid Hormones/physiology , Homeostasis , Humans , Ion Transport , Klotho Proteins , Parathyroid Hormone/physiology , Phosphates/urine , Phosphoproteins/physiology , Potassium Deficiency/physiopathology , Signal Transduction , Sodium-Hydrogen Exchangers/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIc/physiology
4.
Handchir Mikrochir Plast Chir ; 41(1): 56-7, 2009 Feb.
Article in German | MEDLINE | ID: mdl-19085819

ABSTRACT

Surgery is generally thought to be the most successful treatment of flexor tendon sheath ganglions. Most previous investigators judge the blind percutaneous aspiration as unreliable. The purpose of this study was to investigate the feasibility and the success rate of percutaneous apiration guided by ultrasonography. We performed percutaneous aspiration under ultrasonographic control in 60 patients with flexor tendon sheath ganglions. Encouraged by our favourable results we can recommend this procedure instead of the blind percutaneous aspiration and even instead of surgery.


Subject(s)
Ganglion Cysts/diagnostic imaging , Ganglion Cysts/surgery , Hand/diagnostic imaging , Hand/surgery , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional , Suction/methods , Surgery, Computer-Assisted/methods , Ultrasonography, Interventional/methods , Humans , Postoperative Complications/diagnostic imaging , Postoperative Complications/etiology , Recurrence
5.
Kidney Int ; 70(9): 1548-59, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16955105

ABSTRACT

Members of the SLC34 gene family of solute carriers encode for three Na+-dependent phosphate (P i) cotransporter proteins, two of which (NaPi-IIa/SLC34A1 and NaPi-IIc/SLC34A3) control renal reabsorption of P i in the proximal tubule of mammals, whereas NaPi-IIb/SCLC34A2 mediates P i transport in organs other than the kidney. The P i transport mechanism has been extensively studied in heterologous expression systems and structure-function studies have begun to reveal the intricacies of the transport cycle at the molecular level using techniques such as cysteine scanning mutagenesis, and voltage clamp fluorometry. Moreover, sequence differences between the three types of cotransporters have been exploited to obtain information about the molecular determinants of hormonal sensitivity and electrogenicity. Renal handling of P i is regulated by hormonal and non-hormonal factors. Changes in urinary excretion of P i are almost invariably mirrored by changes in the apical expression of NaPi-IIa and NaPi-IIc in proximal tubules. Therefore, understanding the mechanisms that control the apical expression of NaPi-IIa and NaPi-IIc as well as their functional properties is critical to understanding how an organism achieves P i homeostasis.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIc/metabolism , Animals , Homeostasis , Humans , Mice , Parathyroid Hormone/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIc/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIc/genetics , Structure-Activity Relationship
6.
J Membr Biol ; 206(3): 227-38, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16456717

ABSTRACT

The type IIa Na(+)/P(i), cotransporter (NaPi-IIa) mediates electrogenic transport of three Na(+) and one divalent P(i) ion (and one net positive charge) across the cell membrane. Sequence comparison of electrogenic NaPi-IIa and IIb isoforms with the electroneutral NaPi-IIc isoform pointed to the third transmembrane domain (TMD-3) as a possibly significant determinant of substrate binding. To elucidate the role of TMD-3 in the topology and mechanism underlying NaPi-IIa function we subjected it to cysteine scanning mutagenesis. The constructs were expressed in Xenopus oocytes and P(i) transport kinetics were assayed by electrophysiology and radiotracer uptake. Cys substitution resulted in only marginally altered kinetics of P(i) transport in those mutants providing sufficient current for analysis. Only one site, at the extracellular end of TMD-3, appeared to be accessible to methanethiosulfonate reagents. However, additional mutations carried out at D224 (replaced by E, G or N) and N227 (replaced by D or Q) resulted in markedly altered voltage and substrate dependencies of the P(i)-dependent currents. Replacing Asp-224 (highly conserved in electrogenic a and b isoforms) with Gly (the residue found in the electroneutral c isoform) resulted in a mutant that mediated electroneutral Na(+)-dependent P(i) transport. Since electrogenic NaPi-II transports 3 Na(+)/transport cycle, whereas electroneutral NaPi-IIc only transports 2, we speculate that this loss of electrogenicity might result from the loss of one of the three Na(+) binding sites in NaPi-IIa.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/metabolism , Ion Channel Gating/physiology , Membrane Potentials/physiology , Oocytes/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/chemistry , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Cells, Cultured , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Structure-Activity Relationship , Xenopus laevis
7.
J Membr Biol ; 188(3): 227-36, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12181613

ABSTRACT

Analysis of rat and mouse proximal tubular brush-border membrane expression of the type IIa Na/P(i)-cotransporter provides evidence for its cleavage in the large extracellular loop (ECL-2). To study functional properties and membrane distribution of this split NaP(i)-IIa transporter we followed two strategies. In one strategy we expressed the transporter as two complementary parts (p40 and p45) in Xenopus laevis oocytes and as another strategy we cleaved the WT protein with trypsin. Both strategies resulted in a split NaP(i)-IIa protein located in the plasma membrane. The two domains were tied together by a disulfide bridge, most likely involving the cysteines 306 and 334. Surface expression of the NaP(i)-IIa fragments was dependent on the presence of both domains. If both domains were coexpressed, the transporter was functional and transport characteristics were identical to those of the WT-NaP(i)-IIa protein. Corresponding to this, the transporter cleaved by trypsin also retains its transport capacity. These data indicate that cleavage of the type IIa Na/P(i)-cotransporter at ECL-2 is compatible with its cotransport function.


Subject(s)
Gene Expression Regulation , Oocytes/cytology , Oocytes/physiology , Symporters/genetics , Symporters/metabolism , Trypsin/pharmacology , Amino Acid Sequence , Animals , Cell Line , Hydrogen-Ion Concentration , Membrane Potentials/physiology , Molecular Sequence Data , Oocytes/drug effects , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type II , Symporters/drug effects , Trypsin/genetics , Xenopus laevis
9.
J Membr Biol ; 187(2): 85-96, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12029367

ABSTRACT

The effects of the arginine-modifying reagent phenylglyoxal on the kinetics of the type IIa Na + /Pi cotransporter expressed in Xenopus, oocytes were studied by means of 32Pi uptake and electrophysiology. Phenylglyoxal incubation induced up to 60% loss of cotransport function but only marginally altered the Na+-leak. Substrate activation and pH dependency remained essentially unaltered, whereas the voltage dependency of Pi-induced change in electrogenic response was significantly reduced. Presteady-state charge movements were suppressed and the equilibrium charge distribution was shifted slightly towards hyperpolarizing potentials. Charge movements in the absence of external Na+ were also suppressed, which indicated that the empty-carrier kinetics were modified. These effects were incorporated into an ordered alternating access model for NaPi-IIa, whereby the arginine modification by phenylglyoxal was modeled as altered apparent electrical distances moved by mobile charges, together with a slower rate of translocation of the electroneutral, fully loaded carrier.


Subject(s)
Arginine/antagonists & inhibitors , Membrane Potentials/drug effects , Phenylglyoxal/pharmacology , Symporters/genetics , Symporters/metabolism , Animals , Arginine/genetics , Arginine/metabolism , Computer Simulation , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/physiology , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Membrane Potentials/physiology , Models, Biological , Oocytes/physiology , Patch-Clamp Techniques/methods , Phosphorus/metabolism , Reproducibility of Results , Sensitivity and Specificity , Sodium-Phosphate Cotransporter Proteins , Xenopus laevis
11.
Curr Opin Nephrol Hypertens ; 10(5): 555-61, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11496046

ABSTRACT

The type IIa sodium/inorganic phosphate co-transporter is the rate-limiting inorganic phosphate transport pathway in renal brush-border membranes, and is thus a key player in overall inorganic phosphate homeostasis. Its regulation is mostly associated with membrane retrieval/reinsertion (traffic) of the transport protein. This membrane traffic is controlled by specific 'motifs' at the level of the transporter protein and probably involves interacting proteins (e.g. for scaffolding, regulation or sorting). The intracellular signaling mechanisms (e.g. the involvement of kinases) and the involvement of the cytoskeleton are not yet understood. Hereditary alterations in renal inorganic phosphate handling can be associated with factors controlling the expression of the brush-border type IIa sodium/inorganic phosphate co-transporter.


Subject(s)
Kidney/metabolism , Phosphates/metabolism , Symporters/physiology , Absorption/physiology , Amino Acid Motifs/physiology , Animals , Humans , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa , Symporters/genetics
12.
Eur J Pharmacol ; 419(1): 1-7, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11348623

ABSTRACT

The functional pharmacology of receptors composed of the chicken brain GABA(A) receptor gamma 4 subunit and the mammalian GABA(A) receptor alpha 3 and beta2 subunits was studied by heterologous expression in Xenopus laevis oocytes using the two electrode voltage-clamp technique. GABA-evoked currents had an EC(50) of 180+/-30 microM. Responses were blocked by the competitive and non-competitive GABA(A) receptor antagonists, bicuculline methochloride and picrotoxin. Sodium pentobarbital reversibly potentiated the current several-fold, and Zn(2+) ions blocked the current with high potency (IC50=20 microM). GABA-evoked currents were potentiated by the benzodiazepine site full agonists flunitrazepam and triazolam and less by the partial agonists abecarnil and bretazenil. The inverse agonists methyl-beta-carboline-3-carboxylate (beta-CCM) and methyl 6,7-dimethoxy-4-ethyl-beta-carboline-3-carboxylate (DMCM) reduced the current. However, the imidazobenzodiazepine Ro 15-4513, which acts as an inverse agonist at mammalian alphaxbetaygamma2 GABA(A) receptors (where x=1, 2, 3 or 5, and y=1, 2 or 3), acted as a positive agonist at the gamma 4 subunit-containing receptors.


Subject(s)
GABA Modulators/pharmacology , Receptors, GABA-A/metabolism , Animals , Bicuculline/pharmacology , Brain Chemistry , Chickens , Dose-Response Relationship, Drug , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , Humans , Oocytes/metabolism , Patch-Clamp Techniques , Pentobarbital/pharmacology , Picrotoxin/pharmacology , Protein Subunits , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Recombinant Proteins/metabolism , Xenopus laevis , Zinc/chemistry , Zinc/pharmacology , gamma-Aminobutyric Acid/pharmacology
13.
J Gen Physiol ; 117(6): 533-46, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11382804

ABSTRACT

The transport function of the rat type IIa Na(+)/P(i) cotransporter is inhibited after binding the cysteine modifying reagent 2-aminoethyl methanethiosulfonate hydrobromide (MTSEA) to a cysteine residue substituted for a serine at position 460 (S460C) in the predicted third extracellular loop. This suggests that Ser-460 lies in a functionally important region of the protein. To establish a "structure-function" profile for the regions that flank Ser-460, the substituted cysteine accessibility method was employed. 18 mutants were constructed in which selected amino acids from Arg-437 through Leu-465 were substituted one by one for a cysteine. Mutants were expressed in Xenopus oocytes and transport function (cotransport and slippage) and kinetics were assayed by electrophysiology with or without prior treatment with cysteine modifying (methanethiosulfonate, MTS) reagents. Except for mutant I447C, mutants with cysteines at sites from Arg-437 through Thr-449, as well as Pro-461, were inactive. Cotransport function of mutants with Cys substitutions at sites Arg-462 through Leu-465 showed low sensitivity to MTS reagents. The preceding mutants (Cys substitution at Thr-451 to Ser-460) showed a periodic accessibility pattern that would be expected for an alpha-helix motif. Apart from loss of transport function, exposure of mutants A453C and A455C to MTSEA or 2-(triethylammonium)ethyl MTS bromide (MTSET) increased the uncoupled slippage current, which implicated the mutated sites in the leak pathway. Mutants from Ala-453 through Ala-459 showed less pH dependency, but generally stronger voltage dependency compared with the wild type, whereas those flanking this group were more sensitive to pH and showed weaker voltage dependence of cotransport mode kinetics. Our data indicate that parts of the third extracellular loop are involved in the translocation of the fully loaded carrier and show a membrane-associated alpha-helical structure.


Subject(s)
Carrier Proteins/genetics , Cysteine , Mutagenesis, Site-Directed , Symporters , Carrier Proteins/physiology , DNA, Complementary/genetics , Forecasting , Hydrogen-Ion Concentration , Patch-Clamp Techniques , Serine , Sodium-Phosphate Cotransporter Proteins , Structure-Activity Relationship
14.
J Membr Biol ; 180(1): 83-90, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11284206

ABSTRACT

We have combined a functional assay, surface labeling and immunocytochemical methods to compare total and surface-exposed renal type IIa Na+/Pi cotransporter protein. The wild-type type cotransporter (NaPi-IIa) and its functionally comparable cysteine mutant S460C were expressed in Xenopus oocytes. S460C contains a novel cysteine residue that, when modified by preincubation with methanethiosulfonate reagents, leads to complete suppression of cotransport function. This allowed surface labeling of the S460C using MTSEA-Biotin and confirmation by electrophysiology on the same cell. Protein was analyzed by Western blotting before and after streptavidin precipitation and by immunocytochemistry and immunogold electronmicroscopy. MTSEA-Biotin treatment resulted in a complete inhibition of S460C-mediated Na+/Pi-cotransport activity, which indicated that all transporters at the surface were biotinylated. After biotinylation, only a small fraction of total S460C protein was precipitated by streptavidin compared with the total amount of S460C protein detected in the lysate. Light- and electron-microscopy analysis of oocytes showed a large amount of WT and S460C transporter protein beneath the oocyte membrane. These data indicate that the apparent weak labeling efficiencies of surface-biotinylation-based assays of membrane proteins heterologously expressed in oocytes can be related to diminished incorporation of the protein in the oolemma.


Subject(s)
Carrier Proteins/metabolism , Oocytes/metabolism , Symporters , Animals , Biological Transport , Biotin/analogs & derivatives , Biotinylation , Carrier Proteins/genetics , Cell Membrane/metabolism , Cells, Cultured , Cloning, Molecular , Electrochemistry , Indicators and Reagents , Mesylates , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium-Phosphate Cotransporter Proteins , Xenopus laevis
15.
J Physiol ; 531(Pt 2): 383-91, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11230511

ABSTRACT

Type IIa and IIb Na+-Pi cotransporters are highly conserved proteins expressed in brush border membranes of proximal tubules and small intestine, respectively. The kinetics of IIa and IIb differ significantly: type IIb is saturated at lower concentrations of Na+ and Pi. To define the domain responsible for the difference in Na+ affinity we constructed several mouse IIa-IIb chimeras as well as site-directed mutagenized cotransporters. Pi uptake activity was determined after injection of cRNAs into Xenopus laevis oocytes. From the chimera experiments we concluded that the domain containing part of the second intracellular loop, the fifth transmembrane domain (TD) and part of the third extracellular loop determines the specific Na+ activation properties for both types of cotransporter. Within this domain only a few residues located in the fifth TD are not conserved between type IIa and IIb. Site-directed mutagenesis on non-conserved residues was performed. Substitution of F402 of IIa by the corresponding L418 from IIb yielded a cotransporter that behaved like the IIb. On the other hand, substitution of the specific L418 of IIb by the corresponding F402 of IIa produced a cotransporter with a Na+ activation similar to IIa. (Single letter amino acid nomenclature is used throughout the paper.) These data suggest that the specific Na+ activation properties exhibited by type IIa and type IIb Na+-Pi cotransporters are at least in part due to the presence of a specific amino acid (F402 in IIa, and L418 in IIb) within the fifth TD of the protein.


Subject(s)
Amino Acids/metabolism , Carrier Proteins/metabolism , Oocytes/metabolism , Sodium/metabolism , Symporters , Amino Acid Sequence/genetics , Amino Acid Substitution , Animals , Carrier Proteins/genetics , Chimera , Female , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Isoforms/genetics , Protein Structure, Tertiary/genetics , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type II , Sodium-Phosphate Cotransporter Proteins, Type IIa , Sodium-Phosphate Cotransporter Proteins, Type IIb , Xenopus laevis
16.
Clin Radiol ; 56(3): 221-4, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11247700

ABSTRACT

AIM: Post-arthroplasty knee pain is common and clinically it can be difficult to identify those patients with complications requiring active treatment. The aim of this study was to determine the usefulness of(99)Tc(m)-MDP bone scintigraphy. METHOD: A retrospective study of all patients having a(99)Tc(m)-MDP bone scintigram for a painful knee arthroplasty between 1993 and 1999 was performed. Bone scintigrams were classified as normal or abnormal by a single observer. The results of these investigations were correlated with clinical outcome. RESULTS: Seventy-five patients with painful knee arthroplasties were referred for investigation. A total of 80 bone scintigrams were performed. The average patient age was 66.2 years (42 female and 33 male). The mean time period between surgery and onset of knee pain was 3 years. A final clinical diagnosis based on arthroscopy, open surgery, and extended clinical follow-up was available for all patients. Forty-three (53.8%) of the scintigrams were normal and 37 (46.3%) abnormal. Two patients with a normal bone scintigram has loose prostheses. Thirteen patients with an abnormal study had normal prostheses on follow-up and these tended to be patients scanned less than a year after surgery. The sensitivity, specificity, positive predictive value and negative predictive value of an unequivocally normal or abnormal bone scintigram was 92.3, 75.9, 64.9 and 95.0%, respectively. The pattern of isotope uptake in the abnormal studies was not specific enough to reliably differentiate aseptic from septic loosening. CONCLUSION: Radionuclide bone scintigraphy is useful in the assessment of the painful knee arthroplasty. A negative bone scintigram is reassuring and makes loosening or infection unlikely.


Subject(s)
Arthroplasty, Replacement, Knee/adverse effects , Pain, Postoperative/diagnostic imaging , Aged , Arthroplasty, Replacement, Knee/methods , Female , Humans , Male , Pain, Postoperative/etiology , Predictive Value of Tests , Prosthesis Failure , Radionuclide Imaging , Radiopharmaceuticals , Retrospective Studies , Technetium Tc 99m Medronate , Treatment Outcome
17.
Physiol Rev ; 80(4): 1373-409, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11015617

ABSTRACT

Renal proximal tubular reabsorption of P(i) is a key element in overall P(i) homeostasis, and it involves a secondary active P(i) transport mechanism. Among the molecularly identified sodium-phosphate (Na/P(i)) cotransport systems a brush-border membrane type IIa Na-P(i) cotransporter is the key player in proximal tubular P(i) reabsorption. Physiological and pathophysiological alterations in renal P(i) reabsorption are related to altered brush-border membrane expression/content of the type IIa Na-P(i) cotransporter. Complex membrane retrieval/insertion mechanisms are involved in modulating transporter content in the brush-border membrane. In a tissue culture model (OK cells) expressing intrinsically the type IIa Na-P(i) cotransporter, the cellular cascades involved in "physiological/pathophysiological" control of P(i) reabsorption have been explored. As this cell model offers a "proximal tubular" environment, it is useful for characterization (in heterologous expression studies) of the cellular/molecular requirements for transport regulation. Finally, the oocyte expression system has permitted a thorough characterization of the transport characteristics and of structure/function relationships. Thus the cloning of the type IIa Na-P(i )cotransporter (in 1993) provided the tools to study renal brush-border membrane Na-P(i) cotransport function/regulation at the cellular/molecular level as well as at the organ level and led to an understanding of cellular mechanisms involved in control of proximal tubular P(i) handling and, thus, of overall P(i) homeostasis.


Subject(s)
Kidney Tubules, Proximal/metabolism , Phosphates/metabolism , Symporters , Absorption/physiology , Animals , Biological Transport, Active/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Electrophysiology , Gene Expression Regulation/physiology , Mice , Microvilli/metabolism , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary/genetics , Rabbits , Rats , Sequence Homology, Amino Acid , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa
18.
Exp Nephrol ; 8(6): 366-75, 2000.
Article in English | MEDLINE | ID: mdl-11014934

ABSTRACT

A key process in overall P(i)-homeostasis is renal proximal tubular reabsorption of inorganic phosphate (P(i)), which involves secondary active sodium/phosphate (Na(+)/P(i)) cotransport reabsorption at the brush border membrane. Among the two different molecularly identified Na(+)/P(i) cotransporters, the type-IIa Na(+)/P(i) cotransporter (NaPi-IIa) accounts for up to 70% of brush border membrane transport. Regulation of renal P(i) reabsorption centers around brush border membrane insertion and retrieval of transporter protein under the influence of hormonal and nonhormonal factors. Immunohistochemical and fluorescence techniques have provided new insights into the tissue distribution and the regulation processes. The intrinsic electrogenicity of NaPi-IIa, has allowed detailed studies of the transport kinetics of NaPi-IIa and, combined with mutagenesis methods, structure-function information at the protein level is emerging.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/physiology , Symporters , Amino Acid Sequence/genetics , Animals , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Down-Regulation , Humans , Kidney/metabolism , Kinetics , Molecular Sequence Data , Phosphate-Binding Proteins , Protein Structure, Secondary , RNA, Messenger/metabolism , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type IIa , Structure-Activity Relationship , Up-Regulation
19.
J Membr Biol ; 176(2): 133-41, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10926678

ABSTRACT

The rat renal Na/P(i) cotransporter type IIa (rat NaP(i) IIa) is a 637 amino acid protein containing 12 cysteine residues. We examined the effect of different cysteine modifying methanethiosulfonate (MTS)-reagents and the disulfide bond reducing agent tris(2-carboxyethyl)phosphine (TCEP) on the transport activity of wild-type and 12 single cysteine substitution mutants of rat NaPi IIa expressed in Xenopus laevis oocytes. The transport activity of the wild-type protein was resistant to three membrane impermeant MTS-reagents (MTSEA, MTSET and MTSES). In contrast, membrane permeant methyl methanethiosulfonate (MMTS) and TCEP inhibited the transport activity of both the wild-type, as well as all the single mutant proteins. This indicated the existence of more than one functionally important cysteine residue, not accessible extracellularly, and at least 2 disulfide bridges. To identify the disulfide bridges, three double mutants lacking 2 of the 3 cysteine residues predicted to be extracellular in different combinations were examined. This led to the identification of one disulfide bridge between C306 and C334; reconsideration of the topological model predictions suggested a second disulfide bridge between C225 and C520. Evaluation of a fourth double mutant indicated that at least one of two disulfide bridges (C306 and C334; C225 and C520) has to be formed to allow the surface expression of a functional cotransporter. A revised secondary structure is proposed which includes two partially repeated motifs that are connected by disulfide bridges formed between cysteine pairs C306-C334 and C225-C520.


Subject(s)
Carrier Proteins/chemistry , Cysteine/chemistry , Kidney Tubules, Proximal/chemistry , Symporters , Amino Acid Sequence , Amino Acid Substitution , Animals , Carrier Proteins/genetics , Cysteine/genetics , Disulfides , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Mesylates/pharmacology , Methyl Methanesulfonate/analogs & derivatives , Methyl Methanesulfonate/pharmacology , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/drug effects , Protein Structure, Tertiary , Rats , Reducing Agents/pharmacology , Serine/genetics , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type II , Sodium-Phosphate Cotransporter Proteins, Type IIa , Xenopus laevis
20.
Biophys J ; 79(1): 215-30, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10866949

ABSTRACT

In the kidney proximal tubule, acidification of the glomerular filtrate leads to an inhibition of inorganic phosphate (P(i)) reabsorption by type II Na(+)-coupled cotransporters (NaPi-II). As external pH also alters the divalent/monovalent P(i) ratio, it has been difficult to separate putative proton interactions with the cotransporter from direct titration of divalent P(i), the preferred species transported. To distinguish between these possibilities and identify pH-sensitive transitions in the cotransport cycle, the pH-dependent kinetics of two NaPi-II isoforms, expressed in Xenopus laevis oocytes, were investigated electrophysiologically. At -50 mV, both isoforms showed >70% suppression of electrogenic response for an external pH change from 8.0 to 6.2, not attributable to titration of divalent P(i). This was accompanied by a progressive removal of steady-state voltage dependence. The NaPi-II-related uncoupled slippage current was unaffected by a pH change from 7.4 to 6.2, with no shift in the reversal potential, which suggested that protons do not function as substrate. The voltage-dependence of pre-steady-state relaxations was shifted to depolarizing potentials in 100 mM and 0 mM Na(ext)(+) and two kinetic components were resolved, the slower of which was pH-dependent. The changes in kinetics are predicted by a model in which protons interact with the empty carrier and final Na(+) binding step.


Subject(s)
Acids/metabolism , Carrier Proteins/metabolism , Oocytes/metabolism , Symporters , Acids/pharmacology , Animals , Carrier Proteins/genetics , Cells, Cultured , Flounder , Hydrogen-Ion Concentration , Ion Transport/drug effects , Kinetics , Oocytes/cytology , Patch-Clamp Techniques , Phosphates/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Sodium/metabolism , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type II , Transfection , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...