Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Anal Sci ; 40(5): 925-934, 2024 May.
Article in English | MEDLINE | ID: mdl-38528254

ABSTRACT

Stereoinversion of Ser residues within proteins, which has been identified in long-lived proteins, influences protein function. To quantify the stereoinversion of Ser residues, we investigated the potential adaptation of our direct peptide analytical method originally established for analyzing the isomerization of asparaginyl/aspartyl residues. Peptide pairs containing L-Ser or D-Ser residues with lengths of four or five residues were synthesized. Separation conditions for these peptide pairs were systematically examined by precisely adjusting the pH of the elution solvent using reverse-phase high-performance liquid chromatography (HPLC). Optimal separation conditions were successfully developed for all peptide pairs, enabling the direct quantification of Ser residue stereoinversion through a single HPLC run. Subsequently, the degree of Ser stereoinversion within the model peptide, Gly-Ser-Gly-Tyr, was determined using the method established in this study. Surprisingly, the stereoinversion of Ser residues occurred only when the absolute configurations of Ser and Tyr residues of the peptide differed from each other, whereas no stereoinversion was observed when their absolute configurations were identical. The experiments using peptides similar to the model peptide reveal that both the N-terminal amino group and the hydroxyl group of the C-terminal Tyr residue are involved in the stereoinversion of the Ser residue. By applying a simple method to quantify the stereoinversion of Ser residues, valuable insights into the mechanisms governing these stereoinversions were obtained.

2.
Mol Microbiol ; 118(5): 526-540, 2022 11.
Article in English | MEDLINE | ID: mdl-36065735

ABSTRACT

A Clostridioides difficile strain deficient in the ddl gene is unable to synthesize the dipeptide D-Ala-D-Ala, an essential component of peptidoglycan and the target of vancomycin. We isolated spontaneous suppressors of a ∆ddl mutation that allowed cell growth in the absence of D-Ala-D-Ala. The mutations caused constitutive or partly constitutive expression of the vancomycin-inducible vanG operon responsible for the synthesis of D-Ala-D-Ser, which can replace D-Ala-D-Ala in peptidoglycan. The mutations mapped to the vanS or vanR genes, which regulate expression of the vanG operon. The constitutive level of vanG expression was about 10-fold above that obtained by vancomycin induction. The incorporation of D-Ala-D-Ser into peptidoglycan due to high expression of the vanG operon conferred only low-level resistance to vancomycin, but VanG was found to synthesize D-Ala-D-Ala in addition to D-Ala-D-Ser. However, the same, low resistance to vancomycin was also observed in cells completely unable to synthesize D-Ala-D-Ala and grown in the presence of D-Ala-D-Ser. D-Ala-D-Ala presence was required for efficient vancomycin induction of the vanG operon showing that vancomycin is not by itself able to activate VanS. D-Ala-D-Ser, similar to D-Ala-D-Ala, served as an anti-activator of DdlR, the positive regulator of the ddl gene, thereby coupling vanG and ddl expression.


Subject(s)
Clostridioides difficile , Vancomycin Resistance , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Peptidoglycan/metabolism , Clostridioides difficile/genetics , Clostridioides , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Transcription Factors , Anti-Bacterial Agents/pharmacology
3.
Neural Regen Res ; 17(9): 2072-2078, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35142699

ABSTRACT

In our previous studies, we have shown that (D-Ser2) oxyntomodulin (Oxm), a glucagon-like peptide 1 (GLP-1) receptor (GLP1R)/glucagon receptor (GCGR) dual agonist peptide, protects hippocampal neurons against Aß1-42-induced cytotoxicity, and stabilizes the calcium homeostasis and mitochondrial membrane potential of hippocampal neurons. Additionally, we have demonstrated that (D-Ser2) Oxm improves cognitive decline and reduces the deposition of amyloid-beta in Alzheimer's disease model mice. However, the protective mechanism remains unclear. In this study, we showed that 2 weeks of intraperitoneal administration of (D-Ser2) Oxm ameliorated the working memory and fear memory impairments of 9-month-old 3×Tg Alzheimer's disease model mice. In addition, electrophysiological data recorded by a wireless multichannel neural recording system implanted in the hippocampal CA1 region showed that (D-Ser2) Oxm increased the power of the theta rhythm. In addition, (D-Ser2) Oxm treatment greatly increased the expression level of synaptic-associated proteins SYP and PSD-95 and increased the number of dendritic spines in 3×Tg Alzheimer's disease model mice. These findings suggest that (D-Ser2) Oxm improves the cognitive function of Alzheimer's disease transgenic mice by recovering hippocampal synaptic function and theta rhythm.

4.
Biomolecules ; 11(11)2021 11 18.
Article in English | MEDLINE | ID: mdl-34827714

ABSTRACT

In nature, amino acids are found in two forms, L and D enantiomers, except for glycine which does not have a chiral center. The change of one form to the other will lead to a change in the primary structure of proteins and hence may affect the function and biological activity of proteins. Indeed, several D-amino acid-containing peptides (DAACPs) were isolated from patients with cataracts, Alzheimer's and other diseases. Additionally, significant levels of free D-amino acids were found in several diseases, reflecting the disease conditions. Studying the molecular mechanisms of the DAACPs formation and the alteration in D-amino acids metabolism will certainly assist in understanding these diseases and finding new biomarkers and drug targets. In this review, the presence of DAACPs and free D-amino acids and their links with disease development and progress are summarized. Similarly, we highlight some recent advances in analytical techniques that led to improvement in the discovery and analysis of DAACPs and D-amino acids.


Subject(s)
Peptides , Amino Acids , Stereoisomerism
5.
Peptides ; 134: 170399, 2020 12.
Article in English | MEDLINE | ID: mdl-32889021

ABSTRACT

Humanin (HN) is a 24-amino acid mitochondrial-derived peptide, best known for its ability to protect neurons from damage caused by ischemic stroke and neurodegenerative insults and cardiomyocytes from myocardial infarction or doxorubicin (Dox)-induced cardiotoxicity. This study examines the neuroprotective and myoprotective effects of HN novel synthetic analogs HUJInin and c(D-Ser14-HN), prepared by solid-phase peptide synthesis. The cellular models employed were oxygen-glucose-deprivation (OGD) followed by reoxygenation (R)-induced neurotoxicity in PC12 and SH-SY5Y neuronal cell cultures and Dox-induced cardiotoxicity in H9c2 and C2C12 myoblast cell cultures, respectively. Necrotic and apoptotic cell death was measured by LDH release and caspase-3 activity. Erk 1/2 and AKT phosphorylations were examined by western blotting. Mitochondrial calcium and mitochondrial membrane potential were measured using the fluorescent dye tetramethylrhodamine-methyl ester. It was found that HUJInin and c(D-Ser14-HN) conferred significant dose-dependent neuroprotection, a phenomenon related to attenuation of OGD insult-induced Erk 1/2 phosphorylation, stimulation of AKT phosphorylation and improvement of mitochondrial functions. These peptides also conferred myoprotective effect towards Dox-induced apo-necrotic cell death insults. HUJInin and c(D-Ser14-HN) synthetic analogs may provide new lead compounds for the development of a potential candidate drug for stroke treatment and/or Dox-induced cardiotoxicity therapy in cancer patients.


Subject(s)
Doxorubicin/toxicity , Intracellular Signaling Peptides and Proteins/pharmacology , Ischemia/physiopathology , Mitochondria/drug effects , Myoblasts/drug effects , Neurons/drug effects , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Mice , Mitochondria/metabolism , Mitochondria/pathology , Myoblasts/metabolism , Myoblasts/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Phosphorylation , Rats
6.
Enzyme Microb Technol ; 137: 109547, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32423675

ABSTRACT

Recombinant protein overexpressed in Escherichia coli is often less folded, leading to form the insoluble aggregates also called as inclusion bodies (IBs). IBs are classified as active and inactive ones, and enzyme produced as active IBs is the novel carrier-free immobilized material. In this study, we determined that His6-tagged serine racemase (SR) from maize or human produced as partially active IBs maintained the activities for reversible racemization of l-serine to d-serine but lost the activities for irreversible ß-elimination of both enantiomers, in contrast to the soluble one displaying all activities. Fourier transform infrared spectroscopy analysis showed structural changes between the soluble and insoluble SR. Compared with the soluble SR with attachment of the N-terminal cellulose-binding module via the oriented immobilization of the regenerated amorphous cellulose, the insoluble SR with the fusion of the N-terminal aggregation-inducible tag GFIL8 displayed higher production and usage efficiency, lower leaky capacity, more stability at 4 °C storage with the prolonged time, less sensitivity to the limited proteolysis mediated by trypsin, and higher yield of the synthesized d-serine. These advantages allow the SRs as partially active IBs to synthesize d-serine for medical and agricultural applications.


Subject(s)
Escherichia coli/metabolism , Inclusion Bodies/metabolism , Racemases and Epimerases/biosynthesis , Serine/biosynthesis , Zea mays/enzymology , Escherichia coli/genetics , Humans , Protein Binding , Racemases and Epimerases/genetics , Recombinant Proteins/biosynthesis , Spectroscopy, Fourier Transform Infrared
7.
Horm Behav ; 118: 104640, 2020 02.
Article in English | MEDLINE | ID: mdl-31765661

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease that severely affects the health and lifespan of the elderly worldwide. Recently, the correlation between AD and type 2 diabetes mellitus (T2DM) has received intensive attention, and a promising new anti-AD strategy is the use of anti-diabetic drugs. Oxyntomodulin (Oxm) is a peptide hormone and growth factor that acts on neurons in the hypothalamus. OXM activates glucagon-like peptide 1 (GLP-1) and glucagon (Gcg) receptors, facilitates insulin signaling and has neuroprotective effects against Aß1-42-induced cytotoxicity in primary hippocampal neurons. Here, we tested the effects of the protease-resistant analogue (D-Ser2)Oxm on spatial memory and synaptic plasticity and the underlying molecular mechanisms in the APP/PS1 transgenic mouse model of AD. The results showed that (D-Ser2)Oxm not only alleviated the impairments of working memory and long-term spatial memory, but also reduced the number of Aß plaques in the hippocampus, and reversed the suppression of hippocampal synaptic long-term potentiation (LTP). Moreover, (D-Ser2)Oxm administration significantly increased p-PI3K/p-AKT1 expression and decreased p-GSK3ß levels in the hippocampus. These results are the first to show an in vivo neuroprotective role of (D-Ser2)Oxm in APP/PS1 mice, and this role involves the improvement of synaptic plasticity, clearance of Aß and normalization of PI3K/AKT/GSK3ß cell signaling in the hippocampus. This study suggests that (D-Ser2)Oxm holds promise for the prevention and treatment of AD.


Subject(s)
Alzheimer Disease/pathology , Glucagon-Like Peptide 1/agonists , Neuronal Plasticity/drug effects , Oxyntomodulin/pharmacology , Receptors, Glucagon/agonists , Spatial Memory/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Glucagon-Like Peptide 1/pharmacology , Hippocampus/drug effects , Insulin/metabolism , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents/pharmacology , Oxyntomodulin/therapeutic use , Presenilin-1/genetics
8.
Biochem Biophys Res Commun ; 504(1): 40-45, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30173889

ABSTRACT

D-Serine deaminase (DSD) degrades D-Ser to pyruvate and ammonia. Uropathogenic bacteria survive in the toxic D-Ser containing mammalian urine because of DSD activity. The crystal structure of the apo form of Salmonella typhimurium DSD (StDSD) has been reported earlier. In the present work, we have investigated the role of two active site residues, Thr166 and Asp236 by site directed mutagenesis (T166A and D236L). The enzyme activity is lost upon mutation of these residues. The 2.7 Šresolution crystal structure of T166A DSD with bound PLP reported here represents the first structure of the holo form of StDSD. PLP binding induces small changes in the relative dispositions of the minor and major domains of the protein and this inter-domain movement becomes substantial upon interaction with the substrate. The conformational changes bring Thr166 to a position at the active site favorable for the degradation of D-Ser. Examination of the different forms of the enzyme and comparison with structures of homologous enzymes suggests that Thr166 is the most probable base abstracting proton from the Cα atom of the substrate and Asp236 is crucial for binding of the cofactor.


Subject(s)
Aspartic Acid/chemistry , L-Serine Dehydratase/chemistry , Salmonella typhimurium/enzymology , Threonine/chemistry , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , L-Serine Dehydratase/genetics , Models, Molecular , Mutation , Pyridoxal Phosphate/chemistry
9.
Amino Acids ; 49(10): 1743-1754, 2017 10.
Article in English | MEDLINE | ID: mdl-28744579

ABSTRACT

Recently, we cloned and characterized eleven serine and aspartate racemases (SerR and AspR, respectively) from animals. These SerRs and AspRs are not separated by their racemase functions and form a serine/aspartate racemase family cluster based on phylogenetic analysis. Moreover, we have proposed that the AspR-specific triple serine loop region at amino acid positions 150-152 may be responsible for the large AspR activity. In the present study, to test this hypothesis, we prepared and characterized fourteen mutants in this region of animal SerRs and AspRs. The large AspR activity in Acropora and Crassostrea AspR was reduced to <0.04% of wild-type after substitution of the triple serine loop region. Conversely, introducing the triple serine loop region into Acropora, Crassostrea, and Penaeus SerR drastically increased the AspR activity. Those mutants showed similar or higher substrate affinity for aspartate than serine and showed 11-683-fold higher k cat and 28-351-fold higher k cat/K m values for aspartate than serine racemization. Furthermore, we introduced serine residues in all combinations at position 150-152 in mouse SerR. These mutants revealed that a change in the enzyme function from SerR to AspR can be caused by introduction of Ser151 and Ser152, and addition of the third serine residue at position 150 further enhances the enzyme specificity for aspartate due to a decrease in the serine racemase and serine dehydratase activity. Here, we provide convincing evidence that the AspR gene has evolved from the SerR gene by acquisition of the triple serine loop region.


Subject(s)
Amino Acid Isomerases , Anthozoa , Arthropod Proteins , Crassostrea , Mutation, Missense , Penaeidae , Racemases and Epimerases , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/genetics , Amino Acid Substitution , Animals , Anthozoa/enzymology , Anthozoa/genetics , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Crassostrea/enzymology , Crassostrea/genetics , Mice , Penaeidae/enzymology , Penaeidae/genetics , Protein Structure, Secondary , Racemases and Epimerases/chemistry , Racemases and Epimerases/genetics
10.
Amino Acids ; 49(9): 1521-1533, 2017 09.
Article in English | MEDLINE | ID: mdl-28681245

ABSTRACT

More than half a century ago researchers thought that D-amino acids had a minor function compared to L-enantiomers in biological processes. Many evidences have shown that D-amino acids are present in high concentration in microorganisms, plants, mammals and humans and fulfil specific biological functions. In the brain of mammals, D-serine (D-Ser) acts as a co-agonist of the N-methyl-D-aspartate (NMDA)-type glutamate receptors, responsible for learning, memory and behaviour. D-Ser metabolism is relevant for disorders associated with an altered function of the NMDA receptor, such as schizophrenia, ischemia, epilepsy and neurodegenerative disorders. On the other hand, D-aspartate (D-Asp) is one of the major regulators of adult neurogenesis and plays an important role in the development of endocrine function. D-Asp is present in the neuroendocrine and endocrine tissues and testes, and regulates the synthesis and secretion of hormones and spermatogenesis. Also food proteins contain D-amino acids that are naturally originated or processing-induced under conditions such as high temperatures, acid and alkali treatments and fermentation processes. The presence of D-amino acids in dairy products denotes thermal and alkaline treatments and microbial contamination. Two enzymes are involved in the metabolism of D-amino acids: amino acid racemase in the synthesis and D-amino acid oxidase in the degradation.


Subject(s)
Amino Acid Isomerases/chemistry , D-Amino-Acid Oxidase/chemistry , D-Aspartic Acid/chemistry , Serine/chemistry , Amino Acid Isomerases/metabolism , Amphibian Venoms/chemistry , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/chemistry , Bacteria/chemistry , Bacteria/metabolism , D-Amino-Acid Oxidase/metabolism , D-Aspartic Acid/metabolism , Diet , Glyceraldehyde/chemistry , Glyceraldehyde/metabolism , Humans , Plants/chemistry , Plants/metabolism , Pyridoxal Phosphate/chemistry , Pyridoxal Phosphate/metabolism , Schiff Bases/chemistry , Schiff Bases/metabolism , Serine/metabolism , Stereoisomerism
11.
Neuropharmacology ; 117: 408-421, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28219718

ABSTRACT

The Regulator of G protein Signalling 4 (RGS4) is a multitask protein that interacts with and negatively modulates opioid receptor signalling. Previously, we showed that the δ-opioid receptor (δ-OR) forms a multiprotein signalling complex consisting of Gi/Go proteins and the Signal Transducer and Activator of Transcription 5B (STAT5B) that leads to neuronal differentiation and neurite outgrowth upon δ-ΟR activation. Here, we investigated whether RGS4 could participate in signalling pathways to regulate neurotropic events. We demonstrate that RGS4 interacts directly with STAT5B independently of δ-ΟR presence both in vitro and in living cells. This interaction involves the N-terminal portion of RGS4 and the DNA-binding SH3 domain of STAT5B. Expression of RGS4 in HEK293 cells expressing δ-OR and/or erythropoietin receptor results in inhibition of [D-Ser2, Leu5, Thr6]-enkephalin (DSLET)-and erythropoietin-dependent STAT5B phosphorylation and subsequent transcriptional activation. DSLET-dependent neurite outgrowth of neuroblastoma cells is also blocked by RGS4 expression, whereas primary cortical cultures of RGS4 knockout mice (RGS4-/-) exhibit enhanced neuronal sprouting after δ-OR activation. Additional studies in adult brain extracts from RGS4-/- mice revealed increased levels of p-STAT5B. Finally, neuronal progenitor cultures from RGS4-/- mice exhibit enhanced proliferation with concomitant increases in the mRNA levels of the anti-apoptotic STAT5B target genes bcl2 and bcl-xl. These observations suggest that RGS4 is implicated in opioid dependent neuronal differentiation and neurite outgrowth via a "non-canonical" signaling pathway regulating STAT5B-directed responses.


Subject(s)
Neurogenesis/physiology , Neuronal Outgrowth/physiology , Neurons/metabolism , RGS Proteins/metabolism , STAT5 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Cell Survival/physiology , Cerebral Cortex/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/metabolism , Phosphorylation/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , RGS Proteins/genetics , RNA, Messenger/metabolism , Rats , Receptors, Erythropoietin/metabolism , Receptors, Opioid, delta/metabolism , bcl-X Protein/metabolism
12.
Amino Acids ; 48(2): 387-402, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26352274

ABSTRACT

Free D-amino acids have been found in various invertebrate phyla, while amino acid racemase genes have been identified in few species. The purpose of this study is to elucidate the distribution, function, and evolution of amino acid racemases in invertebrate animals. We searched the GenBank databases, and found 11 homologous serine racemase genes from eight species in eight different invertebrate phyla. The cloned genes were identified based on their maximum activity as Acropora millepora (Cnidaria) serine racemase (SerR) and aspartate racemase (AspR), Caenorhabditis elegans (Nematoda) SerR, Capitella teleta (Annelida) SerR, Crassostrea gigas (Mollusca) SerR and AspR, Dugesia japonica (Platyhelminthes) SerR, Milnesium tardigradum (Tardigrada) SerR, Penaeus monodon (Arthropoda) SerR and AspR and Strongylocentrotus purpuratus (Echinodermata) AspR. We found that Acropora, Aplysia, Capitella, Crassostrea and Penaeus had two amino acid racemase paralogous genes and these paralogous genes have evolved independently by gene duplication at their recent ancestral species. The transcriptome analyses using available SRA data and enzyme kinetic data suggested that these paralogous genes are expressed in different tissues and have different functions in vivo. Phylogenetic analyses clearly indicated that animal SerR and AspR are not separated by their particular racemase functions and form a serine/aspartate racemase family cluster. Our results revealed that SerR and AspR are more widely distributed among invertebrates than previously known. Moreover, we propose that the triple serine loop motif at amino acid positions 150-152 may be responsible for the large aspartate racemase activity and the AspR evolution from SerR.


Subject(s)
Amino Acid Isomerases/genetics , Aspartic Acid/metabolism , Invertebrates/enzymology , Racemases and Epimerases/genetics , Serine/metabolism , Amino Acid Isomerases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Escherichia coli/genetics , Invertebrates/genetics , Phylogeny , Pyridoxal Phosphate/metabolism , Racemases and Epimerases/metabolism , Sequence Alignment , Sequence Analysis, DNA
14.
Cell Signal ; 25(12): 2634-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24012499

ABSTRACT

Western blot analysis demonstrated that PC-12 cells express monomeric and dimeric forms of serine racemase (m-SR, d-SR) and that 1321N1 cells express m-SR. Quantitative RT-PCR and functional studies demonstrated that PC-12 cells express homomeric and heteromeric forms of nicotinic acetylcholine receptors (nAChR) while 1321N1 cells primarily express the α7-nAChR subtype. The effect of nAChR agonists and antagonists on SR activity and expression was examined by following concentration-dependent changes in intracellular d-Ser levels and SR protein expression. Incubation with (S)-nicotine increased d-Ser levels, which were attenuated by the α7-nAChR antagonist methyllycaconitine (MLA). Treatment of PC-12 cells with mecamylamine (MEC) produced a bimodal reduction of d-Ser reflecting MEC inhibition of homomeric and heteromeric nAChRs, while a unimodal curve was observed with 1321N1 cells, reflecting predominant expression of α7-nAChR. The nAChR subtype selectivity was probed using α7-nAChR selective inhibitors MLA and (R,S)-dehydronorketamine and α3ß4-nAChR specific inhibitor AT-1001. The compounds reduced d-Ser in PC-12 cells, but only MLA and (R,S)-dehydronorketamine were effective in 1321N1 cells. Incubation of PC-12 and 1321N1 cells with (S)-nicotine, MEC and AT-1001 did not affect m-SR or d-SR expression, while MLA and (R,S)-dehydronorketamine increased m-SR expression but not SR mRNA levels. Treatment with cycloheximide indicated that increased m-SR was due to de novo protein synthesis associated with phospho-active forms of ERK1/2, MARCKS, Akt and rapamycin-sensitive mTOR. This effect was attenuated by treatment with the pharmacological inhibitors U0126, LY294002 and rapamycin, which selectively block the activation of ERK1/2, Akt and mTOR, respectively, and siRNAs directed against ERK1/2, Akt and mTOR. We propose that nAChR-associated changes in Ca(2+) flux affect SR activity, but not expression, and that MLA and (R,S)-dehydronorketamine bind to allosteric sites on the α7-nAChR and promote multiple signaling cascades that converge at mTOR to increase m-SR levels.


Subject(s)
Aconitine/analogs & derivatives , Mecamylamine/pharmacology , Nicotinic Antagonists/pharmacology , PC12 Cells/drug effects , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Receptors, Nicotinic/metabolism , Aconitine/pharmacology , Animals , Cell Line , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Nicotine/metabolism , PC12 Cells/enzymology , PC12 Cells/metabolism , Rats , Receptors, Nicotinic/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
15.
Bioorg Med Chem Lett ; 23(18): 5082-5, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23932788

ABSTRACT

Derivatives of peptides of the TIPP (Tyr-Tic-Phe-Phe; Tic=1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid) family containing a guanidino (Guan) function in place of the N-terminal amino group were synthesized in an effort to improve their blood-brain barrier permeability. Unexpectedly, N-terminal amidination significantly altered the in vitro opioid activity profiles. Guan-analogues of TIPP-related δ opioid antagonists showed δ partial agonist or mixed δ partial agonist/µ partial agonist activity. Guanidinylation of the mixed µ agonist/δ antagonists H-Dmt-Tic-Phe-Phe-NH2 (DIPP-NH2) and H-Dmt-TicΨ[CH2NH]Phe-Phe-NH2 (DIPP-NH2[Ψ]) converted them to mixed µ agonist/δ agonists. A docking study revealed distinct positioning of DIPP-NH2 and Guan-DIPP-NH2 in the δ receptor binding site. Lys(3)-analogues of DIPP-NH2 and DIPP-NH2[Ψ] (guanidinylated or non-guanidinylated) turned out to be mixed µ/κ agonists with δ antagonist-, δ partial agonist- or δ full agonist activity. Compounds with some of the observed mixed opioid activity profiles have therapeutic potential as analgesics with reduced side effects or for treatment of cocaine addiction.


Subject(s)
Guanidines/chemistry , Oligopeptides/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Tetrahydroisoquinolines/pharmacology , Dose-Response Relationship, Drug , Models, Molecular , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Receptors, Opioid, delta/metabolism , Structure-Activity Relationship , Tetrahydroisoquinolines/chemical synthesis , Tetrahydroisoquinolines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL