Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Naunyn Schmiedebergs Arch Pharmacol ; 391(8): 833-846, 2018 08.
Article in English | MEDLINE | ID: mdl-29736621

ABSTRACT

Transient receptor potential (TRP) channels represent a superfamily of unselective cation channels that are subdivided into seven subfamilies based on their sequence homology and differences in gating and functional properties. Little is known about the molecular mechanisms of TRP channel regulation, particularly of the "canonical" TRP (TRPC) subfamily and their activation by polyunsaturated fatty acids (PUFAs). Here, we analyzed the structure-function relationship of Drosophila fruit fly TRPC channels. The primary aim was to uncover the molecular basis of PUFA sensitivity of Drosophila TRP-like (TRPL) and TRPgamma channels. Amino acid (aa) sequence alignment of the three Drosophila TRPC channels revealed 50 aa residues highly conserved in PUFA-sensitive TRPL and TRPgamma channels but not in the PUFA-insensitive TRP channel. Substitution of respective aa in TRPL by corresponding aa of TRP identified 18 residues that are necessary for PUFA-mediated activation of TRPL. Most aa positions are located within a stretch comprising transmembrane domains S2-S4, whereas six aa positions have been assigned to the proximal cytosolic C-terminus. Interestingly, residues I465 and S471 are required for activation by 5,8,11,14-eicosatetraynoic acid (ETYA) but not 5,8,11-eicosatriynoic acid (ETI). As proof of concept, we generated a PUFA-sensitive TRP channel by exchanging the corresponding aa from TRPL to TRP. Our study demonstrates a specific aa pattern in the transmembrane domains S2-S4 and the proximal C-terminus essential for TRP channel activation by PUFAs.


Subject(s)
Drosophila Proteins/physiology , Fatty Acids, Unsaturated/pharmacology , Transient Receptor Potential Channels/physiology , Calcium/physiology , Drosophila Proteins/genetics , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Transient Receptor Potential Channels/genetics
2.
Angew Chem Int Ed Engl ; 56(21): 5750-5754, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28429411

ABSTRACT

G-protein-coupled-receptors (GPCRs) are of fundamental importance for signal transduction through cell membranes. This makes them important drug targets, but structure-based drug design (SBDD) is still hampered by the limitations for structure determination of unmodified GPCRs. We show that the interligand NOEs for pharmacophore mapping (INPHARMA) method can provide valuable information on ligand poses inside the binding site of the unmodified human A2A adenosine receptor reconstituted in nanodiscs. By comparing experimental INPHARMA spectra with back-calculated spectra based on ligand poses obtained from molecular dynamics simulations, a complex structure for A2A R with the low-affinity ligand 3-pyrrolidin-1-ylquinoxalin-2-amine was determined based on the X-ray structure of ligand ZM-241,358 in complex with a modified A2A R.


Subject(s)
Receptor, Adenosine A2A/chemistry , Receptors, G-Protein-Coupled/chemistry , Binding Sites , Humans , Ligands , Lipids , Magnetic Resonance Spectroscopy , Molecular Structure , Protein Binding , Protein Domains
3.
Neurobiol Learn Mem ; 136: 1-12, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27633915

ABSTRACT

Recently we have shown that capsaicin attenuates the strength of LTP in the lateral amygdala (LA) and demonstrated that this effect is mediated by the transient receptor potential (TRP) channel TRPV1. Here we further show that capsaicin, which is thought to act primarily through TRPV1, modifies long term depression (LTD) in the LA. Yet the application of various TRPV1 antagonists does not reverse this effect and it remains in TRPV1-deficient mice. In addition, voltage gated calcium channels, nitric oxide and CB1 receptors are not involved. Using pharmacology and TRPM1-/- mice, our electrophysiological data indicate that capsaicin-induced activation of TRPM1 channels contribute to the induction of LA-LTD. Whereas LA-LTD in general depends on the acitvation of NMDA receptors- and group II metabotropic glutamate receptors (mGluR), the modifying effect of capsaicin on LA-LTD via TRPM1 appears to be specifically mediated by group I mGluRs and in interaction with another member of the TRP family, TRPC5. Additionally, intact GABAergic transmission is required for the capsaicin-effect to take place. This is the first documentation that beside their function in the retina TRPM1 proteins are expressed in the brain and have a functional relevance in modifying synaptic plasticity.


Subject(s)
Basolateral Nuclear Complex/drug effects , Capsaicin/pharmacology , Long-Term Synaptic Depression/drug effects , Sensory System Agents/pharmacology , TRPC Cation Channels/drug effects , TRPM Cation Channels/drug effects , Animals , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , TRPM Cation Channels/deficiency
4.
Sci Rep ; 6: 27903, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27291497

ABSTRACT

Metastasis is a significant health issue. The standard mode of care is combination of chemotherapy and targeted therapeutics but the 5-year survival rate remains low. New/better drug targets that can improve outcomes of patients with metastatic disease are needed. Metastasis is a complex process, with each step conferred by a set of genetic aberrations. Mapping the molecular changes associated with metastasis improves our understanding of the etiology of this disease and contributes to the pipeline of targeted therapeutics. Here, phosphoproteomics of a xenograft-derived in vitro model comprising 4 isogenic cell lines with increasing metastatic potential implicated Transient Receptor Potential Vanilloid subtype 4 in breast cancer metastasis. TRPV4 mRNA levels in breast, gastric and ovarian cancers correlated with poor clinical outcomes, suggesting a wide role of TRPV4 in human epithelial cancers. TRPV4 was shown to be required for breast cancer cell invasion and transendothelial migration but not growth/proliferation. Knockdown of Trpv4 significantly reduced the number of metastatic nodules in mouse xenografts leaving the size unaffected. Overexpression of TRPV4 promoted breast cancer cell softness, blebbing, and actin reorganization. The findings provide new insights into the role of TRPV4 in cancer extravasation putatively by reducing cell rigidity through controlling the cytoskeleton at the cell cortex.


Subject(s)
Actin Cytoskeleton/metabolism , Breast Neoplasms/pathology , TRPV Cation Channels/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Calcium/metabolism , Cell Line, Tumor , Cell Movement , Disease-Free Survival , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Phosphopeptides/analysis , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Transendothelial and Transepithelial Migration , Transplantation, Heterologous , Up-Regulation
5.
Article in English | MEDLINE | ID: mdl-26997877

ABSTRACT

Mutations in CD2-associated protein (CD2AP) have been identified in patients with focal segmental glomerulosclerosis (FSGS); however, reports of CD2AP mutations remain scarce. We performed Sanger sequencing in a patient with steroid-resistant FSGS and identified a heterozygous CD2AP mutation (p.T374A, c.1120 A > G). Our patient displayed mild cognitive decline, a phenotypic characteristic not previously associated with CD2AP-associated FSGS. His proteinuria was remarkably reduced by treatment with cyclosporine A. Our findings expand the genetic spectrum of CD2AP-associated disorders and broaden the associated phenotype with the co-occurrence of cognitive decline. Our case shows that cyclosporin A is a treatment option for CD2AP-associated nephropathy.

6.
J Am Soc Nephrol ; 27(9): 2771-83, 2016 09.
Article in English | MEDLINE | ID: mdl-26892346

ABSTRACT

FSGS is a CKD with heavy proteinuria that eventually progresses to ESRD. Hereditary forms of FSGS have been linked to mutations in the transient receptor potential cation channel, subfamily C, member 6 (TRPC6) gene encoding a nonselective cation channel. Most of these TRPC6 mutations cause a gain-of-function phenotype, leading to calcium-triggered podocyte cell death, but the underlying molecular mechanisms are unclear. We studied the molecular effect of disease-related mutations using tridimensional in silico modeling of tetrameric TRPC6. Our results indicated that G757 is localized in a domain forming a TRPC6-TRPC6 interface and predicted that the amino acid exchange G757D causes local steric hindrance and disruption of the channel complex. Notably, functional characterization of model interface domain mutants suggested a loss-of-function phenotype. We then characterized 19 human FSGS-related TRPC6 mutations, the majority of which caused gain-of-function mutations. However, five mutations (N125S, L395A, G757D, L780P, and R895L) caused a loss-of-function phenotype. Coexpression of wild-type TRPC6 and TRPC6 G757D, mimicking heterozygosity observed in patients, revealed a dominant negative effect of TRPC6 G757D. Our comprehensive analysis of human disease-causing TRPC6 mutations reveals loss of TRPC6 function as an additional concept of hereditary FSGS and provides molecular insights into the mechanism responsible for the loss-of-function phenotype of TRPC6 G757D in humans.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Mutation , TRPC Cation Channels/genetics , DNA Mutational Analysis , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , TRPC6 Cation Channel
7.
Article in English | MEDLINE | ID: mdl-26384487

ABSTRACT

Meantime, it is well accepted that hyperforin, the chemical instable phloroglucinol derivative of Hypericum perforatum, St. John's wort, is the pharmacophore of St. John's wort extracts. With the decline of this scientific discussion, another controversial aspect has been arisen, the question regarding the underlying mechanism leading to the pharmacological profile of the plant extract used in therapy of depression. We will summarize the different concepts described for hyperforin's antidepressive activity. Starting with unspecific protein-independent mechanisms due to changes in pH, we will summarize data of protein-based concepts beginning with concepts based on involvement of a variety of proteins and will finally present concepts based on the modulation of a single protein.


Subject(s)
Phloroglucinol/analogs & derivatives , TRPC Cation Channels/drug effects , Terpenes/pharmacology , Animals , Cytochrome P-450 Enzyme Inducers/pharmacology , Drug Stability , Humans , Phloroglucinol/chemistry , Phloroglucinol/pharmacology , TRPC Cation Channels/physiology , TRPC6 Cation Channel , Terpenes/chemistry
8.
Biochem J ; 469(1): 59-69, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26173259

ABSTRACT

Class IB phosphoinositide 3-kinases γ (PI3Kγ) are second-messenger-generating enzymes downstream of signalling cascades triggered by G-protein-coupled receptors (GPCRs). PI3Kγ variants have one catalytic p110γ subunit that can form two different heterodimers by binding to one of a pair of non-catalytic subunits, p87 or p101. Growing experimental data argue for a different regulation of p87-p110γ and p101-p110γ allowing integration into distinct signalling pathways. Pharmacological tools enabling distinct modulation of the two variants are missing. The ability of an anti-p110γ monoclonal antibody [mAb(A)p110γ] to block PI3Kγ enzymatic activity attracted us to characterize this tool in detail using purified proteins. In order to get insight into the antibody-p110γ interface, hydrogen-deuterium exchange coupled to MS (HDX-MS) measurements were performed demonstrating binding of the monoclonal antibody to the C2 domain in p110γ, which was accompanied by conformational changes in the helical domain harbouring the Gßγ-binding site. We then studied the modulation of phospholipid vesicles association of PI3Kγ by the antibody. p87-p110γ showed a significantly reduced Gßγ-mediated phospholipid recruitment as compared with p101-p110γ. Concomitantly, in the presence of mAb(A)p110γ, Gßγ did not bind to p87-p110γ. These data correlated with the ability of the antibody to block Gßγ-stimulated lipid kinase activity of p87-p110γ 30-fold more potently than p101-p110γ. Our data argue for differential regulatory functions of the non-catalytic subunits and a specific Gßγ-dependent regulation of p101 in PI3Kγ activation. In this scenario, we consider the antibody as a valuable tool to dissect the distinct roles of the two PI3Kγ variants downstream of GPCRs.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Class Ib Phosphatidylinositol 3-Kinase , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Animals , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Deuterium Exchange Measurement , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , HEK293 Cells , Humans , Sf9 Cells , Spodoptera
9.
J Med Chem ; 58(1): 212-21, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-24983663

ABSTRACT

The importance of complete elucidation of the biological functions of phosphoinositide 3-kinases (PI3K) was realized years ago. They generate 3-phosphoinositides, which are known to function as important second messengers in many inter- and intracellular signaling pathways. However, the functional role of class II PI3Ks is still unclear. Herein, we describe the synthesis of a panel of compounds that were tested against all eight mammalian PI3K-isoforms. We found inhibitors with some selectivity for class II PI3K-C2γ and also compounds with preferred inhibition of class II PI3K-C2ß, providing structural leads to develop selective tool compounds.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Class II Phosphatidylinositol 3-Kinases/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Humans , Models, Chemical , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Pyrazines/chemistry , Pyrazines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Benzenesulfonamides
10.
Eur J Dermatol ; 24(6): 650-61, 2014.
Article in English | MEDLINE | ID: mdl-25514792

ABSTRACT

Regular keratinocyte differentiation is crucial for the formation of an intact epidermal barrier and is triggered by extracellular calcium. Disturbances of epidermal barrier formation and aberrant keratinocyte differentiation are involved in the pathophysiology of several skin diseases, such as psoriasis, atopic dermatitis, basal and squamous skin cancer, and genetic skin diseases such as Darier's disease and Olmstedt syndrome. In this review, we summarize current knowledge about the underlying molecular mechanisms of calcium-induced differentiation in keratinocytes. We provide an overview of calcium's genomic and non-genomic mechanisms to induce differentiation and discuss the calcium gradient in the epidermis, giving rise to cornified skin and lipid envelope formation. We focus on the calcium-sensing receptor, transient receptor potential channels, and STIM/Orai as the major constituents of calcium sensing and calcium entry in the keratinocytes. Finally, skin diseases linked to impaired differentiation will be discussed, paying special attention to disturbed TRP channel expression and TRP channel mutations.


Subject(s)
Calcium/metabolism , Cell Differentiation/physiology , Keratinocytes/physiology , Receptors, Calcium-Sensing/metabolism , Skin Diseases/metabolism , Transient Receptor Potential Channels/metabolism , Calcium Channels/metabolism , Carcinoma, Basal Cell/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Differentiation/genetics , Darier Disease/metabolism , Dermatitis, Atopic/metabolism , Humans , Keratinocytes/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , ORAI1 Protein , Psoriasis/metabolism , Skin Neoplasms/metabolism , Stromal Interaction Molecule 1
11.
Cancer Cell ; 26(5): 738-53, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25517751

ABSTRACT

Autophagy promotes tumor growth by generating nutrients from the degradation of intracellular structures. Here we establish, using shRNAs, a dominant-negative mutant, and a pharmacologic inhibitor, mefenamic acid (MFA), that the Transient Receptor Potential Melastatin 3 (TRPM3) channel promotes the growth of clear cell renal cell carcinoma (ccRCC) and stimulates MAP1LC3A (LC3A) and MAP1LC3B (LC3B) autophagy. Increased expression of TRPM3 in RCC leads to Ca(2+) influx, activation of CAMKK2, AMPK, and ULK1, and phagophore formation. In addition, TRPM3 Ca(2+) and Zn(2+) fluxes inhibit miR-214, which directly targets LC3A and LC3B. The von Hippel-Lindau tumor suppressor (VHL) represses TRPM3 directly through miR-204 and indirectly through another miR-204 target, Caveolin 1 (CAV1).


Subject(s)
Autophagy , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , MicroRNAs/physiology , TRPM Cation Channels/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Caveolin 1/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Kidney Neoplasms/genetics , Mice, Nude , Neoplasm Transplantation , Oncogenes , RNA Interference , TRPM Cation Channels/metabolism , Tumor Burden , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
12.
Am J Physiol Endocrinol Metab ; 307(9): E800-12, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205820

ABSTRACT

Bordetella pertussis toxin (PTx), also known as islet-activating protein, induces insulin secretion by ADP-ribosylation of inhibitory G proteins. PTx-induced insulin secretion may result either from inactivation of Gα(o) proteins or from combined inactivation of Gα(o), Gα(i1), Gα(i2), and Gα(i3) isoforms. However, the specific role of Gα(i2) in pancreatic ß-cells still remains unknown. In global (Gα(i2)(-/-)) and ß-cell-specific (Gα(i2)(ßcko)) gene-targeted Gα(i2) mouse models, we studied glucose homeostasis and islet functions. Insulin secretion experiments and intracellular Ca²âº measurements were used to characterize Gα(i2) function in vitro. Gα(i2)(-/-) and Gα(i2)(ßcko) mice showed an unexpected metabolic phenotype, i.e., significantly lower plasma insulin levels upon intraperitoneal glucose challenge in Gα(i2)(-/-) and Gα(i2)(ßcko) mice, whereas plasma glucose concentrations were unchanged in Gα(i2)(-/-) but significantly increased in Gα(i2)(ßcko) mice. These findings indicate a novel albeit unexpected role for Gα(i2) in the expression, turnover, and/or release of insulin from islets. Detection of insulin secretion in isolated islets did not show differences in response to high (16 mM) glucose concentrations between control and ß-cell-specific Gα(i2)-deficient mice. In contrast, the two- to threefold increase in insulin secretion evoked by L-arginine or L-ornithine (in the presence of 16 mM glucose) was significantly reduced in islets lacking Gα(i2). In accord with a reduced level of insulin secretion, intracellular calcium concentrations induced by the agonistic amino acid L-arginine did not reach control levels in ß-cells. The presented analysis of gene-targeted mice provides novel insights in the role of ß-cell Gα(i2) showing that amino acid-induced insulin-release depends on Gα(i2).


Subject(s)
Arginine/metabolism , GTP-Binding Protein alpha Subunit, Gi2/agonists , Hyperglycemia/prevention & control , Insulin/metabolism , Islets of Langerhans/metabolism , Ornithine/metabolism , Up-Regulation , Animals , Blood Glucose/analysis , Calcium Signaling , Crosses, Genetic , Down-Regulation , Fluorescent Antibody Technique , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hyperglycemia/blood , Hyperglycemia/metabolism , Hypoglycemia/blood , Hypoglycemia/metabolism , Hypoglycemia/prevention & control , Insulin/blood , Insulin Secretion , Islets of Langerhans/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ornithine/blood , Specific Pathogen-Free Organisms , Tissue Culture Techniques
13.
J Pharmacol Exp Ther ; 348(3): 393-400, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24259679

ABSTRACT

The acylated phloroglucinol, hyperforin, the main active ingredient of St. John's Wort, exerts antidepressant properties via indirect inhibition of serotonin reuptake by selectively activating the canonical transient receptor potential channel 6 (TRPC6). Hyperforin treatment can lead to drug-drug interactions due to potent activation of the nuclear receptor PXR (NR1I2), a key transcriptional regulator of genes involved in drug metabolism and transport. It was previously shown that synthetic acylated phloroglucinol derivatives activate TRPC6 with similar potency as hyperforin. However, their interaction potential with PXR remained unknown. Here we investigated five synthetic TRPC6-activating phloroglucinol derivatives and four TRPC6-nonactivating compounds compared with hyperforin and rifampicin for their potential to activate PXR in silico and in vitro. Computational PXR pharmacophore modeling did not indicate potent agonist or antagonist interactions for the TRPC6-activating derivatives, whereas one of them was suggested by docking studies to show both agonist and antagonist interactions. Hyperforin and rifampicin treatment of HepG2 cells cotransfected with human PXR expression vector and a CYP3A4 promoter-reporter construct resulted in potent PXR-dependent induction, whereas all TRPC6-activating compounds failed to show any PXR activation or to antagonize rifampicin-mediated CYP3A4 promoter induction. Hyperforin and rifampicin treatment of primary human hepatocytes resulted in highly correlated induction of PXR target genes, whereas treatment with the phloroglucinol derivatives elicited moderate gene expression changes that were only weakly correlated with those of rifampicin and hyperforin treatment. These results show that TRPC6-activating phloroglucinols do not activate PXR and should therefore be promising new candidates for further drug development.


Subject(s)
Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Receptors, Steroid/agonists , Terpenes/pharmacology , Cytochrome P-450 CYP3A/genetics , Genes, Reporter , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Docking Simulation , Phloroglucinol/chemistry , Pregnane X Receptor , Promoter Regions, Genetic , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/chemistry , Terpenes/chemistry , Transcription, Genetic , Transcriptome
14.
Proc Natl Acad Sci U S A ; 110(47): 18862-7, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24190998

ABSTRACT

Phosphoinositide 3-kinase gamma (PI3Kγ) has profound roles downstream of G-protein-coupled receptors in inflammation, cardiac function, and tumor progression. To gain insight into how the enzyme's activity is shaped by association with its p101 adaptor subunit, lipid membranes, and Gßγ heterodimers, we mapped these regulatory interactions using hydrogen-deuterium exchange mass spectrometry. We identify residues in both the p110γ and p101 subunits that contribute critical interactions with Gßγ heterodimers, leading to PI3Kγ activation. Mutating Gßγ-interaction sites of either p110γ or p101 ablates G-protein-coupled receptor-mediated signaling to p110γ/p101 in cells and severely affects chemotaxis and cell transformation induced by PI3Kγ overexpression. Hydrogen-deuterium exchange mass spectrometry shows that association with the p101 regulatory subunit causes substantial protection of the RBD-C2 linker as well as the helical domain of p110γ. Lipid interaction massively exposes that same helical site, which is then stabilized by Gßγ. Membrane-elicited conformational change of the helical domain could help prepare the enzyme for Gßγ binding. Our studies and others identify the helical domain of the class I PI3Ks as a hub for diverse regulatory interactions that include the p101, p87 (also known as p84), and p85 adaptor subunits; Rab5 and Gßγ heterodimers; and the ß-adrenergic receptor kinase.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Models, Molecular , Phosphatidylinositol 3-Kinases/metabolism , Protein Conformation , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Chemotaxis , Class Ib Phosphatidylinositol 3-Kinase/genetics , Deuterium Exchange Measurement , Enzyme Activation , HEK293 Cells , Humans , Mass Spectrometry , Mice , Microscopy, Confocal , NIH 3T3 Cells , Receptors, G-Protein-Coupled/agonists , Signal Transduction/genetics , ras Proteins/metabolism
15.
Molecules ; 18(10): 12012-28, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-24084011

ABSTRACT

Pregnenolone sulfate is a steroid metabolite with a plethora of actions and functions. As a neurosteroid, pregnenolone sulfate modulates a variety of ion channels, transporters, and enzymes. Interestingly, as a sulfated steroid, pregnenolone sulfate is not the final- or waste-product of pregnenolone being sulfated via a phase II metabolism reaction and renally excreted, as one would presume from the pharmacology textbook knowledge. Pregnenolone sulfate is also the source and thereby the starting point for subsequent steroid synthesis pathways. Most recently, pregnenolone sulfate has been functionally "upgraded" from modulator of ion channels to an activating ion channel ligand. This review will focus on molecular aspects of the neurosteroid, pregnenolone sulfate, its metabolism, concentrations in serum and tissues and last not least will summarize the functional data.


Subject(s)
Pregnenolone/physiology , Transient Receptor Potential Channels/metabolism , Animals , Humans , Ligands , Receptors, GABA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
16.
J Biol Chem ; 288(43): 31059-68, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24014027

ABSTRACT

Class IB phosphoinositide 3-kinase γ (PI3Kγ) comprises a single catalytic p110γ subunit, which binds to two non-catalytic subunits, p87 or p101, and controls a plethora of fundamental cellular responses. The non-catalytic subunits are assumed to be redundant adaptors for Gßγ enabling G-protein-coupled receptor-mediated regulation of PI3Kγ. Growing experimental data provide contradictory evidence. To elucidate the roles of the non-catalytic subunits in determining the specificity of PI3Kγ, we tested the impact of p87 and p101 in heterodimeric p87-p110γ and p101-p110γ complexes on the modulation of PI3Kγ activity in vitro and in living cells. RT-PCR, biochemical, and imaging data provide four lines of evidence: (i) specific expression patterns of p87 and p101, (ii) up-regulation of p101, providing the basis to consider p87 as a protein forming a constitutively and p101 as a protein forming an inducibly expressed PI3Kγ, (iii) differences in basal and stimulated enzymatic activities, and (iv) differences in complex stability, all indicating apparent diversity within class IB PI3Kγ. In conclusion, expression and activities of PI3Kγ are modified differently by p87 and p101 in vitro and in living cells, arguing for specific regulatory roles of the non-catalytic subunits in the differentiation of PI3Kγ signaling pathways.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Gene Expression Regulation, Enzymologic/physiology , Protein Multimerization/physiology , Signal Transduction/physiology , Animals , Class Ib Phosphatidylinositol 3-Kinase/genetics , Female , HEK293 Cells , Humans , Male , Sf9 Cells , Spodoptera , Substrate Specificity/physiology
17.
Hippocampus ; 23(1): 40-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22815087

ABSTRACT

The standardized extract of the St. John's wort plant (Hypericum perforatum) is commonly used to treat mild to moderate depression. Its active constituent is hyperforin, a phloroglucinol derivative that reduces the reuptake of serotonin and norepinephrine by increasing intracellular Na(+) concentration through the activation of nonselective cationic TRPC6 channels. TRPC6 channels are also Ca(2+) -permeable, resulting in intracellular Ca(2+) elevations. Indeed, hyperforin activates TRPC6-mediated currents and Ca(2+) transients in rat PC12 cells, which induce their differentiation, mimicking the neurotrophic effect of nerve growth factor. Here, we show that hyperforin modulates dendritic spine morphology in CA1 and CA3 pyramidal neurons of hippocampal slice cultures through the activation of TRPC6 channels. Hyperforin also evoked intracellular Ca(2+) transients and depolarizing inward currents sensitive to the TRPC channel blocker La(3+) , thus resembling the actions of the neurotrophin brain-derived neurotrophic factor (BDNF) in hippocampal pyramidal neurons. These results suggest that the antidepressant actions of St. John's wort are mediated by a mechanism similar to that engaged by BDNF.


Subject(s)
Dendritic Spines/drug effects , Hypericum/chemistry , Phloroglucinol/analogs & derivatives , Plant Extracts/pharmacology , Pyramidal Cells/drug effects , TRPC Cation Channels/agonists , Terpenes/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Dendritic Spines/metabolism , Depression/drug therapy , Depression/metabolism , Gene Expression/physiology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Lanthanum/pharmacology , Organ Culture Techniques , Phloroglucinol/pharmacology , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , Transfection
18.
Sci Signal ; 5(253): ra89, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23211529

ABSTRACT

Synergistic activation by heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) and receptor tyrosine kinases distinguishes p110ß from other class IA phosphoinositide 3-kinases (PI3Ks). Activation of p110ß is specifically implicated in various physiological and pathophysiological processes, such as the growth of tumors deficient in phosphatase and tensin homolog deleted from chromosome 10 (PTEN). To determine the specific contribution of GPCR signaling to p110ß-dependent functions, we identified the site in p110ß that binds to the Gßγ subunit of G proteins. Mutation of this site eliminated Gßγ-dependent activation of PI3Kß (a dimer of p110ß and the p85 regulatory subunit) in vitro and in cells, without affecting basal activity or phosphotyrosine peptide-mediated activation. Disrupting the p110ß-Gßγ interaction by mutation or with a cell-permeable peptide inhibitor blocked the transforming capacity of PI3Kß in fibroblasts and reduced the proliferation, chemotaxis, and invasiveness of PTEN-null tumor cells in culture. Our data suggest that specifically targeting GPCR signaling to PI3Kß could provide a therapeutic approach for tumors that depend on p110ß for growth and metastasis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblasts/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Class I Phosphatidylinositol 3-Kinases , Fibroblasts/pathology , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Phosphatidylinositol 3-Kinases/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics
19.
Biochem J ; 441(3): 851-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22054284

ABSTRACT

G-protein-regulated PI3Kγ (phosphoinositide 3-kinase γ) plays a crucial role in inflammatory and allergic processes. PI3Kγ, a dimeric protein formed by the non-catalytic p101 and catalytic p110γ subunits, is stimulated by receptor-released Gßγ complexes. We have demonstrated previously that Gßγ stimulates both monomeric p110γ and dimeric p110γ/p101 lipid kinase activity in vitro. In order to identify the Gß residues responsible for the Gßγ-PI3Kγ interaction, we examined Gß1 mutants for their ability to stimulate lipid and protein kinase activities and to recruit PI3Kγ to lipid vesicles. Our findings revealed different interaction profiles of Gß residues interacting with p110γ or p110γ/p101. Moreover, p101 was able to rescue the stimulatory activity of Gß1 mutants incapable of modulating monomeric p110γ. In addition to the known adaptor function of p101, in the present paper we show a novel regulatory role of p101 in the activation of PI3Kγ.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/physiology , GTP-Binding Protein beta Subunits/metabolism , Animals , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/chemistry , Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Drug Resistance/genetics , Drug Resistance/physiology , Enzyme Activation/genetics , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Proteolysis/drug effects , Spodoptera , Transfection , Trypsin/metabolism , Trypsin/pharmacology
20.
J Neurochem ; 119(3): 474-85, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21955047

ABSTRACT

The functional implications of transient receptor potential melastatin 3 (TRPM3) activation, the most recently described member of the melastatin subfamily of cation permeable TRP channels, have begun to be elucidated in recent years. The discovery of TRPM3 activation by the steroid pregnenolone sulfate (PregS) has shed new light on the physiological role of this channel. For example, TRPM3 activation enhances insulin secretion from ß pancreatic cells, induces contraction of vascular smooth muscle, and is also involved in the detection of noxious heat. Although TRPM3 expression has been detected in several regions of the developing and mature brain, little is known about the roles of TRPM3 in brain physiology. In this study, we demonstrate the abundant expression of TRPM3 steroid-sensitive channels in the developing cerebellar cortex. We also show that TRPM3-like channels are expressed at glutamatergic synapses in neonatal Purkinje cells. We recently showed that PregS potentiates spontaneous glutamate release onto neonatal Purkinje cells during a period of active glutamatergic synapse formation; we now show that this effect of PregS is mediated by TRPM3-like channels. Mefenamic acid, a recently discovered TRPM3 antagonist, blocked the effect of PregS on glutamate release. The PregS effect on glutamate release was mimicked by other TRPM3 agonists (nifedipine and epipregnanolone sulfate) but not by a TRMP3-inactive steroid (progesterone). Our findings identify TRPM3 channels as novel modulators of glutamatergic transmission in the developing brain.


Subject(s)
Cerebellum/growth & development , Cerebellum/metabolism , Glutamic Acid/physiology , Neurons/physiology , Pregnenolone/pharmacology , Synaptic Transmission/physiology , TRPM Cation Channels/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cerebellum/drug effects , Glutamic Acid/metabolism , Male , Molecular Sequence Data , Neurons/metabolism , Protein Transport/genetics , Purkinje Cells/drug effects , Purkinje Cells/metabolism , Purkinje Cells/physiology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , TRPM Cation Channels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...