Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 285(3): 742-50, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11453656

ABSTRACT

Despite attempts in several laboratories, it has been difficult to prepare antiserum to the thyrotropin-releasing hormone receptor (TRHR). We have prepared a polyclonal anti-rat TRHR antiserum by immunization of rabbits with a synthetic peptide corresponding to the C-terminus of the TRHR. The specificity of the antiserum was assessed by enzyme-linked immunosorbent assay. The affinity-purified antibody recognized a major broad band at 50-60 kDa and a minor broad band at 100-120 kDa in Western blot analysis of membrane proteins from TRHR-transfected, but not control, HEK293t cells. Binding to both bands was abolished by preincubation with the immunizing peptide but not control peptide. The approach was repeated with rat pituitary F4C1 cells, which lack endogenous TRHRs; membranes from F4C1 cells transfected with TRHR cDNA, but not control cells, showed specific binding by Western blot. Using laser confocal microscopy, the TRHR was visualized on the plasma membrane of transfected, but not control, F4C1 cells. Similar confocal findings were observed in TRHR-transfected HEK293t cells. Within 5 min after TRH addition, the TRHR signal translocated from the plasma membrane to the cytoplasm of F4C1 cells transfected with TRHR cDNA. Ten minutes after TRH addition, the TRHR signal formed aggregates in the cytoplasm. Thirty minutes after TRH treatment, both cytoplasmic and plasma membrane localizations were observed, suggesting recycling of some TRHRs back to the plasma membrane. These observations are consistent with our previous findings using an epitope-tagged TRHR. In conclusion, we have prepared an antiserum that recognizes the native TRHR by Western blot analysis and confocal microscopy.


Subject(s)
Antibody Specificity/immunology , Immune Sera/chemistry , Immune Sera/isolation & purification , Receptors, Thyrotropin-Releasing Hormone/immunology , Animals , Binding, Competitive/immunology , Blotting, Western , Cell Line , Cell Membrane/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immune Sera/metabolism , Immunohistochemistry , Immunosorbent Techniques , Microscopy, Confocal , Pituitary Gland/cytology , Pituitary Gland/metabolism , Rabbits , Rats , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Transfection
2.
J Neurosci ; 21(8): 2622-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11306615

ABSTRACT

Drosophila phototransduction is an important model system for studies of inositol lipid signaling. Light excitation in Drosophila photoreceptors depends on phospholipase C, because null mutants of this enzyme do not respond to light. Surprisingly, genetic elimination of the apparently single inositol trisphosphate receptor (InsP(3)R) of Drosophila has no effect on phototransduction. This led to the proposal that Drosophila photoreceptors do not use the InsP(3) branch of phospholipase C (PLC)-mediated signaling for phototransduction, unlike most other inositol lipid-signaling systems. To examine this hypothesis we applied the membrane-permeant InsP(3)R antagonist 2-aminoethoxydiphenyl borate (2-APB), which has proved to be an important probe for assessing InsP(3)R involvement in various signaling systems. We first examined the effects of 2-APB on Xenopus oocytes. We found that 2-APB is efficient at reversibly blocking the robust InsP(3)-mediated Ca(2+) release and store-operated Ca(2+) entry in Xenopus oocytes at a stage operating after production of InsP(3) but before the opening of the surface membrane Cl(-) channels by Ca(2+). We next demonstrated that 2-APB is effective at reversibly blocking the response to light of Drosophila photoreceptors in a light-dependent manner at a concentration range similar to that effective in Xenopus oocytes and other cells. We show furthermore that 2-APB does not directly block the light-sensitive channels, indicating that it operates upstream in the activation of these channels. The results indicate an important link in the coupling mechanism of vertebrate store-operated channels and Drosophila TRP channels, which involves the InsP(3) branch of the inositol lipid-signaling pathway.


Subject(s)
Calcium Signaling/physiology , Drosophila Proteins , Vision, Ocular/physiology , Animals , Boron Compounds/pharmacology , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calmodulin-Binding Proteins/metabolism , Cells, Cultured , Chloride Channels/immunology , Chloride Channels/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drosophila , Electroretinography/drug effects , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Insect Proteins/metabolism , Light , Membrane Proteins/metabolism , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Photoreceptor Cells, Invertebrate/drug effects , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/radiation effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Transient Receptor Potential Channels , Vision, Ocular/drug effects , Vision, Ocular/radiation effects , Xenopus
3.
Trends Biochem Sci ; 26(3): 161-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11246021

ABSTRACT

The idea that both the substrate and the enzyme contribute to catalysis (substrate assisted catalysis; SAC) is applicable to guanine nucleotide-binding proteins (G proteins). Naturally occurring SAC uses GTP as a general base in the GTPase reaction catalyzed by G proteins. Engineered SAC has identified a putative rate-limiting step for the GTPase reaction and shown that GTPase-deficient oncogenic Ras mutants are not irreversibly impaired. Thus, anti-cancer drugs could potentially be designed to restore the blocked GTPase reaction.


Subject(s)
GTP-Binding Proteins/metabolism , Binding Sites , Catalysis , GTP-Binding Proteins/genetics , GTPase-Activating Proteins/metabolism , Mutation , Substrate Specificity
4.
Mol Cell Neurosci ; 15(5): 429-45, 2000 May.
Article in English | MEDLINE | ID: mdl-10833300

ABSTRACT

The Drosophila light-sensitive channels TRP and TRPL are prototypical members of an ion channel family responsible for a variety of receptor-mediated Ca(2+) influx phenomena, including store-operated calcium influx. While phospholipase Cbeta is essential, downstream events leading to TRP and TRPL activation remain unclear. We investigated the role of the InsP(3) receptor (InsP(3)R) by generating mosaic eyes homozygous for a deficiency of the only known InsP(3)R gene in Drosophila. Absence of gene product was confirmed by RT-PCR, Western analysis, and immunocytochemistry. Mutant photoreceptors underwent late onset retinal degeneration; however, whole-cell recordings from young flies demonstrated that phototransduction was unaffected, quantum bumps, macroscopic responses in the presence and absence of external Ca(2+), light adaptation, and Ca(2+) release from internal stores all being normal. Using the specific TRP channel blocker La(3+) we demonstrated that both TRP and TRPL channel functions were unaffected. These results indicate that InsP(3)R-mediated store depletion does not underlie TRP and TRPL activation in Drosophila photoreceptors.


Subject(s)
Calcium Channels/genetics , Drosophila melanogaster/physiology , Gene Deletion , Photoreceptor Cells, Invertebrate/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Vision, Ocular/physiology , Animals , Calcium/metabolism , Calcium Channels/metabolism , Homozygote , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors , Light , Microscopy, Electron , Microscopy, Electron, Scanning , Mutation/physiology , Photoreceptor Cells, Invertebrate/radiation effects , Photoreceptor Cells, Invertebrate/ultrastructure , Receptors, Cytoplasmic and Nuclear/metabolism , Reference Values , Tissue Distribution
5.
Nat Cell Biol ; 2(5): 296-301, 2000 May.
Article in English | MEDLINE | ID: mdl-10806481

ABSTRACT

In Drosophila photoreceptors, phospholipase C (PLC) and other signalling components form multiprotein structures through the PDZ scaffold protein INAD. Association between PLC and INAD is important for termination of responses to light; the underlying mechanism is, however, unclear. Here we report that the maintenance of large amounts of PLC in the signalling membranes by association with INAD facilitates response termination, and show that PLC functions as a GTPase-activating protein (GAP). The inactivation of the G protein by its target, the PLC, is crucial for reliable production of single-photon responses and for the high temporal and intensity resolution of the response to light.


Subject(s)
GTP-Binding Proteins/metabolism , Isoenzymes/metabolism , Type C Phospholipases/metabolism , Vision, Ocular/physiology , Animals , Drosophila , Gene Expression Regulation, Enzymologic , Heat-Shock Response/physiology , Isoenzymes/genetics , Mutagenesis/physiology , Patch-Clamp Techniques , Phenotype , Phospholipase C beta , Photic Stimulation , Photoreceptor Cells, Invertebrate/enzymology , Type C Phospholipases/genetics
6.
J Neurosci ; 19(20): 8799-807, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10516299

ABSTRACT

Phototransduction in Drosophila is mediated by a G-protein-coupled phospholipase C transduction cascade in which each absorbed photon generates a discrete electrical event, the quantum bump. In whole-cell voltage-clamp recordings, cAMP, as well as its nonhydrolyzable and membrane-permeant analogs 8-bromo-cAMP (8-Br-cAMP) and dibutyryl-cAMP, slowed down the macroscopic light response by increasing quantum bump latency, without changes in bump amplitude or duration. In contrast, cGMP or 8-Br-cGMP had no effect on light response amplitude or kinetics. None of the cyclic nucleotides activated any channels in the plasma membrane. The effects of cAMP were mimicked by application of the non-specific phosphodiesterase inhibitor IBMX and the adenylyl cyclase activator forskolin; zaprinast, a specific cGMP-phosphodiesterase inhibitor, was ineffective. Bump latency was also increased by targeted expression of either an activated G(s) alpha subunit, which increased endogenous adenylyl cyclase activity, or an activated catalytic protein kinase A (PKA) subunit. The action of IBMX was blocked by pretreatment with the PKA inhibitor H-89. The effects of cAMP were abolished in mutants of the ninaC gene, suggesting this nonconventional myosin as a possible target for PKA-mediated phosphorylation. Dopamine (10 microM) and octopamine (100 microM) mimicked the effects of cAMP. These results indicate the existence of a G-protein-coupled adenylyl cyclase pathway in Drosophila photoreceptors, which modulates the phospholipase C-based phototransduction cascade.


Subject(s)
Cyclic AMP/physiology , Drosophila melanogaster/physiology , Light , Photoreceptor Cells/physiology , Photoreceptor Cells/radiation effects , Animals , Cyclic AMP-Dependent Protein Kinases/physiology , GTP-Binding Proteins/physiology , Kinetics , Vision, Ocular/physiology
7.
Proc Natl Acad Sci U S A ; 96(12): 7065-70, 1999 Jun 08.
Article in English | MEDLINE | ID: mdl-10359839

ABSTRACT

Interest in the guanosine triphosphatase (GTPase) reaction of Ras as a molecular drug target stems from the observation that, in a large number of human tumors, Ras is characteristically mutated at codons 12 or 61, more rarely 13. Impaired GTPase activity, even in the presence of GTPase activating proteins, has been found to be the biochemical reason behind the oncogenicity of most Gly12/Gln61 mutations, thus preventing Ras from being switched off. Therefore, these oncogenic Ras mutants remain constitutively activated and contribute to the neoplastic phenotype of tumor cells. Here, we show that the guanosine 5'-triphosphate (GTP) analogue diaminobenzophenone-phosphoroamidate-GTP (DABP-GTP) is hydrolyzed by wild-type Ras but more efficiently by frequently occurring oncogenic Ras mutants, to yield guanosine 5'-diphosphate-bound inactive Ras and DABP-Pi. The reaction is independent of the presence of Gln61 and is most dramatically enhanced with Gly12 mutants. Thus, the defective GTPase reaction of the oncogenic Ras mutants can be rescued by using DABP-GTP instead of GTP, arguing that the GTPase switch of Ras is not irreversibly damaged. An exocyclic aromatic amino group of DABP-GTP is critical for the reaction and bypasses the putative rate-limiting step of the intrinsic Ras GTPase reaction. The crystal structures of Ras-bound DABP-beta,gamma-imido-GTP show a disordered switch I and identify the Gly12/Gly13 region as the hydrophobic patch to accommodate the DABP-moiety. The biochemical and structural studies help to define the requirements for the design of anti-Ras drugs aimed at the blocked GTPase reaction.


Subject(s)
Benzophenones/analysis , Genes, ras , Guanosine Triphosphate/analogs & derivatives , Mutation , ras Proteins/genetics , ras Proteins/metabolism , Benzophenones/chemistry , Benzophenones/metabolism , Crystallography, X-Ray , Escherichia coli , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/analysis , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Molecular Sequence Data , Protein Binding
8.
FEBS Lett ; 433(3): 326-30, 1998 Aug 21.
Article in English | MEDLINE | ID: mdl-9744820

ABSTRACT

Hydrolysis of GTP, bound to members of the G-protein superfamily, terminates their downstream signaling activity. A conserved glutamine serves a critical role in this pivotal guanosine triphosphatase (GTPase) reaction. However, the role of the catalytic glutamine in GTP hydrolysis is still not well understood. We have employed substrate-assisted catalysis to probe the catalytic mechanism of Gs alpha using GTP analogues. These GTP analogues, each having different functional groups, were designed to support or refute particular putative GTPase mechanisms. We have found that a hydrogen donor group, in close proximity to the gamma-phosphate of GTP, is necessary and sufficient to substitute for the function of the catalytic glutamine in the GTPase reaction.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/metabolism , Adenosine Triphosphate/metabolism , Adenylyl Cyclases/metabolism , Animals , Cell Membrane/metabolism , GTP Phosphohydrolases/chemistry , GTP-Binding Protein alpha Subunits, Gs/chemistry , Glutamine/metabolism , Hydrolysis , Kinetics , Parotid Gland/metabolism , Rats , Substrate Specificity
9.
Eur J Biochem ; 249(1): 330-6, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9363787

ABSTRACT

Signaling by guanine-nucleotide-binding proteins (G-proteins) occurs when they are charged with GTP, while hydrolysis of the bound nucleotide turns the signaling off. Despite a wealth of biochemical and structural information, the mechanism of GTP hydrolysis by G-proteins remains controversial. We have employed substrate-assisted catalysis as a novel approach to study catalysis by G-proteins. In these studies, we have used diaminobenzophenone-phosphonoamidate-GTP, a unique GTP analog bearing the functional groups that are missing in the GTPase-deficient [Leu227]G(s alpha) mutant. This mutant, found in various human tumors, fails to hydrolyze GTP for an extended period. In contrast, the GTP analog is hydrolyzed by this mutant and by the wild-type enzyme at the same rate. On the other hand, modification of G(s alpha) by cholera toxin, which catalyses ADP-ribosylation of Arg201 of G(s alpha), decreased the rates of hydrolysis of both GTP and its analog by 95%. These results attest to the specificity of the GTP analog as a unique substrate for the [Leu227]G(s alpha) mutant and to the essential role of Gln227 in GTP hydrolysis. Furthermore, the finding that the GTP analog was hydrolyzed at the same rate as GTP by the wild-type enzyme, favors a model in which formation of a pentavalent transition state intermediate, presumably stabilized by the catalytic glutamine, is not the rate-limiting step of the GTPase reaction.


Subject(s)
GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Point Mutation , Animals , Benzophenones/chemistry , Benzophenones/metabolism , Binding Sites/genetics , Catalysis , Cholera Toxin/pharmacology , Computer Simulation , GTP-Binding Proteins/chemistry , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , In Vitro Techniques , Kinetics , Models, Molecular , Protein Conformation , Rats , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 94(11): 5894-9, 1997 May 27.
Article in English | MEDLINE | ID: mdl-9159171

ABSTRACT

Phototransduction in Drosophila occurs through inositol lipid signaling that results in Ca2+ mobilization. In this system, we investigate the hitherto unknown physiological roles of calmodulin (CaM) in light adaptation and in regulation of the inward current that is brought about by depletion of cellular Ca2+ stores. To see the effects of a decreased Ca-CaM content in photoreceptor cells, we used several methods. Transgenic Drosophila P[ninaCDeltaB] flies, which have CaM-deficient photoreceptors, were studied. The peptide inhibitor M5, which binds to Ca-CaM and prevents its action, was applied. A Ca2+-free medium, which prevents Ca2+ influx and thereby diminishes the generation of Ca-CaM, was used. The decrease in the Ca-CaM level caused the following effects. (i) Fluorescence of Ca2+ indicator revealed an enhanced light-induced Ca2+ release from internal stores. (ii) Measurements of the light-induced current in P[ninaCDeltaB] cells showed a reduced light adaptation. (iii) Internal dialysis of M5 initially enhanced excitation and subsequently disrupted the light-induced current. (iv) An inward dark current appeared after depletion of the Ca2+ stores with ryanodine and caffeine. Importantly, application of Ca-CaM into the photoreceptor cells prevented all of the above effects. We propose that negative feedback of Ca-CaM on Ca2+ release from ryanodine-sensitive stores mediates light adaptation, is essential for light excitation, and keeps the store-operated inward current under a tight control.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Photoreceptor Cells, Invertebrate/physiology , Acclimatization , Animals , Animals, Genetically Modified , Caffeine/pharmacology , Calmodulin/deficiency , Darkness , Drosophila melanogaster , Heparin/pharmacology , In Vitro Techniques , Kinetics , Light , Patch-Clamp Techniques , Photic Stimulation , Photoreceptor Cells, Invertebrate/drug effects , Ryanodine/pharmacology
11.
J Pept Res ; 49(5): 421-6, 1997 May.
Article in English | MEDLINE | ID: mdl-9211223

ABSTRACT

A biased library of 60 novel backbone-bicyclic Substance P analogs was prepared by the simultaneous multiple peptide synthesis method. The peptides, containing both a lactam and a disulfide ring, were synthesized by combined Boc and Fmoc chemistries, and were cyclized on the resin. Cleavage of the S-benzyl group and oxidation of the sulfhydryl groups was enabled by adaptation of the diphenylsulfoxidetrichloromethylsilane method to solid-phase synthesis. The peptides were screened for NK-1 and NK-3 activity, and were found to be weak agonists.


Subject(s)
Peptides, Cyclic/pharmacology , Substance P/analogs & derivatives , Substance P/pharmacology , Animals , Disulfides , Guinea Pigs , Ileum , In Vitro Techniques , Indicators and Reagents , Lactams , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Neurons/physiology , Peptide Library , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Protein Conformation , Receptors, Neurokinin-1/physiology , Receptors, Neurokinin-3/physiology , Substance P/chemical synthesis , Substance P/chemistry
12.
Science ; 275(5303): 1119-21, 1997 Feb 21.
Article in English | MEDLINE | ID: mdl-9027311

ABSTRACT

Phototransduction in Drosophila occurs through the ubiquitous phosphoinositide-mediated signal transduction system. Major unresolved questions in this pathway are the identity and role of the internal calcium stores in light excitation and the mechanism underlying regulation of Ca2+ release from internal stores. Treatment of Drosophila photoreceptors with ryanodine and caffeine disrupted the current induced by light, whereas subsequent application of calcium-calmodulin (Ca-CaM) rescued the inactivated photoresponse. In calcium-deprived wild-type Drosophila and in calmodulin-deficient transgenic flies, the current induced by light was disrupted by a specific inhibitor of Ca-CaM. Furthermore, inhibition of Ca-CaM revealed light-induced release of calcium from intracellular stores. It appears that functional ryanodine-sensitive stores are essential for the photoresponse. Moreover, calcium release from these stores appears to be a component of Drosophila phototransduction, and Ca-CaM regulates this process.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Light , Photoreceptor Cells, Invertebrate/metabolism , Signal Transduction , Animals , Animals, Genetically Modified , Caffeine/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Drosophila/genetics , Drosophila/metabolism , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Patch-Clamp Techniques , Photoreceptor Cells, Invertebrate/drug effects , Ryanodine/pharmacology , Signal Transduction/drug effects , Thapsigargin/pharmacology
13.
Proc Natl Acad Sci U S A ; 93(24): 14146-51, 1996 Nov 26.
Article in English | MEDLINE | ID: mdl-8943075

ABSTRACT

Capacitative Ca2+ entry is a component of the inositol-lipid signaling in which depletion of inositol 1,4,5-trisphosphate (InsP3)-sensitive Ca2+ stores activates Ca2+ influx by a mechanism that is still unknown. This pathway plays a central role in cellular signaling, which is mediated by many hormones, neurotransmitters, and growth factors. Studies of Drosophila photoreceptors provided the first putative capacitative Ca2+ entry mutant designated transient receptor potential (trp) and a Drosophila gene encoding TRP-like protein (trpl). It is not clear how the Ca2+ store depletion signal is relayed to the plasma membrane and whether both TRP and TRPL participate in this process. We report here that coexpressing Drosophila TRP and TRPL in Xenopus oocytes synergistically enhances the endogenous Ca(2+)-activated Cl- current and produces a divalent inward current. Both of these currents are activated by Ca2+ store depletion. In the absence of Ca2+, Mg2+ is the main charge carrier of the divalent current. This current is characterized by lanthanum sensitivity and a voltage-dependent blocking effect of Mg2+, which is relieved at both hyperpolarizing (inward rectification) and depolarizing (outward rectification) potentials. The store-operated divalent current is neither observed in native oocytes nor in oocytes expressing either TRP or TRPL alone. The production of this current implicates a cooperative action of TRP and TRPL in the depletion-activated current.


Subject(s)
Calcium Channels/biosynthesis , Calcium/metabolism , Calmodulin-Binding Proteins/biosynthesis , Drosophila Proteins , Drosophila/physiology , Membrane Proteins/biosynthesis , Oocytes/physiology , Photoreceptor Cells/physiology , Animals , Calcium Channels/physiology , Calmodulin-Binding Proteins/physiology , Female , Ion Channels/biosynthesis , Kinetics , Light , Membrane Potentials/physiology , Membrane Proteins/physiology , TRPC Cation Channels , Transient Receptor Potential Channels , Xenopus
14.
Curr Opin Neurobiol ; 6(4): 459-66, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8794093

ABSTRACT

Invertebrate photoreceptors use the ubiquitous inositol-lipid signaling pathway for phototransduction. This pathway depends on Ca2+ release from internal stores and on Ca2+ entry via light-activated channels to replenish the loss of Ca2+ in those stores. The Drosophila transient receptor potential (TRP) protein is essential for the high Ca2+ permeability and other biophysical properties of these light-activated channels, which affect both excitation and adaptation in photoreceptor cells. Physiological and heterologous expression studies indicate that TRP is a putative subunit of a surface membrane channel that can be activated by depletion of internal Ca2+ stores. Furthermore, trp is an archetypal member of a multigene family whose products share a structure that is highly conserved throughout evolution, from worms to humans.


Subject(s)
Calcium Channels , Calcium/physiology , Drosophila/physiology , Ion Channels/physiology , Photoreceptor Cells, Invertebrate/physiology , Adaptation, Ocular/physiology , Animals , Biological Evolution , Cations, Divalent/pharmacokinetics , Conserved Sequence , Ion Channels/genetics , Ion Channels/metabolism , Ion Channels/radiation effects , Light , Mutation , Permeability , TRPC Cation Channels
15.
J Med Chem ; 39(16): 3174-8, 1996 Aug 02.
Article in English | MEDLINE | ID: mdl-8759639

ABSTRACT

The application of the concept of backbone cyclization to linear substance P (SP) analogs is presented. We describe the synthesis, characterization, and biological activity of a series of backbone-to-amino-terminus cyclic analogs of the C-terminal hexapeptide of SP. These analogs were designed on the basis of NMR data and molecular modeling of the selective NK-1 analog WS-septide (Ac[Arg6,Pro9]SP6-11). A series of peptides with the general formula: cyclo[-CH2)m-NH-CO-(CH2)n-CO-Arg-Phe-Phe-N-]-CH2-CO-Leu-Met-NH2 (n = 2, 3, 6 and m = 2, 3, 4) was synthesized by solid phase methodology using Fmoc chemistry for the main chain and Boc chemistry for the building units [Na-(omega-aminoalkyl)Gly] side chains. Cyclization was performed on the resin after removal of the Boc protecting group from the omega-aminoalkyl chain. Cyclic and precyclic analogs were compared. They were purified by HPLC and characterized by mass spectroscopy and NMR. Biological activity and selectivity to the NK-1 neurokinin receptor were found to depend on cyclization and the ring size: The most active and selective analog had a ring of 20 atoms. This analog was found to have enhanced metabolic stability in various tissue preparation compared to WS-septide.


Subject(s)
Peptide Fragments/pharmacology , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Receptors, Neurokinin-1/agonists , Substance P/pharmacology , Amino Acid Sequence , Animals , Guinea Pigs , Molecular Sequence Data , Muscle, Smooth/drug effects , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Protein Binding , Protein Conformation , Pyrrolidonecarboxylic Acid/analogs & derivatives , Rats , Receptors, Neurokinin-1/metabolism , Structure-Activity Relationship , Substance P/chemical synthesis , Substance P/chemistry
16.
J Pept Sci ; 2(4): 261-9, 1996.
Article in English | MEDLINE | ID: mdl-9231334

ABSTRACT

Novel backbone-to-side chain and backbone-to-backbone cyclic analogues of substance P (SP) were prepared by solid-phase synthesis and screened for biological activity. An analogue containing a thioetherlactam ring between positions 9 and 11 showed an EC50 value of 20 nM toward the neurokinin 1 (NK-1) and was inactive toward the NK-2 and NK-3 receptors. On the other hand, in a multiple backbone cyclic peptide library of similar analogues, in which the sulphur was excluded from the ring, very low activity was detected. The activity was re-evaluated and was found to be even lower (EC50 = 0.11 mM) than the previously published data. These results indicate that the thioether moiety has a crucial role in receptor activation. The results also show tolerance of the NK-1 receptor, but not NK-2 or NK-3, to cyclization of the C-terminal portion of the SP6-11 hexapeptide.


Subject(s)
Substance P/chemical synthesis , Substance P/metabolism , Sulfur/physiology , Animals , Biological Assay , Cyclization , Guinea Pigs , Ileum/drug effects , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-2/drug effects , Receptors, Neurokinin-3/drug effects , Substance P/analogs & derivatives , Sulfur/chemistry
17.
Anal Biochem ; 236(2): 302-8, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8660509

ABSTRACT

Determination of microgram quantities of protein in the Bradford Coomassie brilliant blue assay is accomplished by measurement of absorbance at 590 nm. However, as intrinsic nonlinearity compromises the sensitivity and accuracy of this method. It is shown that under standard assay conditions, the ratio of the absorbances, 590 nm over 450 nm, is strictly linear with protein concentration. This simple procedure increases the accuracy and improves the sensitivity of the assay about 10-fold, permitting quantitation down to 50 ng of bovine serum albumin. Furthermore, protein assay in presence of up to 35-fold weight excess of sodium dodecyl sulfate (detergent) over bovine serum albumin (protein) can be performed. A linear equation that perfectly fits the experimental data is provided on the basis of mass action and Beer's law.


Subject(s)
Serum Albumin, Bovine/analysis , Spectrophotometry/methods , Calibration , Detergents , Indicators and Reagents , Linear Models , Microchemistry , Rosaniline Dyes , Sensitivity and Specificity , Sodium Dodecyl Sulfate
18.
Mol Neurobiol ; 12(2): 163-80, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8818149

ABSTRACT

Inositol lipid signaling relies on an InsP3-induced Ca2+ release from intracellular stores and on extracellular Ca2+ entry, which takes place when the Ca2+ stores become depleted of Ca2+. This interplay between Ca2+ release and Ca2+ entry has been termed capacitative Ca2+ entry and the inward current calcium release activated current (CRAC) to indicate gating of Ca2+ entry by Ca2+-store depletion. The signaling pathway and the gating mechanism of capacitative Ca2+ entry, however, are largely unknown and the molecular participants in this process have not been identified. In this article we review genetic, molecular, and functional studies of wild-type and mutant Drosophila photoreceptors, suggesting that the transient receptor potential mutant (trp) is the first putative capacitative Ca2+ entry mutant. Furthermore, several lines of evidence suggest that the trp gene product TRP is a candidate subunit of the plasma membrane channel that is activated by Ca2+ store depletion.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Drosophila/physiology , Phosphatidylinositols/physiology , Photoreceptor Cells/physiology , Signal Transduction , Animals , Calcium Channels/biosynthesis , Calcium Channels/genetics , Drosophila/genetics , Genes, Insect , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate/physiology , Light , Models, Structural , Phosphatidylinositols/pharmacology , TRPC Cation Channels
19.
Int J Pept Protein Res ; 45(6): 508-13, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7558580

ABSTRACT

The analogues [Glp6,Glu(OBzl)11]SP(6-11) and [Glp5,Glu(OBzl)11]SP(5-11) of the C-terminal hexapeptide and heptapeptide of Substance P have been synthesized by conventional solution methods. In each analogue the N-terminal glutamine has been replaced by pyroglutamic acid, while the COOCH2C6H5 ester group has replaced the SCH3 group of the Met11 side chain. The in vitro activity of both analogues has been determined on three biological preparations: guinea pig ileum (GPI), rat vas deferens (RVD) and rat portal vein (RPV). The results showed that both analogues are highly potent and selective agonists on GPI through the NK-1 receptor. They are more potent than SP itself, with 1.54 and 1.25 respective values of relative potency on GPI. Their selectivity has been studied by utilizing atropine-treated guinea pig ileum (GPI+At). The analogues showed low activity on RVD and RPV tissues, which represent NK-2 and NK-3 monoreceptor assay, respectively.


Subject(s)
Glutamine/chemistry , Methionine/chemistry , Muscle, Smooth, Vascular/drug effects , Oligopeptides/chemistry , Peptide Fragments/chemical synthesis , Pyrrolidonecarboxylic Acid/chemistry , Substance P/analogs & derivatives , Substance P/agonists , Amino Acid Sequence , Animals , Biological Assay , Guinea Pigs , Molecular Sequence Data , Muscle, Smooth/drug effects , Peptide Fragments/pharmacology , Rats , Receptors, Neurokinin-1/analysis , Receptors, Neurokinin-2/analysis , Receptors, Neurokinin-3/analysis , Substance P/chemical synthesis , Substance P/chemistry , Substance P/pharmacology
20.
Biochem J ; 306 ( Pt 1): 253-8, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7864818

ABSTRACT

A novel photoaffinity label, m-acetylanilido-GTP (m-AcAGTP), was synthesized and used to identify GTP-binding proteins (G-proteins). This GTP analogue is easily prepared and can be used for photoaffinity labelling of G-proteins without chromatographic purification. In the presence of the beta-adrenergic agonist isoprenaline, it activates turkey erythrocyte adenylate cyclase. This activation persists even when the beta-adrenergic receptor is subsequently blocked by antagonist, indicating that the GTP analogue is resistant to hydrolysis. The apparent Ka for activation of turkey erythrocyte adenylate cyclase by m-AcAGTP was found to be 0.21 microM, a value similar to that for guanosine 5'-[beta,gamma-imido]triphosphate. m-AcAGTP also effectively inhibited the light-dependent GTPase of Musca fly eye membranes. Photoaffinity labelling of fly eye membranes with [alpha-32P]m-AcAGTP, followed by immunoprecipitation of G-protein Gq, identified a labelled protein band with the mobility of a 41.5 kDa protein on SDS/PAGE. Labelling of this protein was enhanced 9-fold in blue over red illuminated membranes, containing metarhodopsin and rhodopsin respectively. Labelling of alpha-subunits of heterotrimeric G-proteins was also demonstrated in turkey erythrocyte membranes. The ease of preparation of m-AcAGTP and the chemical properties of the photoreactive acetophenone make this affinity label an important new tool in studies of cellular phenomena mediated by guanine nucleotide-binding proteins.


Subject(s)
Affinity Labels , GTP-Binding Proteins/analysis , Guanosine Triphosphate/analogs & derivatives , Adenylyl Cyclases/metabolism , Animals , Enzyme Activation/drug effects , Erythrocytes/enzymology , GTP-Binding Proteins/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/chemical synthesis , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Guanylyl Imidodiphosphate/pharmacology , Immunosorbent Techniques , Isoproterenol/pharmacology , Photochemistry , Propranolol/pharmacology , Turkeys/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...