Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Braz. J. Pharm. Sci. (Online) ; 58: e191093, 2022. tab, graf
Article in English | LILACS | ID: biblio-1383999

ABSTRACT

Abstract In recent years, improvements have been made, through biotechnological processes, in the production and development of peptides capable of increasing collagen and elastin synthesis for anti-aging skin care. However, proteins have many limitations due to their structural, chemical and physical fragility to external aggressions, which may cause conformational changes, leading to loss of biological activity. Therefore, it is important to create delivery systems that protect these biomolecules from damage, allowing them to reach their target. This work aimed to develop a system able to carry bovine serum albumin (BSA), used as a model of a protein, and to incorporate this system in a semisolid formulation suitable for skin application. A microemulgel based on a solid-in-oil-in-water (S/O/W) microemulsion was prepared. Firstly, the association efficiency (AE) of lyophilized BSA-sucrose ester complex and the size of S/O nanodispersion were assessed; then, the characterization and stability evaluation of the final semisolid formulation through evaluation of pH, texture and rheological behavior were performed. The average value of AE was 54.74% ± 2.17. It was possible to develop an S/O/W microemulsion, which allowed the subsequent development of an S/O/W microemulgel that assured suitable pH, texture and rheological characteristics for skin application.


Subject(s)
Serum Albumin, Bovine , Proteins/adverse effects , Collagen , Peptides/agonists , Skin/drug effects , Biological Products , Aging , Hydrogen-Ion Concentration
2.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34272276

ABSTRACT

CD8+ T cells are inherently cross-reactive and recognize numerous peptide antigens in the context of a given major histocompatibility complex class I (MHCI) molecule via the clonotypically expressed T cell receptor (TCR). The lineally expressed coreceptor CD8 interacts coordinately with MHCI at a distinct and largely invariant site to slow the TCR/peptide-MHCI (pMHCI) dissociation rate and enhance antigen sensitivity. However, this biological effect is not necessarily uniform, and theoretical models suggest that antigen sensitivity can be modulated in a differential manner by CD8. We used two intrinsically controlled systems to determine how the relationship between the TCR/pMHCI interaction and the pMHCI/CD8 interaction affects the functional sensitivity of antigen recognition. Our data show that modulation of the pMHCI/CD8 interaction can reorder the agonist hierarchy of peptide ligands across a spectrum of affinities for the TCR.


Subject(s)
CD8 Antigens/immunology , Peptides/agonists , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Antigens/chemistry , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Reactions , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Kinetics , Ligands , Lymphocyte Activation , Models, Immunological , Mutation
3.
Compr Physiol ; 10(1): 99-124, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31853954

ABSTRACT

Obesity is a global healthcare challenge that gives rise to devastating diseases such as the metabolic syndrome, type-2 diabetes (T2D), and a variety of cardiovascular diseases. The escalating prevalence of obesity has led to an increased interest in pharmacological options to counteract excess weight gain. Gastrointestinal hormones such as glucagon, amylin, and glucagon-like peptide-1 (GLP-1) are well recognized for influencing food intake and satiety, but the therapeutic potential of these native peptides is overall limited by a short half-life and an often dose-dependent appearance of unwanted effects. Recent clinical success of chemically optimized GLP-1 mimetics with improved pharmacokinetics and sustained action has propelled pharmacological interest in using bioengineered gut hormones to treat obesity and diabetes. In this article, we summarize the basic biology and signaling mechanisms of selected gut peptides and discuss how they regulate systemic energy and glucose metabolism. Subsequently, we focus on the design and evaluation of unimolecular drugs that combine the beneficial effects of selected gut hormones into a single entity to optimize the beneficial impact on systems metabolism. © 2020 American Physiological Society. Compr Physiol 10:99-124, 2020.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Hormones/agonists , Obesity/drug therapy , Peptides/agonists , Animals , Bariatric Surgery , Diabetes Mellitus, Type 2/metabolism , Gastrointestinal Hormones/metabolism , Humans , Obesity/metabolism , Obesity/surgery , Peptides/metabolism
4.
Bioorg Med Chem Lett ; 27(20): 4626-4629, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28935264

ABSTRACT

Neuromedin U (NMU) mediates various physiological functions via NMUR1 and NMUR2 receptors. NMUR2 has been considered a promising treatment option for diabetes and obesity. Although NMU-8, a shorter peptide, has potent agonist activity for both receptors, it is metabolically unstable. Therefore, NMU-8 analogs modified with long-chain alkyl moieties via a linker were synthesized. An octadecanoyl analog (17) with amino acid substitutions [αMePhe19, Nle21, and Arg(Me)24] and a linker [Tra-γGlu-PEG(2)] dramatically increased NMUR2 selectivity, with retention of high agonist activity. Subcutaneous administration of 17 induced anorectic activity in C57BL/6J mice. Owing to its high metabolic stability, 17 would be useful in clarifying the physiological role and therapeutic application of NMU.


Subject(s)
Appetite Depressants/metabolism , Peptides/metabolism , Receptors, Neurotransmitter/metabolism , Alkylation , Amino Acid Sequence , Animals , Appetite Depressants/chemistry , Appetite Depressants/pharmacology , Eating/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Peptides/agonists , Receptors, Neurotransmitter/antagonists & inhibitors , Structure-Activity Relationship
5.
PLoS One ; 12(8): e0182555, 2017.
Article in English | MEDLINE | ID: mdl-28771632

ABSTRACT

The emergence of multi-drug resistant Neisseria gonorrhoeae has generated an urgent need for novel therapies or a vaccine to prevent gonococcal disease. In this study we investigate the potential of targeting the surface exposed nitrite reductase, AniA, to block activity by producing functional blocking antibodies. AniA activity is essential for anaerobic growth and biofilm formation of N. gonorrhoeae and functional blocking antibodies may prevent colonisation and disease. Seven peptides covering regions adjacent to the active site were designed based on the AniA structure. Six of the seven peptide conjugates generated immune responses. Peptide 7, GALGQLKVEGAEN, was able to elicit antibodies capable of blocking AniA activity. Antiserum raised against the peptide 7 conjugate detected AniA in 20 N. gonorrhoeae clinical isolates. Recombinant AniA protein antigens were also assessed in this study and generated high-titre, functional blocking antibody responses. Peptide 7 conjugates or truncated recombinant AniA antigens have potential for inclusion in a vaccine against N. gonorrhoeae.


Subject(s)
Antibodies, Blocking/immunology , Antigens, Bacterial/chemistry , Bacterial Outer Membrane Proteins/chemistry , Neisseria gonorrhoeae/enzymology , Peptides/immunology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/chemistry , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Catalytic Domain , Female , Immunization , Mice , Neisseria gonorrhoeae/immunology , Peptides/agonists , Peptides/chemical synthesis , Rabbits , Recombinant Proteins/immunology
6.
Mol Cell Biochem ; 427(1-2): 13-22, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27995416

ABSTRACT

The aim of our study was to determine whether the use of cisplatin in the presence echistatin in MDA-MB-231 breast cancer cells leads to a reduction of toxic effects associated with the use of cisplatin. The expression of ß1-integrin and insulin-like growth factor 1 receptor (IGF-IR), signaling pathway protein expression: protein kinase B (AKT), mitogen-activated protein kinases (ERK1/ERK2), nuclear factor kappa B (NFκB), and caspase-3 and -9 activity was measured after 24 h of incubation with tested compounds to explain detailed molecular mechanism of induction of apoptosis. The viability of MDA-MB-231 breast cancer cells was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Annexin V-FITC/propidium iodide staining assay was performed to detect the induction of apoptosis. Inhibition DNA biosynthesis was determined by [3H]thymidine incorporation into DNA. The expression of of ß1-integrin, IGF-IR, AKT, ERK1/ERK2, NFκB, caspase-3 and -9 was evaluated using Western blot. The results suggest that treatment of MDA-MB-231 breast cancer cells for 24 h cisplatin plus echistatin severely inhibits cell growth and activates apoptosis by upregulation of caspase-3 and -9 expressions. The effect was stronger than treatment cisplatin and echistatin alone. In this study, we have found that cisplatin plus echistatin treatment decreases collagen biosynthesis in MDA-MB-231 breast cancer cells stronger than the individual compounds. The inhibition was found to be dependent on the ß1-integrin and IGF receptor activation. A significant reduction of ERK1/ERK2, AKT expression in cancer cells after cisplatin plus echistatin treatment was also found. The cancer cells treated by echistatin, cisplatin, and in particular the combination of both compounds drastically increased expression of NFκB transcription factor. Our results suggest that combined therapy cisplatin plus echistatin is a possible way to improve selectiveness of cisplatin. This mechanism probably is due to downregulation of expression of ß1-integrin and IGF-IR receptors, and the signaling pathway proteins induced by these receptors. Our results suggest that therapy cisplatin plus echistatin is a possible way to improve selectiveness of cisplatin.


Subject(s)
Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Peptides/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cisplatin/agonists , Drug Synergism , Female , Humans , Intercellular Signaling Peptides and Proteins , Neoplasm Proteins/metabolism , Peptides/agonists
7.
J Vis Exp ; (117)2016 11 10.
Article in English | MEDLINE | ID: mdl-27911362

ABSTRACT

As with small molecule drug discovery, screening for peptide agonists requires the serial dilution of peptides to produce concentration-response curves. Screening peptides affords an additional layer of complexity as conventional tip-based sample handling methods expose peptides to a large surface area of plasticware, providing an increased opportunity for peptide loss via adsorption. Preventing excessive exposure to plasticware reduces peptide loss via adherence to plastics and thus minimizes inaccuracies in potency prediction, and we have previously described the benefits of non-contact acoustic dispensing for in vitro high-throughput screening of peptide agonists1. Here we discuss a fully integrated automation solution for non-contact acoustic preparation of peptide serial dilutions in microtiter plates utilizing the example of screening for peptide agonists at the mouse glucagon-like peptide-1 receptor (GLP-1R). Our methods allow for high-throughput cell-based assays to screen for agonists and are easily scalable to support increased sample throughput, or to allow for increased numbers of assay plate copies (e.g., for a panel of more target cell lines).


Subject(s)
Biological Assay , Peptides/agonists , Acoustics , Animals , Automation , Mice
8.
Peptides ; 83: 49-56, 2016 09.
Article in English | MEDLINE | ID: mdl-27397853

ABSTRACT

The ability to design agonists that target peptide signaling is a strategy to delineate underlying mechanisms and influence biology. A sequence that uniquely characterizes a peptide provides a distinct site to generate novel agonists. Drosophila melanogaster sulfakinin encodes non-sulfated drosulfakinin I (nsDSK I; FDDYGHMRF-NH2) and nsDSK II (GGDDQFDDYGHMRF-NH2). Drosulfakinin is typical of sulfakinin precursors, which are conserved throughout invertebrates. Non-sulfated DSK II is structurally related to DSK I, however, it contains a unique 5-residue N-terminal extension; drosulfakinins signal through G-protein coupled receptors, DSK-R1 and DSK-R2. Drosulfakinin II distinctly influences adult and larval gut motility and larval locomotion; yet, its structure-activity relationship was unreported. We hypothesized substitution of an N-terminal extension residue may alter nsDSK II activity. By targeting the extension we identified, not unexpectedly, analogs mimicking nsDSK II, yet, surprisingly, we also discovered novel agonists with increased (super) and opposite (protean) effects. We determined [A3] nsDSK II increased larval gut contractility rather than, like nsDSK II, decrease it. [N4] nsDSK II impacted larval locomotion, although nsDSK II was inactive. In adult gut, [A1] nsDSK II, [A2] nsDSKII, and [A3] nsDSK II mimicked nsDSK II, and [A4] nsDSK II and [A5] nsDSK II were more potent; [N3] nsDSK II and [N4] nsDSK II mimicked nsDSK II. This study reports nsDSK II signals through DSK-R2 to influence gut motility and locomotion, identifying a novel role for the N-terminal extension in sulfakinin biology and receptor activation; it also led to the discovery of nsDSK II structural analogs that act as super and protean agonists.


Subject(s)
Drosophila Proteins/genetics , Locomotion/genetics , Peptides/agonists , Peptides/genetics , Receptors, G-Protein-Coupled/genetics , Amino Acid Sequence/genetics , Animals , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Intercellular Signaling Peptides and Proteins , Larva/drug effects , Larva/genetics , Locomotion/drug effects , Neuropeptides/chemistry , Neuropeptides/genetics , Oligopeptides/chemistry , Oligopeptides/genetics , Peptides/chemistry , Peptides/pharmacology , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
9.
Mol Pharmacol ; 90(3): 214-24, 2016 09.
Article in English | MEDLINE | ID: mdl-27338081

ABSTRACT

Adhesion G protein-coupled receptors (aGPCRs) have emerging roles in development and tissue maintenance and is the most prevalent GPCR subclass mutated in human cancers, but to date, no drugs have been developed to target them in any disease. aGPCR extracellular domains contain a conserved subdomain that mediates self-cleavage proximal to the start of the 7-transmembrane domain (7TM). The two receptor protomers, extracellular domain and amino terminal fragment (NTF), and the 7TM or C-terminal fragment remain noncovalently bound at the plasma membrane in a low-activity state. We recently demonstrated that NTF dissociation liberates the 7TM N-terminal stalk, which acts as a tethered-peptide agonist permitting receptor-dependent heterotrimeric G protein activation. In many cases, natural aGPCR ligands are extracellular matrix proteins that dissociate the NTF to reveal the tethered agonist. Given the perceived difficulty in modifying extracellular matrix proteins to create aGPCR probes, we developed a serum response element (SRE)-luciferase-based screening approach to identify GPR56/ADGRG1 small-molecule inhibitors. A 2000-compound library comprising known drugs and natural products was screened for GPR56-dependent SRE activation inhibitors that did not inhibit constitutively active Gα13-dependent SRE activation. Dihydromunduletone (DHM), a rotenoid derivative, was validated using cell-free aGPCR/heterotrimeric G protein guanosine 5'-3-O-(thio)triphosphate binding reconstitution assays. DHM inhibited GPR56 and GPR114/ADGRG5, which have similar tethered agonists, but not the aGPCR GPR110/ADGRF1, M3 muscarinic acetylcholine, or ß2 adrenergic GPCRs. DHM inhibited tethered peptide agonist-stimulated and synthetic peptide agonist-stimulated GPR56 but did not inhibit basal activity, demonstrating that it antagonizes the peptide agonist. DHM is a novel aGPCR antagonist and potentially useful chemical probe that may be developed as a future aGPCR therapeutic.


Subject(s)
Benzopyrans/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Retinoids/pharmacology , Small Molecule Libraries/pharmacology , Animals , Benzopyrans/analysis , Benzopyrans/chemistry , Cell Adhesion/drug effects , Flavonoids/pharmacology , HEK293 Cells , High-Throughput Screening Assays , Humans , Peptides/agonists , Peptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Reproducibility of Results , Retinoids/analysis , Retinoids/chemistry , Small Molecule Libraries/analysis , Small Molecule Libraries/chemistry
10.
Bioorg Med Chem ; 23(8): 1808-16, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25797164

ABSTRACT

In behavioral research, the sex peptide receptor in Drosophila melanogaster (DrmSPR) is the most interesting G protein-coupled receptor (GPCR) and is involved in post-mating responses such as increased egg-laying and decreased receptivity of the female; during these responses, the receptors are activated by a specific natural peptide agonist (sex peptide, SP). To discover small molecule agonists for DrmSPR, a compound library based on a pyrazolodiazepine scaffold, which was previously reported as a potential privileged structure, was screened. Structure-activity relationship (SAR) studies of the hit compounds, which exhibited weak agonistic effects (69-72% activation at 100µM), were explored through the synthesis of various analogs with substituents at the R1, R2, R3 and R4 positions of the pyrazolodiazepine skeleton. As a result, compounds 21 and 31 of the 6-benzyl pyrazolodiazepine derivative series were found to be small molecule agonists for DrmSPR with EC50 values of 3-4µM.


Subject(s)
Azepines/chemistry , Azepines/pharmacology , Drosophila Proteins/agonists , Peptides/agonists , Pyrazoles/chemistry , Pyrazoles/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Drosophila/drug effects , Drosophila/physiology , Drosophila Proteins/metabolism , Drug Discovery , Female , Male , Peptides/metabolism , Receptors, Peptide , Structure-Activity Relationship
11.
Biotechnol Prog ; 29(6): 1550-7, 2013.
Article in English | MEDLINE | ID: mdl-24014069

ABSTRACT

Human mesenchymal stem cells (MSCs) are attractive for use in cartilage tissue engineering. Cells are often seeded in a structural scaffold containing growth factors. Peptide mimics of full-length growth factors are a promising alternative because they are less expensive and easier to manufacture. We investigated four short peptides for their effect on the chondrogenesis of human MSCs. The peptides were originally designed to mimic bone morphogenetic protein-2 (BMP-2), transforming growth factor-beta 1 (TGF-ß1), and insulin, all of which have been shown to affect MSC chondrogenesis. Previous studies demonstrated that the peptides elicited bioactivity in other cell types, but the peptides have not been investigated for their effect on chondrogenesis in human MSCs. In a preliminary investigation, peptides were added to a pellet culture of human MSCs and assayed for their effect on glycosaminoglycan (GAG) production. These experiments determined peptide concentrations used in a full-factorial experiment to investigate any interactions. The experiment revealed the BMP peptide as a robust stimulant for GAG production. .


Subject(s)
Cell Differentiation/drug effects , Chondrogenesis/drug effects , Peptides/administration & dosage , Tissue Engineering , Bone Morphogenetic Protein 2/chemistry , Humans , Insulin/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Peptides/agonists , Peptides/chemistry , Transforming Growth Factor beta1/chemistry
12.
Biochem Pharmacol ; 83(5): 559-66, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22051078

ABSTRACT

Neuropeptides play a crucial role in the normal function of the central nervous system and peptide receptors hold great promise as therapeutic targets for the treatment of several CNS disorders. In general, the development of peptide therapeutics has been limited by the lack of drug-like properties of peptides and this has made it very difficult to transform them into marketable therapeutic molecules. Some of these challenges include poor in vivo stability, poor solubility, incompatibility with oral administration, shelf stability, cost of manufacture. Recent technical advances have overcome many of these limitations and have led to rapid growth in the development of peptides for a wide range of therapeutic indications such as diabetes, cancer and pain. This review examines the therapeutic potential of peptide agonists for the treatment of major CNS disorders such as schizophrenia, anxiety, depression and autism. Both clinical and preclinical data has been accumulated supporting the potential utility of agonists at central neurotensin, cholecystokinin, neuropeptide Y and oxytocin receptors. Some of the successful approaches that have been developed to increase the stability and longevity of peptides in vivo and improve their delivery are also described and potential strategies for overcoming the major challenge that is unique to CNS therapeutics, penetration of the blood-brain barrier, are discussed.


Subject(s)
Central Nervous System Diseases/drug therapy , Peptides/therapeutic use , Biological Transport , Blood-Brain Barrier/physiology , Humans , Peptides/agonists , Peptides/chemistry , Peptides/metabolism , Receptors, Peptide/metabolism
13.
J Immunol ; 187(3): 1475-85, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21709160

ABSTRACT

The peptide F2L was previously characterized as a high-affinity natural agonist for the human formyl peptide receptor (FPR) 3. F2L is an acetylated 21-aa peptide corresponding with the N terminus of the intracellular heme-binding protein 1 (HEBP1). In the current work, we have investigated which proteases were able to generate the F2L peptide from its precursor HEBP1. Structure-function analysis of F2L identified three amino acids, G(3), N(7), and S(8), as the most important for interaction of the peptide with FPR3. We expressed a C-terminally His-tagged form of human HEBP1 in yeast and purified it to homogeneity. The purified protein was used as substrate to identify proteases generating bioactive peptides for FPR3-expressing cells. A conditioned medium from human monocyte-derived macrophages was able to generate bioactivity from HEBP1, and this activity was inhibited by pepstatin A. Cathepsin D was characterized as the protease responsible for HEBP1 processing, and the bioactive product was identified as F2L. We have therefore determined how F2L, the specific agonist of FPR3, is generated from the intracellular protein HEBP1, although it is unknown in which compartment the processing by cathepsin D occurs in vivo.


Subject(s)
Carrier Proteins/metabolism , Cathepsin D/physiology , Chemotactic Factors/agonists , Hemeproteins/metabolism , Peptides/agonists , Protein Precursors/metabolism , Protein Processing, Post-Translational/immunology , Receptors, Formyl Peptide/metabolism , Amino Acid Sequence , Animals , CHO Cells , Carrier Proteins/biosynthesis , Cathepsin D/deficiency , Cells, Cultured , Chemotactic Factors/biosynthesis , Chemotactic Factors/metabolism , Cricetinae , Cricetulus , Heme-Binding Proteins , Hemeproteins/biosynthesis , Humans , Ligands , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , Peptides/metabolism , Protein Binding/immunology , Protein Precursors/biosynthesis , Receptors, Formyl Peptide/biosynthesis
14.
Neurosci Res ; 68(1): 35-43, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20562017

ABSTRACT

Hexafins are recently identified low-molecular-weight peptide agonists of the fibroblast growth factor receptor (FGFR), derived from the beta6-beta7 loop region of various FGFs. Synthetic hexafin peptides have been shown to bind to and induce tyrosine phosphorylation of FGFR1, stimulate neurite outgrowth, and promote neuronal survival in vitro. Thus, the pronounced biological activities of hexafins in vitro make them attractive compounds for pharmacological studies in vivo. The present study investigated the effects of subcutaneous administration of hexafin1 and hexafin2 (peptides derived from FGF1 and FGF2, respectively) on social memory, exploratory activity, and anxiety-like behavior in adult rats. Treatment with hexafin1 and hexafin2 resulted in prolonged retention of social memory. Furthermore, rats treated with hexafin2 exhibited decreased anxiety-like behavior in the elevated plus maze. Employing an R6/2 mouse model of Huntington's disease (HD), we found that although hexafin2 did not affect the progression of motor symptoms, it alleviated deficits in activity related to social behavior, including sociability and social novelty. Thus, hexafin2 may have therapeutic potential for the treatment of HD.


Subject(s)
Behavior, Animal/drug effects , Fibroblast Growth Factors/agonists , Peptides/agonists , Peptides/pharmacology , Receptors, Fibroblast Growth Factor/agonists , Animals , Behavior, Animal/physiology , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Fibroblast Growth Factor 1/chemical synthesis , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/chemical synthesis , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factors/physiology , Humans , Injections, Subcutaneous/methods , Male , Mice , Mice, Transgenic , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Peptides/chemical synthesis , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/physiology
15.
Proc Natl Acad Sci U S A ; 107(14): 6520-5, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20308537

ABSTRACT

Upon mating, females of many animal species undergo dramatic changes in their behavior. In Drosophila melanogaster, postmating behaviors are triggered by sex peptide (SP), which is produced in the male seminal fluid and transferred to female during copulation. SP modulates female behaviors via sex peptide receptor (SPR) located in a small subset of internal sensory neurons that innervate the female uterus and project to the CNS. Although required for postmating responses only in these female sensory neurons, SPR is expressed broadly in the CNS of both sexes. Moreover, SPR is also encoded in the genomes of insects that lack obvious SP orthologs. These observations suggest that SPR may have additional ligands and functions. Here, we identify myoinhibitory peptides (MIPs) as a second family of SPR ligands that is conserved across a wide range of invertebrate species. MIPs are potent agonists for Drosophila, Aedes, and Aplysia SPRs in vitro, yet are unable to trigger postmating responses in vivo. In contrast to SP, MIPs are not produced in male reproductive organs, and are not required for postmating behaviors in Drosophila females. We conclude that MIPs are evolutionarily conserved ligands for SPR, which are likely to mediate functions other than the regulation of female reproductive behaviors.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , CHO Cells , Central Nervous System/metabolism , Consummatory Behavior , Cricetinae , Cricetulus , Drosophila Proteins/agonists , Drosophila Proteins/genetics , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Female , Ligands , Male , Models, Molecular , Molecular Sequence Data , Peptides/agonists , Peptides/chemistry , Peptides/genetics , Phylogeny , Protein Structure, Tertiary , Receptors, Peptide , Sex Attractants/genetics , Sex Attractants/metabolism
16.
Br J Pharmacol ; 158(1): 361-71, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19719785

ABSTRACT

BACKGROUND AND PURPOSE: Proteinase-activated receptor 2 (PAR(2)) is a G-protein coupled receptor associated with many pathophysiological functions. To date, the development of PAR(2) antagonists has been limited. Here, we identify a number of novel peptide-mimetic PAR(2) antagonists and demonstrate inhibitory effects on PAR(2)-mediated intracellular signalling pathways and vascular responses. EXPERIMENTAL APPROACH: The peptide-mimetic compound library based on the structures of PAR(2) agonist peptides were screened for inhibition of PAR(2)-induced calcium mobilisation in human keratinocytes. Representative compounds were further evaluated by radioligand binding and inhibition of NFkappaB transcriptional activity and IL-8 production. The vascular effects of the antagonists were assessed using in vitro and in vivo models. KEY RESULTS: Two compounds, K-12940 and K-14585, significantly reduced SLIGKV-induced Ca(2+) mobilisation in primary human keratinocytes. Both K-12940 and K-14585 exhibited competitive inhibition for the binding of a high-affinity radiolabelled PAR(2)-ligand, [(3)H]-2-furoyl-LIGRL-NH(2), to human PAR(2) with K(i) values of 1.94 and 0.627 microM respectively. NFkappaB reporter activity and IL-8 production were also significantly reduced. Furthermore, relaxation of rat-isolated aorta induced by SLIGRL-NH(2) was inhibited competitively by K-14585. K-14585 also significantly lowered plasma extravasation in the dorsal skin of guinea pigs and reduced salivation in mice. CONCLUSIONS AND IMPLICATIONS: K-12940 and K-14585 antagonized PAR(2) competitively, resulting in inhibition of PAR(2)-mediated signalling and physiological responses both in vitro and in vivo. These peptide-mimetic PAR(2) antagonists could be useful in evaluating PAR(2)-mediated biological events and might lead to a new generation of therapeutically useful antagonists.


Subject(s)
Capillary Permeability/physiology , Keratinocytes/physiology , Oligopeptides/pharmacology , Peptides/antagonists & inhibitors , Peptides/physiology , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/physiology , Urea/analogs & derivatives , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Capillary Permeability/drug effects , Cell Line , Cells, Cultured , Guinea Pigs , Humans , In Vitro Techniques , Keratinocytes/drug effects , Keratinocytes/enzymology , Male , Mice , Molecular Mimicry , Peptides/agonists , Rats , Rats, Wistar , Receptor, PAR-2/agonists , Urea/pharmacology
17.
Scand J Immunol ; 70(3): 264-76, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19703016

ABSTRACT

HIV-infected individuals have an increased risk of invasive bacterial infections, even at early clinical stages with relatively normal CD4(+) T-cell counts. The pathogenic mechanisms behind this are not fully understood. However, an increasing number of studies indicate that HIV may impair the innate immunity to bacteria by infecting key cells of the monocyte/macrophage lineage. In this study, the effects of HIV infection on the protein profile of undifferentiated monocyte-like THP-1 cells were examined by a mass spectrometric approach based on stable isotope labelling with amino acid in cell culture (SILAC). We identified 651 proteins, of which nine proteins were down-regulated and 17 proteins were up-regulated in HIV-infected THP-1 cells as compared to uninfected controls. Most remarkably, the IL-1 receptor associated kinase 4 (IRAK-4), which is essential for virtually all TLR signalling, was suppressed, whereas the precursor for the antibiotic peptide Dermcidin was up-regulated in HIV-infected cells. Upon stimulation of either TLR2 or TLR4, the HIV-infected THP-1 cells displayed reduced TNF-alpha secretion. The HIV-induced down-regulation of IRAK-4 was reconfirmed in monocyte-derived macrophage cell cultures. These data suggests that HIV may impair the TLR signalling cascade for pathogen recognition in cells of the monocyte/macrophage lineage and thus, may reduce the ability of the innate immune system to sense invading pathogens and initiate appropriate responses.


Subject(s)
HIV Infections/immunology , HIV/pathogenicity , Interleukin-1 Receptor-Associated Kinases/metabolism , Macrophages/immunology , Monocytes/immunology , Peptides/metabolism , Cell Line , Down-Regulation/immunology , HIV Infections/virology , Humans , Immunity, Innate , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/immunology , Macrophages/metabolism , Macrophages/virology , Monocytes/metabolism , Monocytes/virology , Peptides/agonists , Peptides/immunology , Proteomics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/immunology
18.
J Biol Chem ; 284(11): 6885-92, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19136563

ABSTRACT

Aminoglycosides such as gentamicin have the ability to suppress translation termination at premature stop mutations, leading to a partial restoration of protein expression and function. This observation led to studies showing that this approach may provide a viable treatment for patients with genetic diseases such as cystic fibrosis that are caused by premature stop mutations. Although aminoglycoside treatment is sometimes associated with harmful side effects, several studies have shown that the co-administration of polyanions such as poly-L-aspartic acid (PAA) can both reduce toxicity and increase the intracellular aminoglycoside concentration. In the current study we examined how the co-administration of gentamicin with PAA influenced the readthrough of premature stop codons in cultured cells and a cystic fibrosis mouse model. Using a dual luciferase readthrough reporter system in cultured cells, we found that the co-administration of gentamicin with PAA increased readthrough 20-40% relative to cells treated with the same concentration of gentamicin alone. Using a Cftr-/- hCFTR-G542X mouse model, we found that PAA also increased the in vivo nonsense suppression induced by gentamicin. Following the withdrawal of gentamicin, PAA significantly prolonged the time interval during which readthrough could be detected, as shown by short circuit current measurements and immunofluorescence. Because the use of gentamicin to suppress disease-causing nonsense mutations will require their long term administration, the ability of PAA to reduce toxicity and increase both the level and duration of readthrough has important implications for this promising therapeutic approach.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/drug therapy , Gentamicins/pharmacology , Mutation, Missense , Peptides/pharmacology , Protein Synthesis Inhibitors/pharmacology , Amino Acid Substitution , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Drug Synergism , Gentamicins/agonists , Gentamicins/therapeutic use , Humans , Mice , Mice, Inbred CFTR , Mice, Knockout , Peptides/agonists , Peptides/therapeutic use , Protein Synthesis Inhibitors/agonists , Protein Synthesis Inhibitors/therapeutic use
19.
Mol Pharmacol ; 73(2): 431-40, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17965196

ABSTRACT

Natriuretic peptide receptor (NPR) A is composed of an extracellular domain (ECD) with a ligand binding site, a single transmembrane region, a kinase homology domain, and a guanylyl cyclase domain. The natural agonists atrial and brain natriuretic peptides (ANP, BNP) bind and activate NPRA, leading to cyclic GMP production, which is responsible for their role in cardiovascular homeostasis. Previous studies suggested that stabilization of a dimeric form of NPRA by agonist is essential for receptor activation. However, ligand specificity and sequential steps of this dimerization process have not been investigated. We used radioligand binding, fluorescence resonance energy transfer homoquenching, and molecular modeling to characterize the interaction of human NPRA-ECD with ANP, BNP, the superagonist (Arg(10),Leu(12),Ser(17),Leu(18))-rANP-(1-28), the minimized analog mini-ANP and the antagonist (Arg(6),beta-cyclohexyl-Ala(8),d-Tic(16),Arg(17),Cys(18))-rANP-(6-18)-amide (A71915). ANP binds to preformed ECD dimers and spontaneous dimerization is the rate-limiting step of the ligand binding process. All the studied peptides, including A71915 antagonist, induce a dose-dependent fluorescence homoquenching, specific to dimerization, with potencies highly correlated with their binding affinities. A71915 induced more quenching than other peptides, suggesting stabilization by the antagonist of ECD dimer in a distinct inactive conformation. In summary, these results indicate that the ligand-induced dimerization process of NPRA is different from that for cytokine receptor model. Agonists or antagonists bind to preformed dimeric ECD, leading to dimer stabilization in an active or inactive conformation, respectively. Furthermore, the highly sensitive fluorescence assay designed to assess dimerization could serve as a powerful tool for further detailing the kinetic steps involved in natriuretic peptide receptor binding and activation.


Subject(s)
Atrial Natriuretic Factor/physiology , Extracellular Fluid/metabolism , Guanylate Cyclase/metabolism , Peptides/metabolism , Receptors, Atrial Natriuretic Factor/agonists , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Binding Sites/physiology , Cell Line , Dimerization , Extracellular Fluid/chemistry , Extracellular Fluid/physiology , Guanylate Cyclase/chemistry , Humans , Insecta , Peptides/agonists , Peptides/physiology , Protein Structure, Tertiary , Receptors, Atrial Natriuretic Factor/chemistry
20.
J Immunol ; 179(4): 2556-64, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17675518

ABSTRACT

Allergic airways disease is initiated and perpetuated by an aberrant Th2 inflammatory response regulated in part by the cytokines IL-4 and IL-13, each of which induces activation of the STAT-6 transcription factor. Data from murine models indicate that the clinical manifestations of acute asthma are STAT-6 dependent, and thus, STAT-6 is a target for drug development in allergic airways disease. We designed a novel chimeric peptide (STAT-6 inhibitory peptide (STAT-6-IP)) comprised of a sequence predicted to bind to and inhibit STAT-6, fused to a protein transduction domain, to facilitate cellular uptake of the STAT-6-binding peptide. Our data demonstrate that the STAT-6-IP inhibited OVA-induced production of Th2 cytokines IL-4 and IL-13 in vitro. In contrast, the STAT-6-IP did not affect production of IFN-gamma, demonstrating specificity for Th2 cytokine inhibition. Following intranasal administration, the STAT-6-IP was localized to epithelial cells in the airways. Finally, in in vivo murine models of allergic rhinitis and asthma, intranasal delivery of the STAT-6-IP inhibited OVA-induced lung inflammation and mucus production as well as accumulation of eosinophils and IL-13 in bronchoalveolar lavage fluid and OVA-dependent airway hyperresponsiveness. Together these data show that local application of cell-penetrating peptide inhibitors of STAT-6 has significant potential for the treatment of allergic rhinitis and asthma.


Subject(s)
Asthma/drug therapy , Peptides/agonists , Rhinitis, Allergic, Perennial/drug therapy , STAT6 Transcription Factor/administration & dosage , STAT6 Transcription Factor/antagonists & inhibitors , Acute Disease , Administration, Intranasal , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/pathology , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Eosinophils/immunology , Eosinophils/pathology , Interleukin-13/immunology , Interleukin-4/immunology , Mice , Mucus/immunology , Ovalbumin/toxicity , Peptides/genetics , Peptides/immunology , Pneumonia/chemically induced , Pneumonia/drug therapy , Pneumonia/immunology , Pneumonia/pathology , Protein Binding/drug effects , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Rhinitis, Allergic, Perennial/chemically induced , Rhinitis, Allergic, Perennial/immunology , Rhinitis, Allergic, Perennial/pathology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , Th2 Cells/immunology , Th2 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...