Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Invest New Drugs ; 41(4): 596-605, 2023 08.
Article in English | MEDLINE | ID: mdl-37415001

ABSTRACT

Tepotinib is a highly selective, potent, mesenchymal-epithelial transition factor (MET) inhibitor, approved for the treatment of non-small cell lung cancer harboring MET exon 14 skipping alterations. The aims of this work were to investigate the potential for drug-drug interactions via cytochrome P450 (CYP) 3A4/5 or P-glycoprotein (P-gp) inhibition. In vitro studies were conducted in human liver microsomes, human hepatocyte cultures and Caco-2 cell monolayers to investigate whether tepotinib or its major metabolite (MSC2571109A) inhibited or induced CYP3A4/5 or inhibited P-gp. Two clinical studies were conducted to investigate the effect of multiple dose tepotinib (500 mg once daily orally) on the single dose pharmacokinetics of a sensitive CYP3A4 substrate (midazolam 7.5 mg orally) and a P-gp substrate (dabigatran etexilate 75 mg orally) in healthy participants. Tepotinib and MSC2571109A showed little evidence of direct or time-dependent CYP3A4/5 inhibition (IC50 > 15 µM) in vitro, although MSC2571109A did show mechanism-based CYP3A4/5 inhibition. Tepotinib did not induce CYP3A4/5 activity in vitro, although both tepotinib and MSC2571109A increased CYP3A4 mRNA. In clinical studies, tepotinib had no effect on the pharmacokinetics of midazolam or its metabolite 1'-hydroxymidazolam. Tepotinib increased dabigatran maximum concentration and area under the curve extrapolated to infinity by 38% and 51%, respectively. These changes were not considered to be clinically relevant. Tepotinib was considered safe and well tolerated in both studies. The potential of tepotinib to cause clinically relevant DDI with CYP3A4- or P-gp-dependent drugs at the clinical dose is considered low. Study 1 (midazolam): NCT03628339 (registered 14 August 2018). Study 2 (dabigatran): NCT03492437 (registered 10 April 2018).


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Cytochrome P-450 CYP3A/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Midazolam/pharmacokinetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dabigatran/pharmacokinetics , Caco-2 Cells , ATP Binding Cassette Transporter, Subfamily B , Drug Interactions
2.
Cancer Chemother Pharmacol ; 90(1): 53-69, 2022 07.
Article in English | MEDLINE | ID: mdl-35771259

ABSTRACT

PURPOSE: Tepotinib is a highly selective MET inhibitor approved for treatment of non-small cell lung cancer (NSCLC) harboring METex14 skipping alterations. Analyses presented herein evaluated the relationship between tepotinib exposure, and efficacy and safety outcomes. METHODS: Exposure-efficacy analyses included data from an ongoing phase 2 study (VISION) investigating 500 mg/day tepotinib in NSCLC harboring METex14 skipping alterations. Efficacy endpoints included objective response, duration of response, and progression-free survival. Exposure-safety analyses included data from VISION, plus four completed studies in advanced solid tumors/hepatocellular carcinoma (30-1400 mg). Safety endpoints included edema, serum albumin, creatinine, amylase, lipase, alanine aminotransferase, aspartate aminotransferase, and QT interval corrected using Fridericia's method (QTcF). RESULTS: Tepotinib exhibited flat exposure-efficacy relationships for all endpoints within the exposure range observed with 500 mg/day. Tepotinib also exhibited flat exposure-safety relationships for all endpoints within the exposure range observed with 30-1400 mg doses. Edema is the most frequently reported adverse event and the most frequent cause of tepotinib dose reductions and interruptions; however, the effect plateaued at low exposures. Concentration-QTc analyses using data from 30 to 1400 mg tepotinib resulted in the upper bounds of the 90% confidence interval being less than 10 ms for the mean exposures at the therapeutic (500 mg) and supratherapeutic (1000 mg) doses. CONCLUSIONS: These analyses provide important quantitative pharmacologic support for benefit/risk assessment of the 500 mg/day dosage of tepotinib as being appropriate for the treatment of NSCLC harboring METex14 skipping alterations. REGISTRATION NUMBERS: NCT01014936, NCT01832506, NCT01988493, NCT02115373, NCT02864992.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/pathology , Edema , Humans , Lung Neoplasms/pathology , Mutation , Piperidines , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-met/genetics , Pyridazines , Pyrimidines
3.
Cancer Chemother Pharmacol ; 89(5): 655-669, 2022 05.
Article in English | MEDLINE | ID: mdl-35385993

ABSTRACT

PURPOSE: Tepotinib is a highly selective, potent, mesenchymal-epithelial transition factor (MET) inhibitor, approved for the treatment of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping. Objectives of this population pharmacokinetic (PK) analysis were to evaluate the dose-exposure relationship of tepotinib and its major circulating metabolite, MSC2571109A, and to identify the intrinsic/extrinsic factors that are predictive of PK variability. METHODS: Data were included from 12 studies in patients with cancer and in healthy participants. A sequential modeling approach was used to analyze the parent and metabolite data, including covariate analyses. Potential associations between observed covariates and PK parameters were illustrated using bootstrap analysis-based forest plots. RESULTS: A two-compartment model with sequential zero- and first-order absorption, and a first-order elimination from the central compartment, best described the plasma PK of tepotinib in humans across the dose range of 30-1400 mg. The bioavailability of tepotinib was shown to be dose dependent, although bioavailability decreased primarily at doses above the therapeutic dose of 500 mg. The intrinsic factors of race, age, sex, body weight, mild/moderate hepatic impairment and mild/moderate renal impairment, along with the extrinsic factors of opioid analgesic and gefitinib intake, had no relevant effect on tepotinib PK. Tepotinib has a long effective half-life of ~ 32 h. CONCLUSIONS: Tepotinib shows dose proportionality up to at least the therapeutic dose, and time-independent clearance with a profile appropriate for once-daily dosing. None of the covariates identified had a clinically meaningful effect on tepotinib exposure or required dose adjustments.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Lung Neoplasms/drug therapy , Piperidines , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met , Pyridazines , Pyrimidines
4.
PLoS One ; 9(3): e92605, 2014.
Article in English | MEDLINE | ID: mdl-24663062

ABSTRACT

BACKGROUND: RGS9-deficient mice show drug-induced dyskinesia but normal locomotor activity under unchallenged conditions. RESULTS: Genes related to Ca2+ signaling and their functions were regulated in RGS9-deficient mice. CONCLUSION: Changes in Ca2+ signaling that compensate for RGS9 loss-of-function can explain the normal locomotor activity in RGS9-deficient mice under unchallenged conditions. SIGNIFICANCE: Identified signaling components may represent novel targets in antidyskinetic therapy. The long splice variant of the regulator of G-protein signaling 9 (RGS9-2) is enriched in striatal medium spiny neurons and dampens dopamine D2 receptor signaling. Lack of RGS9-2 can promote while its overexpression prevents drug-induced dyskinesia. Other animal models of drug-induced dyskinesia rather pointed towards overactivity of dopamine receptor-mediated signaling. To evaluate changes in signaling pathways mRNA expression levels were determined and compared in wild-type and RGS9-deficient mice. Unexpectedly, expression levels of dopamine receptors were unchanged in RGS9-deficient mice, while several genes related to Ca2+ signaling and long-term depression were differentially expressed when compared to wild type animals. Detailed investigations at the protein level revealed hyperphosphorylation of DARPP32 at Thr34 and of ERK1/2 in striata of RGS9-deficient mice. Whole cell patch clamp recordings showed that spontaneous synaptic events are increased (frequency and size) in RGS9-deficient mice while long-term depression is reduced in acute brain slices. These changes are compatible with a Ca2+-induced potentiation of dopamine receptor signaling which may contribute to the drug-induced dyskinesia in RGS9-deficient mice.


Subject(s)
Gene Expression Regulation , Neostriatum/metabolism , RGS Proteins/deficiency , Receptors, Dopamine D2/metabolism , Animals , Calcium Signaling , Cyclic AMP/metabolism , Dyskinesia, Drug-Induced/genetics , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Long-Term Synaptic Depression , Male , Mice , Neuronal Plasticity , Phosphorylation , Synapses/metabolism
5.
J Biol Chem ; 286(45): 39623-31, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21940628

ABSTRACT

Interaction and cross-talk of G-protein-coupled receptors (GPCRs) are of considerable interest because an increasing number of examples implicate a profound functional and physiological relevance of homo- or hetero-oligomeric GPCRs. The ghrelin (growth hormone secretagogue receptor (GHSR)) and melanocortin-3 (MC3R) receptors are both known to have orexigenic effects on the hypothalamic control of body weight. Because in vitro studies indicate heterodimerization of GHSR and MC3R, we investigated their functional interplay. Combined in situ hybridization and immunohistochemistry indicated that the vast majority of GHSR-expressing neurons in the arcuate nucleus also express MC3R. In vitro coexpression of MC3R and GHSR promoted enhanced melanocortin-induced intracellular cAMP accumulation compared with activation of MC3R in the absence of GHSR. In contrast, agonist-independent basal signaling activity and ghrelin-induced signaling of GHSR were impaired, most likely due to interaction with MC3R. By taking advantage of naturally occurring GHSR mutations and an inverse agonist for GHSR, we demonstrate that the observed enhanced MC3R signaling capability depends directly on the basal activity of GHSR. In conclusion, we demonstrate a paradigm-shifting example of GPCR heterodimerization allowing for mutually opposite functional influence of two hypothalamic receptors controlling body weight. We found that the agonist-independent active conformation of one GPCR can determine the signaling modalities of another receptor in a heterodimer. Our discovery also implies that mutations within one of two interacting receptors might affect both receptors and different pathways simultaneously. These findings uncover mechanisms of important relevance for pharmacological targeting of GPCR in general and hypothalamic body weight regulation in particular.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Multimerization/physiology , Receptor, Melanocortin, Type 3/metabolism , Receptors, Ghrelin/metabolism , Signal Transduction/physiology , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP/genetics , Cyclic AMP/metabolism , Gene Expression Regulation/physiology , Ghrelin/genetics , Ghrelin/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/genetics , Receptors, Ghrelin/agonists , Receptors, Ghrelin/genetics
6.
Mol Cell Endocrinol ; 331(2): 170-8, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20708652

ABSTRACT

Once introduced into the very early eukaryotic blueprint, seven-transmembrane receptors soon became the central and versatile components of the evolutionary highly successful G protein-coupled transmembrane signaling mechanism. In contrast to all other components of this signal transduction pathway, G protein-coupled receptors (GPCR) evolved in various structural families, eventually comprising hundreds of members in vertebrate genomes. Their functional diversity is in contrast to the conserved transmembrane core and the invariant set of intracellular signaling mechanisms, and it may be the interplay of these properties that is the key to the evolutionary success of GPCR. The GPCR repertoires retrieved from extant vertebrate genomes are the recent endpoints of this long evolutionary process. But the shaping of the fine structure and the repertoire of GPCR is still ongoing, and signatures of recent selection acting on GPCR genes can be made visible by modern population genetic methods. The very dynamic evolution of GPCR can be analyzed from different perspectives: at the levels of sequence comparisons between species from different families, orders and classes, and at the level of populations within a species. Here, we summarize the main conclusions from studies at these different levels with a specific focus on the more recent evolutionary dynamics of GPCR.


Subject(s)
Evolution, Molecular , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Conserved Sequence , Genome , Humans , Protein Conformation , Sequence Analysis, Protein , Signal Transduction
7.
PLoS One ; 5(5): e10580, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20485495

ABSTRACT

Several Ca(2+)-permeable channels, including the non-selective cation channel TRPV4, are subject to Ca(2+)-dependent facilitation. Although it has been clearly demonstrated in functional experiments that calmodulin (CaM) binding to intracellular domains of TRP channels is involved in this process, the molecular mechanism remains elusive. In this study, we provide experimental evidence for a comprehensive molecular model that explains Ca(2+)-dependent facilitation of TRPV4. In the resting state, an intracellular domain from the channel N terminus forms an autoinhibitory complex with a C-terminal domain that includes a high-affinity CaM binding site. CaM binding, secondary to rises in intracellular Ca(2+), displaces the N-terminal domain which may then form a homologous interaction with an identical domain from a second subunit. This represents a novel potentiation mechanism that may also be relevant in other Ca(2+)-permeable channels.


Subject(s)
Calcium/pharmacology , Ion Channel Gating/drug effects , TRPV Cation Channels/chemistry , TRPV Cation Channels/metabolism , Amino Acid Sequence , Binding Sites , Biological Assay , Calmodulin/metabolism , Cell Line , Humans , Molecular Sequence Data , Mutation/genetics , Peptides/metabolism , Protein Binding/drug effects , Protein Multimerization/drug effects , Protein Structure, Tertiary , Structure-Activity Relationship
8.
BMC Biotechnol ; 9: 46, 2009 May 18.
Article in English | MEDLINE | ID: mdl-19450256

ABSTRACT

BACKGROUND: Controlling enzyme activity by ligand binding to a regulatory domain of choice may have many applications e.g. as biosensors and as tools in regulating cellular functions. However, until now only a small number of ligand-binding domains have been successfully linked to enzyme activity. G protein-coupled receptors (GPCR) are capable of recognizing an extraordinary structural variety of extracellular signals including inorganic and organic molecules. Ligand binding to GPCR results in conformational changes involving the transmembrane helices. Here, we assessed whether ligand-induced conformational changes within the GPCR helix bundle can be utilized to control the activity of an integrated enzyme. RESULTS: As a proof of principle, we inserted the luciferase amino acid sequence into the third intracellular loop of the M3 muscarinic acetylcholine receptor. This fusion protein retained both receptor and enzyme function. Receptor blockers slightly but significantly reduced enzyme activity. By successive deletion mutagenesis the enzyme activity was optimally coupled to ligand-induced conformational helix movements. CONCLUSION: Our results demonstrate that in engineered GPCR-enzyme chimeras, intracellular enzyme activity can be directly controlled by a GPCR serving as the extracellular ligand-binding domain.


Subject(s)
Luciferases, Firefly/metabolism , Receptors, Muscarinic/metabolism , Recombinant Fusion Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Ligands , Mutagenesis, Insertional , Protein Binding , Protein Engineering , Protein Structure, Secondary , Rats , Transfection
9.
Am J Physiol Renal Physiol ; 295(4): F1177-90, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18715941

ABSTRACT

Polyuria, hypernatremia, and hypovolemia are the major clinical signs of inherited nephrogenic diabetes insipidus (NDI). Hypernatremia is commonly considered a secondary sign caused by the net loss of water due to insufficient insertion of aquaporin-2 water channels into the apical membrane of the collecting duct cells. In the present study, we employed transcriptome-wide expression analysis to study gene expression in V2 vasopressin receptor (Avpr2)-deficient mice, an animal model for X-linked NDI. Gene expression changes in NDI mice indicate increased proximal tubular sodium reabsorption. Expression of several key genes including Na+-K+-ATPase and carbonic anhydrases was increased at the mRNA levels and accompanied by enhanced enzyme activities. In addition, altered expression was also observed for components of the eicosanoid and thyroid hormone pathways, including cyclooxygenases and deiodinases, in both kidney and hypothalamus. These effects are likely to contribute to the clinical NDI phenotype. Finally, our data highlight the involvement of the renin-angiotensin-aldosterone system in NDI pathophysiology and provide clues to explain the effectiveness of diuretics and indomethacin in the treatment of NDI.


Subject(s)
Diabetes Insipidus, Nephrogenic/physiopathology , Hypothalamus/physiology , Kidney Tubules, Proximal/physiology , Oligonucleotide Array Sequence Analysis , Receptors, Vasopressin/genetics , Water-Electrolyte Balance/physiology , Animals , Diabetes Insipidus, Nephrogenic/metabolism , Disease Models, Animal , Female , Gene Expression/physiology , Gene Expression Profiling , Homeostasis/physiology , Hypernatremia/metabolism , Hypernatremia/physiopathology , Mice , Receptors, Vasopressin/deficiency , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism
10.
PLoS One ; 3(5): e2209, 2008 May 21.
Article in English | MEDLINE | ID: mdl-18493316

ABSTRACT

Genome-wide scans for positive selection in humans provide a promising approach to establish links between genetic variants and adaptive phenotypes. From this approach, lists of hundreds of candidate genomic regions for positive selection have been assembled. These candidate regions are expected to contain variants that contribute to adaptive phenotypes, but few of these regions have been associated with phenotypic effects. Here we present evidence that a derived nonsynonymous substitution (370A) in EDAR, a gene involved in ectodermal development, was driven to high frequency in East Asia by positive selection prior to 10,000 years ago. With an in vitro transfection assay, we demonstrate that 370A enhances NF-kappaB activity. Our results suggest that 370A is a positively selected functional genetic variant that underlies an adaptive human phenotype.


Subject(s)
Alleles , Edar Receptor/genetics , NF-kappa B/metabolism , Selection, Genetic , Asia , Humans
11.
Eur J Cell Biol ; 87(5): 311-23, 2008 May.
Article in English | MEDLINE | ID: mdl-18387691

ABSTRACT

The protease cathepsin D (Cath D) and its proteolytically inactive proform, procathepsin D (ProCath D), turned out to be multifunctional within and outside the cell. Elevated levels of ProCath D occur in malignant tumors and in organs under chronic inflammation. One important source for this increase of ProCath D might be endothelial cells. Here we examined the expression of Cath D in the human endothelial cell line EA.hy 926 and in primary endothelial cells isolated from human umbilical cord veins (HUVEC). After serum-free incubation with or without human interferon-gamma (hIFN-gamma) and/or human tumor necrosis factor-alpha (hTNF-alpha) immature and mature Cath D forms were examined in cell extracts and in cell-conditioned medium concentrates by Western blotting. Lysates of EA.hy 926 cells as well as of HUVEC contained active Cath D as two-chain form, but only negligible amounts of ProCath D and Cath D intermediates. Yet both endothelial cell cultures accumulated ProCath D in their conditioned media in the absence of any stimulus. The treatment with hIFN-gamma and/or hTNF-alpha had little effect on intracellular levels of Cath D, whereas the cytokine stimulation increased the extracellular presence of ProCath D in both endothelial cell cultures. The extracellular increase of ProCath D was not related to induction of apoptosis, as validated by cleaved caspase-3 in cell lysates. Acidification of cytokine-treated media converted ProCath D into Cath D, which was associated with cathepsin-like activity using a fluorogenic substrate-linked assay. We conclude, in vitro, endothelial cells are a cytokine-dependent source for extracellular ProCath D.


Subject(s)
Cathepsin D/metabolism , Endothelial Cells/drug effects , Enzyme Precursors/metabolism , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Aspartic Acid Endopeptidases/metabolism , Cathepsin D/genetics , Cell Line , Cells, Cultured , Culture Media , Endothelial Cells/cytology , Enzyme Precursors/genetics , Extracellular Space , Humans , Hydrogen-Ion Concentration
12.
PLoS One ; 3(2): e1686, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18301763

ABSTRACT

We identified and examined a candidate gene for local directional selection in Europeans, TRPV6, and conclude that selection has acted on standing genetic variation at this locus, creating parallel soft sweep events in humans. A novel modification of the extended haplotype homozygosity (EHH) test was utilized, which compares EHH for a single allele across populations, to investigate the signature of selection at TRPV6 and neighboring linked loci in published data sets for Europeans, Asians and African-Americans, as well as in newly-obtained sequence data for additional populations. We find that all non-African populations carry a signature of selection on the same haplotype at the TRPV6 locus. The selective footprints, however, are significantly differentiated between non-African populations and estimated to be younger than an ancestral population of non-Africans. The possibility of a single selection event occurring in an ancestral population of non-Africans was tested by simulations and rejected. The putatively-selected TRPV6 haplotype contains three candidate sites for functional differences, namely derived non-synonymous substitutions C157R, M378V and M681T. Potential functional differences between the ancestral and derived TRPV6 proteins were investigated by cloning the ancestral and derived forms, transfecting cell lines, and carrying out electrophysiology experiments via patch clamp analysis. No statistically-significant differences in biophysical channel function were found, although one property of the protein, namely Ca(2+) dependent inactivation, may show functionally relevant differences between the ancestral and derived forms. Although the reason for selection on this locus remains elusive, this is the first demonstration of a widespread parallel selection event acting on standing genetic variation in humans, and highlights the utility of between population EHH statistics.


Subject(s)
Calcium Channels/genetics , Genetics, Population , Selection, Genetic , TRPV Cation Channels/genetics , White People/genetics , Amino Acid Substitution , Calcium Channels/physiology , Electrophysiology , Genetic Variation , Humans , Patch-Clamp Techniques , Racial Groups/genetics , TRPV Cation Channels/physiology
13.
J Physiol ; 581(Pt 2): 619-29, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17347272

ABSTRACT

The range of actions of the second messenger Ca(2+) is a key determinant of neuronal excitability and plasticity. For dendritic spines, there is on-going debate regarding how diffusional efflux of Ca(2+) affects spine signalling. However, the consequences of spino-dendritic coupling for dendritic Ca(2+) homeostasis and downstream signalling cascades have not been explored to date. We addressed this question by four-dimensional computer simulations, which were based on Ca(2+)-imaging data from mice that either express or lack distinct endogenous Ca(2+)-binding proteins. Our simulations revealed that single active spines do not affect dendritic Ca(2+) signalling. Neighbouring, coactive spines, however, induce sizeable increases in dendritic [Ca(2+)](i) when they process slow synaptic Ca(2+) signals, such as those implicated in the induction of long-term plasticity. This spino-dendritic coupling is mediated by buffered diffusion, specifically by diffusing calbindin-bound Ca(2+). This represents a central mechanism for activating calmodulin in dendritic shafts and therefore a novel form of signal integration in spiny dendrites.


Subject(s)
Calcium Signaling , Calcium-Binding Proteins/metabolism , Dendritic Spines/physiology , Neuronal Plasticity , Purkinje Cells/physiology , Synaptic Transmission , Animals , Calbindins , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calmodulin/metabolism , Computer Simulation , Dendritic Spines/metabolism , Diffusion , Kinetics , Mice , Mice, Knockout , Models, Neurological , Parvalbumins/metabolism , Purkinje Cells/metabolism , S100 Calcium Binding Protein G/metabolism
14.
Expert Rev Endocrinol Metab ; 1(6): 727-741, 2006 Nov.
Article in English | MEDLINE | ID: mdl-30754158

ABSTRACT

Maintenance of water and electrolyte homeostasis is central to mammalian survival and, therefore, under stringent hormonal control. Water homeostasis is achieved by balancing fluid intake with water excretion, governed by the antidiuretic action of arginine vasopressin. Arginine vasopressin stimulation of renal V2 vasopressin receptors in the basolateral membrane of principal cells induces aquaporin-2-mediated water reabsorption in the kidney. The importance of this system is apparent when mutations inactivate V2 vasopressin receptors and aquaporin-2 and cause the clinical phenotype of nephrogenic diabetes insipidus. To date, over 190 mutations in the V2 vasopressin receptors gene (AVPR2) and approximately 38 mutations in the aquaporin-2 gene have been identified in patients with inherited nephrogenic diabetes insipidus. Extensive in vitro expression and mutagenesis studies of V2 vasopressin receptors and aquaporin-2 have provided detailed insights into the molecular mechanisms of G-protein-coupled receptor and water channel dysfunction per se. Targeted deletions of AVPR2 and AQP2 in mice have extended the knowledge of nephrogenic diabetes insipidus pathophysiology and have stimulated testing of old and new ideas to therapeutically restore normal kidney function in animal models and patients with this disease. In this review, we summarize the current knowledge relevant to understand the molecular basis of inherited nephrogenic diabetes insipidus forms and the rationales for the current pharmacological treatment of patients with this illness.

15.
J Biol Chem ; 278(29): 26541-9, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12724311

ABSTRACT

Most Ca2+-permeable ion channels are inhibited by increases in the intracellular Ca2+ concentration ([Ca2+]i), thus preventing potentially deleterious rises in [Ca2+]i. In this study, we demonstrate that currents through the osmo-, heat- and phorbol ester-sensitive, Ca2+-permeable nonselective cation channel TRPV4 are potentiated by intracellular Ca2+. Spontaneous TRPV4 currents and currents stimulated by hypotonic solutions or phorbol esters were reduced strongly at all potentials in the absence of extracellular Ca2+. The other permeant divalent cations Ba2+ and Sr2+ were less effective than Ca2+ in supporting channel activity. An intracellular site of Ca2+ action was supported by the parallel decrease in spontaneous currents and [Ca2+]i on removal of extracellular Ca2+ and the ability of Ca2+ release from intracellular stores to restore TRPV4 activity in the absence of extracellular Ca2+. During TRPV4 activation by hypotonic solutions or phorbol esters, Ca2+ entry through the channel increased the rate and extent of channel activation. Currents were also potentiated by ionomycin in the presence of extracellular Ca2+. Ca2+-dependent potentiation of TRPV4 was often followed by inhibition. By mutagenesis, we localized the structural determinant of Ca2+-dependent potentiation to an intracellular, C-terminal calmodulin binding domain. This domain binds calmodulin in a Ca2+-dependent manner. TRPV4 mutants that did not bind calmodulin lacked Ca2+-dependent potentiation. We conclude that TRPV4 activity is tightly controlled by intracellular Ca2+. Ca2+ entry increases both the rate and extent of channel activation by a calmodulin-dependent mechanism. Excessive increases in [Ca2+]i via TRPV4 are prevented by a Ca2+-dependent negative feedback mechanism.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Cation Transport Proteins , Ion Channels/chemistry , Ion Channels/metabolism , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Binding Sites/genetics , Calcium/pharmacology , Cations, Divalent/metabolism , Cell Line , Cloning, Molecular , DNA/genetics , Feedback , Humans , In Vitro Techniques , Ion Channels/drug effects , Ion Channels/genetics , Ionomycin/pharmacology , Ionophores/pharmacology , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , TRPV Cation Channels
16.
J Biol Chem ; 278(6): 3562-71, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12456670

ABSTRACT

Mammalian members of the classical transient receptor potential channel (TRPC) subfamily (TRPC1-7) are Ca(2+)-permeable cation channels involved in receptor-mediated increases in intracellular Ca(2+). Unlike most other TRP-related channels, which are inhibited by La(3+) and Gd(3+), currents through TRPC4 and TRPC5 are potentiated by La(3+). Because these differential effects of lanthanides on TRPC subtypes may be useful for clarifying the role of different TRPCs in native tissues, we characterized the potentiating effect in detail and localized the molecular determinants of potentiation by mutagenesis. Whole cell currents through TRPC5 were reversibly potentiated by micromolar concentrations of La(3+) or Gd(3+), whereas millimolar concentrations were inhibitory. By comparison, TRPC6 was blocked to a similar extent by La(3+) or Gd(3+) at micromolar concentrations and showed no potentiation. Dual effects of lanthanides on TRPC5 were also observed in outside-out patches. Even at micromolar concentrations, the single channel conductance was reduced by La(3+), but reduction in conductance was accompanied by a dramatic increase in channel open probability, leading to larger integral currents. Neutralization of the negatively charged amino acids Glu(543) and Glu(595)/Glu(598), situated close to the extracellular mouth of the channel pore, resulted in a loss of potentiation, and, for Glu(595)/Glu(598) in a modification of channel inhibition. We conclude that in the micromolar range, the lanthanide ions La(3+) and Gd(3+) have opposite effects on whole cell currents through TRPC5 and TRPC6 channels. The potentiation of TRPC4 and TRPC5 by micromolar La(3+) at extracellular sites close to the pore mouth is a promising tool for identifying the involvement of these isoforms in receptor-operated cation conductances of native cells.


Subject(s)
Calcium Channels/physiology , Cation Transport Proteins , Egtazic Acid/analogs & derivatives , Ion Channels/drug effects , Lanthanoid Series Elements/pharmacology , Animals , Base Sequence , Cell Line , DNA Primers , Egtazic Acid/pharmacology , Humans , Mice , TRPC Cation Channels , TRPC6 Cation Channel
17.
Am J Physiol Cell Physiol ; 282(2): C347-59, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11788346

ABSTRACT

To investigate the possible role of members of the mammalian transient receptor potential (TRP) channel family (TRPC1-7) in vasoconstrictor-induced Ca(2+) entry in vascular smooth muscle cells, we studied [Arg(8)]-vasopressin (AVP)-activated channels in A7r5 aortic smooth muscle cells. AVP induced an increase in free cytosolic Ca(2+) concentration ([Ca(2+)](i)) consisting of Ca(2+) release and Ca(2+) influx. Whole cell recordings revealed the activation of a nonselective cation current with a doubly rectifying current-voltage relation strikingly similar to those described for some heterologously expressed TRPC isoforms. The current was also stimulated by direct activation of G proteins as well as by activation of the phospholipase Cgamma-coupled platelet-derived growth factor receptor. Currents were not activated by store depletion or increased [Ca(2+)](i). Application of 1-oleoyl-2-acetyl-sn-glycerol stimulated the current independently of protein kinase C, a characteristic property of the TRPC3/6/7 subfamily. Like TRPC6-mediated currents, cation currents in A7r5 cells were increased by flufenamate. Northern hybridization revealed mRNA coding for TRPC1 and TRPC6. We therefore suggest that TRPC6 is a molecular component of receptor-stimulated Ca(2+)-permeable cation channels in A7r5 smooth muscle cells.


Subject(s)
Calcium Channels/physiology , Cations/metabolism , Ion Channels/physiology , Muscle, Smooth, Vascular/physiology , Receptors, Cell Surface/physiology , Animals , Arginine Vasopressin/pharmacology , Blotting, Northern , Calcium/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Calcium Signaling , Cations/chemistry , Cations/pharmacology , Cell Line , Electric Conductivity , Electrophysiology , Flufenamic Acid/pharmacology , Imidazoles/pharmacology , Ion Channels/drug effects , Muscle, Smooth, Vascular/cytology , Osmolar Concentration , RNA, Messenger/metabolism , Rats , TRPC Cation Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...