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










Publication year range
1.
J Biol Chem ; 295(29): 9959-9973, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32482890

ABSTRACT

Intracellular collagen assembly begins with the oxidative folding of ∼30-kDa C-terminal propeptide (C-Pro) domains. Folded C-Pro domains then template the formation of triple helices between appropriate partner strands. Numerous C-Pro missense variants that disrupt or delay triple-helix formation are known to cause disease, but our understanding of the specific proteostasis defects introduced by these variants remains immature. Moreover, it is unclear whether or not recognition and quality control of misfolded C-Pro domains is mediated by recognizing stalled assembly of triple-helical domains or by direct engagement of the C-Pro itself. Here, we integrate biochemical and cellular approaches to illuminate the proteostasis defects associated with osteogenesis imperfecta-causing mutations within the collagen-α2(I) C-Pro domain. We first show that "C-Pro-only" constructs recapitulate key aspects of the behavior of full-length Colα2(I) constructs. Of the variants studied, perhaps the most severe assembly defects are associated with C1163R C-Proα2(I), which is incapable of forming stable trimers and is retained within cells. We find that the presence or absence of an unassembled triple-helical domain is not the key feature driving cellular retention versus secretion. Rather, the proteostasis network directly engages the misfolded C-Pro domain itself to prevent secretion and initiate clearance. Using MS-based proteomics, we elucidate how the endoplasmic reticulum (ER) proteostasis network differentially engages misfolded C1163R C-Proα2(I) and targets it for ER-associated degradation. These results provide insights into collagen folding and quality control with the potential to inform the design of proteostasis network-targeted strategies for managing collagenopathies.


Subject(s)
Collagen Type I/metabolism , Endoplasmic Reticulum/metabolism , Mutation , Osteogenesis Imperfecta/metabolism , Proteostasis , Collagen Type I/genetics , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , Osteogenesis Imperfecta/genetics , Protein Domains
2.
Methods Mol Biol ; 1944: 95-114, 2019.
Article in English | MEDLINE | ID: mdl-30840237

ABSTRACT

We present the development, optimization, and application of constructs, cell lines, covalent cross-linking methods, and immunoprecipitation strategies that enable robust and accurate determination of collagen interactomes via mass spectrometry-based proteomics. Using collagen type-I as an example, protocols for working with large, repetitive, and GC-rich collagen genes are described, followed by strategies for engineering cells that stably and inducibly express antibody epitope-tagged collagen-I. Detailed steps to optimize collagen interactome cross-linking and perform immunoprecipitations are then presented. We conclude with a discussion of methods to elute collagen interactomes and prepare samples for mass spectrometry-mediated identification of interactors. Throughout, caveats and potential problems researchers may encounter when working with collagen are discussed. We note that the protocols presented herein may be readily adapted to define interactomes of other collagen types, as well as to determine comparative interactomes of normal and disease-causing collagen variants using quantitative isotopic labeling (SILAC)- or isobaric mass tags (iTRAQ or TMT)-based mass spectrometry analysis.


Subject(s)
Collagen/metabolism , Intracellular Space/metabolism , Isotope Labeling/methods , Mass Spectrometry/methods , Protein Interaction Mapping/methods , Proteomics/methods , Collagen/analysis , Humans , Immunoprecipitation
3.
J Am Chem Soc ; 140(51): 18093-18103, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30427676

ABSTRACT

The discovery and optimization of biomolecules that reliably function in metazoan cells is imperative for both the study of basic biology and the treatment of disease. We describe the development, characterization, and proof-of-concept application of a platform for directed evolution of diverse biomolecules of interest (BOIs) directly in human cells. The platform relies on a custom-designed adenovirus variant lacking multiple genes, including the essential DNA polymerase and protease genes, features that allow us to evolve BOIs encoded by genes as large as 7 kb while attaining the mutation rates and enforcing the selection pressure required for successful directed evolution. High mutagenesis rates are continuously attained by trans-complementation of a newly engineered, highly error-prone form of the adenoviral polymerase. Selection pressure that couples desired BOI functions to adenoviral propagation is achieved by linking the functionality of the encoded BOI to the production of adenoviral protease activity by the human cell. The dynamic range for directed evolution can be enhanced to several orders of magnitude via application of a small-molecule adenoviral protease inhibitor to modulate selection pressure during directed evolution experiments. This platform makes it possible, in principle, to evolve any biomolecule activity that can be coupled to adenoviral protease expression or activation by simply serially passaging adenoviral populations carrying the BOI. As proof-of-concept, we use the platform to evolve, directly in the human cell environment, several transcription factor variants that maintain high levels of function while gaining resistance to a small-molecule inhibitor. We anticipate that this platform will substantially expand the repertoire of biomolecules that can be reliably and robustly engineered for both research and therapeutic applications in metazoan systems.


Subject(s)
Directed Molecular Evolution/methods , Transcription Factors/metabolism , Adenoviridae/genetics , Bacillus Phages/enzymology , DNA-Directed DNA Polymerase/genetics , Doxorubicin/pharmacology , Drug Resistance/genetics , HEK293 Cells , Humans , Integrases/genetics , Leucine-tRNA Ligase/genetics , Mutagenesis , Peptide Hydrolases/genetics , Proof of Concept Study , Protein Engineering , Transcription Factors/genetics , Viral Proteins/genetics
4.
Biochemistry ; 57(19): 2814-2827, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29676157

ABSTRACT

Collagen overproduction is a feature of fibrosis and cancer, while insufficient deposition of functional collagen molecules and/or the secretion of malformed collagen is common in genetic disorders like osteogenesis imperfecta. Collagen secretion is an appealing therapeutic target in these and other diseases, as secretion directly connects intracellular biosynthesis to collagen deposition and biological function in the extracellular matrix. However, small molecule and biological methods to tune collagen secretion are severely lacking. Their discovery could prove useful not only in the treatment of disease, but also in providing tools for better elucidating mechanisms of collagen biosynthesis. We developed a cell-based, high-throughput luminescent assay of collagen type I secretion and used it to screen for small molecules that selectively enhance or inhibit that process. Among several validated hits, the Hsp90 inhibitor 17-allylaminogeldanamycin (17-AAG) robustly decreases the secretion of collagen-I by our model cell line and by human primary cells. In these systems, 17-AAG and other pan-isoform Hsp90 inhibitors reduce collagen-I secretion post-translationally and are not global inhibitors of protein secretion. Surprisingly, the consequences of Hsp90 inhibitors cannot be attributed to inhibition of the endoplasmic reticulum's Hsp90 isoform, Grp94. Instead, collagen-I secretion likely depends on the activity of cytosolic Hsp90 chaperones, even though such chaperones cannot directly engage nascent collagen molecules. Our results highlight the value of a cell-based high-throughput screen for selective modulators of collagen secretion and suggest an unanticipated role for cytosolic Hsp90 in collagen secretion.


Subject(s)
Collagen Type I/chemistry , HSP90 Heat-Shock Proteins/chemistry , High-Throughput Screening Assays , Membrane Glycoproteins/chemistry , Benzoquinones/pharmacology , Cell Line , Collagen Type I/biosynthesis , Collagen Type I/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Protein Isoforms/chemistry
5.
Curr Top Microbiol Immunol ; 414: 1-25, 2018.
Article in English | MEDLINE | ID: mdl-28929194

ABSTRACT

Cells address challenges to protein folding in the secretory pathway by engaging endoplasmic reticulum (ER)-localized protective mechanisms that are collectively termed the unfolded protein response (UPR). By the action of the transmembrane signal transducers IRE1, PERK, and ATF6, the UPR induces networks of genes whose products alleviate the burden of protein misfolding. The UPR also plays instructive roles in cell differentiation and development, aids in the response to pathogens, and coordinates the output of professional secretory cells. These functions add to and move beyond the UPR's classical role in addressing proteotoxic stress. Thus, the UPR is not just a reaction to protein misfolding, but also a fundamental driving force in physiology and pathology. Recent efforts have yielded a suite of chemical genetic methods and small molecule modulators that now provide researchers with both stress-dependent and -independent control of UPR activity. Such tools provide new opportunities to perturb the UPR and thereby study mechanisms for maintaining proteostasis in the secretory pathway. Numerous observations now hint at the therapeutic potential of UPR modulation for diseases related to the misfolding and aggregation of ER client proteins. Growing evidence also indicates the promise of targeting ER proteostasis nodes downstream of the UPR. Here, we review selected advances in these areas, providing a resource to inform ongoing studies of secretory proteostasis and function as they relate to the UPR.


Subject(s)
Proteostasis/physiology , Unfolded Protein Response/physiology , Animals , Endoplasmic Reticulum/metabolism , Humans , Protein Folding
6.
J Pept Sci ; 23(4): 266-271, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28220557

ABSTRACT

An aza-amino acid scan of peptide inhibitors of the chromobox homolog 7 (CBX7) was performed to study the conformational requirements for affinity to the methyllysine reader protein. Twelve azapeptide analogues were prepared using three different approaches employing respectively N-(Fmoc)aza-amino acid chlorides and submonomer azapeptide synthesis to install systematically aza-residues at the first four residues of the peptide, as well as to provide aza-lysine residues possessing saturated and unsaturated side chains. The aza-peptide ligands were evaluated in a chromobox homolog 7 binding assay, providing useful insight into structural requirements for affinity. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Amino Acids/pharmacology , Aza Compounds/pharmacology , Peptides/pharmacology , Polycomb Repressive Complex 1/antagonists & inhibitors , Amino Acids/chemistry , Aza Compounds/chemistry , Humans , Ligands , Molecular Conformation , Peptides/chemical synthesis , Peptides/chemistry , Polycomb Repressive Complex 1/metabolism
7.
J Immunol ; 198(5): 2047-2062, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28148737

ABSTRACT

Preterm birth (PTB) is commonly accompanied by in utero fetal inflammation, and existing tocolytic drugs do not target fetal inflammatory injury. Of the candidate proinflammatory mediators, IL-1 appears central and is sufficient to trigger fetal loss. Therefore, we elucidated the effects of antenatal IL-1 exposure on postnatal development and investigated two IL-1 receptor antagonists, the competitive inhibitor anakinra (Kineret) and a potent noncompetitive inhibitor 101.10, for efficacy in blocking IL-1 actions. Antenatal exposure to IL-1ß induced Tnfa, Il6, Ccl2, Pghs2, and Mpges1 expression in placenta and fetal membranes, and it elevated amniotic fluid IL-1ß, IL-6, IL-8, and PGF2α, resulting in PTB and marked neonatal mortality. Surviving neonates had increased Il1b, Il6, Il8, Il10, Pghs2, Tnfa, and Crp expression in WBCs, elevated plasma levels of IL-1ß, IL-6, and IL-8, increased IL-1ß, IL-6, and IL-8 in fetal lung, intestine, and brain, and morphological abnormalities: e.g., disrupted lung alveolarization, atrophy of intestinal villus and colon-resident lymphoid follicle, and degeneration and atrophy of brain microvasculature with visual evoked potential anomalies. Late gestation treatment with 101.10 abolished these adverse outcomes, whereas Kineret exerted only modest effects and no benefit for gestation length, neonatal mortality, or placental inflammation. In a LPS-induced model of infection-associated PTB, 101.10 prevented PTB, neonatal mortality, and fetal brain inflammation. There was no substantive deviation in postnatal growth trajectory or adult body morphometry after antenatal 101.10 treatment. The results implicate IL-1 as an important driver of neonatal morbidity in PTB and identify 101.10 as a safe and effective candidate therapeutic.


Subject(s)
Brain/immunology , Fetal Development/drug effects , Inflammation/immunology , Interleukin-1beta/immunology , Placenta/immunology , Pregnancy/immunology , Premature Birth/immunology , Animals , Animals, Newborn , Brain/drug effects , Disease Models, Animal , Female , Humans , Inflammation/drug therapy , Inflammation Mediators/metabolism , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1beta/antagonists & inhibitors , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Peptides/therapeutic use , Placenta/drug effects , Premature Birth/drug therapy
8.
ACS Chem Biol ; 11(5): 1408-21, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26848503

ABSTRACT

Collagen-I is the most abundant protein in the human body, yet our understanding of how the endoplasmic reticulum regulates collagen-I proteostasis (folding, quality control, and secretion) remains immature. Of particular importance, interactomic studies to map the collagen-I proteostasis network have never been performed. Such studies would provide insight into mechanisms of collagen-I folding and misfolding in cells, an area that is particularly important owing to the prominence of the collagen misfolding-related diseases. Here, we overcome key roadblocks to progress in this area by generating stable fibrosarcoma cells that inducibly express properly folded and modified collagen-I strands tagged with distinctive antibody epitopes. Selective immunoprecipitation of collagen-I from these cells integrated with quantitative mass spectrometry-based proteomics permits the first mapping of the collagen-I proteostasis network. Biochemical validation of the resulting map leads to the assignment of numerous new players in collagen-I proteostasis, and the unanticipated discovery of apparent aspartyl-hydroxylation as a new post-translational modification in the N-propeptide of collagen-I. Furthermore, quantitative analyses reveal that Erp29, an abundant endoplasmic reticulum proteostasis machinery component with few known functions, plays a key role in collagen-I retention under ascorbate-deficient conditions. In summary, the work here provides fresh insights into the molecular mechanisms of collagen-I proteostasis, yielding a detailed roadmap for future investigations. Straightforward adaptations of the cellular platform developed will also enable hypothesis-driven, comparative research on the likely distinctive proteostasis mechanisms engaged by normal and disease-causing, misfolding collagen-I variants, potentially motivating new therapeutic strategies for currently incurable collagenopathies.


Subject(s)
Collagen Type I/metabolism , Protein Folding , Protein Interaction Maps , Cell Line , Collagen Type I/chemistry , Collagen Type I, alpha 1 Chain , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Humans , Mass Spectrometry/methods , Models, Molecular , Protein Interaction Mapping/methods , Protein Processing, Post-Translational , Proteomics/methods , Proteostasis Deficiencies/metabolism
9.
Neuropharmacology ; 108: 440-50, 2016 09.
Article in English | MEDLINE | ID: mdl-26006268

ABSTRACT

Parkinson's disease (PD) is characterized by a steady loss of dopamine neurons through apoptotic, inflammatory and oxidative stress processes. In that line of view, the pituitary adenylate cyclase-activating polypeptide (PACAP), with its ability to cross the blood-brain barrier and its anti-apoptotic, anti-inflammatory and anti-oxidative properties, has proven to offer potent neuroprotection in various PD models. Nonetheless, its peripheral actions, paired with low metabolic stability, hampered its clinical use. We have developed Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27) as an improved PACAP-derived neuroprotective compound. In vitro, this analog stimulated cAMP production, maintained mitochondrial potential and protected SH-SY5Y neuroblastoma cells from 1-methyl-4-phenylpyridinium (MPP(+)) toxicity, as potently as PACAP. Furthermore, contrasting with PACAP, it is stable in human plasma and against dipeptidyl peptidase IV activity. When injected intravenously to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice, PACAP and Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27) restored tyrosine hydoxylase expression into the substantia nigra and modulated the inflammatory response. Albeit falls of mean arterial pressure (MAP) were observed with both PACAP- and Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27)-treated mice, the intensity of the decrease as well as its duration were significantly less marked after iv injections of the analog than after those of the native polypeptide. Moreover, no significant changes in heart rate were measured with the animals for both compounds. Thus, Ac-[Phe(pI)(6), Nle(17)]PACAP(1-27) appears as a promising lead molecule for the development of PACAP-derived drugs potentially useful for the treatment of PD or other neurodegenerative diseases.


Subject(s)
Cardiovascular Diseases/prevention & control , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Animals , CHO Cells , Cardiovascular Diseases/chemically induced , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology
10.
Eur J Med Chem ; 104: 106-14, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26448038

ABSTRACT

Class B G protein-coupled receptors are activated by their cognate ligands following a two-step binding model involving a specific network of ligand-receptor intermolecular interactions. In particular, a N-capping structure present in the ligand would contribute significantly to position the N-terminal segment of the ligand once bound to its receptor. The aim of the current study was to implement the use of Pd-catalyzed Sonogashira coupling for the investigation of this structural motif. First, we have developed and evaluated various Sonogashira-based procedures for on-resin post-synthesis modification using a Leu-enkephalin derivative as a model peptide. Next, we have prepared a small library of PACAP-based analogs and evaluated the pharmacological profile of a few of them using a competitive binding assay, as well as functional and survival assays. Notably, our results suggest that the modification of the N-capping region could alter the binding specificity of PACAP without altering its biological activity, thereby opening the way for the design of more selective compounds. Finally, the possibility to achieve sequential multiple point substitutions via the Sonogashira cross-coupling method, during solid phase peptide synthesis, was also evaluated. Altogether, we demonstrated the versatility of such a procedure for the incorporation of various mono- and multiple alkyne-derived modifications during solid phase peptide synthesis and confirmed its usefulness for the structure-activity study of a class B GPCR ligand.


Subject(s)
Organometallic Compounds/chemistry , Palladium/chemistry , Peptides/chemistry , Peptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , Catalysis , Cell Line, Tumor , Cell Survival/drug effects , Cricetulus , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Peptides/chemical synthesis , Structure-Activity Relationship
11.
Biopolymers ; 104(5): 629-35, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25787802

ABSTRACT

The conformation of the N-amino-imidazolidin-2-one (Aid) peptidomimetic was investigated using NMR spectroscopy and X-ray crystallography. In solution, the tetrapeptide model p-bromobenzoyl-Aid-l-Phe-N'-iso-propylamide (1) exhibited shielded and solvent exposed amide protons indicative of a turn conformation. In the crystal lattice of 1, four turn conformers were present in the unit cell differing primarily by the ring puckering of the Aid residue. Two pairs of ß-turn conformers were observed each possessing an intramolecular ten-member hydrogen bond between the benzamide carbonyl and iso-propylamine NH moieties. The pairs had types II and II' ß-turn geometry that differed slightly about the ϕ and ψ torsion angles. Moreover, the X-ray analysis of 1 has been compared with that of the related unsaturated N-amino-imidazolin-2-one (Nai) analogs indicating the influences of ring flattening and substituents on conformation. Insertion of the preorganized Aid structure into biologically active peptides has thus been shown to offer a potentially valuable means for exploring the importance of turn geometry for activity, particularly in drug discovery.


Subject(s)
Imidazolidines/chemistry , Molecular Mimicry , Crystallography, X-Ray
12.
J Pept Sci ; 21(5): 387-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25400083

ABSTRACT

The solid-phase synthesis of azapeptides possessing a C-terminal aza-residue has been accomplished by a protocol featuring regioselective alkylation of benzhydrylidene-aza-glycinamide and illustrated by the syntheses of [aza-Lys(6)] growth-hormone-releasing peptide-6 analogs.


Subject(s)
Aza Compounds/chemistry , Peptides/chemical synthesis , Alkylation , Animals , Humans , Molecular Structure , Peptides/chemistry , Structure-Activity Relationship
13.
Org Lett ; 16(13): 3588-91, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24959890

ABSTRACT

Aza-peptides with basic amino acid residues (lysine, ornithine, arginine) and derivatives were synthesized by an effective approach featuring alkylation of a hydrazone-protected aza-glycine residue with α-bromo ω-chloro propane and butane to provide the corresponding alkyl chloride side chains. Displacement of the chloride with azide and various amines gave entry to azaOrn, azaLys, and azaArg containing peptides as demonstrated by the solution and solid-phase syntheses of 29 examples, including an aza-library of Growth Hormone Releasing Peptide-6 analogs.


Subject(s)
Amino Acids, Basic/chemistry , Arginine/chemistry , Aza Compounds/chemical synthesis , Lysine/chemistry , Oligopeptides/chemical synthesis , Ornithine/chemistry , Peptides/chemical synthesis , Aza Compounds/chemistry , Molecular Structure , Oligopeptides/chemistry , Peptides/chemistry , Solid-Phase Synthesis Techniques
14.
ACS Appl Mater Interfaces ; 6(12): 9085-92, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24870015

ABSTRACT

Although the conventional methods for strong attachment of chitosan onto stainless steel require many steps in different solvents, it has been demonstrated in this work that covalent grafting of chitosan on a steel surface can be easily achieved through the formation of a self-adhesive surface based on aryldiazonium seed layers. Initially, a polyaminophenyl layer is grafted on a stainless steel surface by means of the one-step GraftFast(TM) process (diazonium induced anchoring process). The grafted aminophenyl groups are then converted to an aryldiazonium seed layer by simply dipping the substrate in a sodium nitrite acidic solution. That diazonium-rich grafted layer can be used as a self-adhesive surface for subsequent spontaneous coating of chitosan onto the steel surface. X-ray photoelectron and impedance electrochemical spectroscopies were used to characterize the pristine and modified steel samples. As evidenced from impedance and linear polarization results, the primary polyaminophenyl layer characterized by a high charge transfer resistance contributed to better protection against corrosion of the resulting chitosan-coated steel in sulfuric acid medium.

15.
Org Lett ; 16(8): 2232-5, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24697286

ABSTRACT

The synthesis of N-aminoimidazolidin-2-one (Aid) peptidomimetics has been achieved by alkylation of the urea nitrogen of a semicarbazone residue using ethylene bromide. The Aid scaffold combines electronic and structural constraints to rigidify the peptide backbone in the equivalent of an aza variant of a Freidinger-Veber lactam. The syntheses and isolation of 25 Aid peptides, including eight GHRP-6 analogues, are reported to demonstrate the utility of this method for controlling conformation.


Subject(s)
Peptides/chemistry , Peptides/chemical synthesis , Semicarbazones/chemistry , Alkylation , Amino Acid Sequence , Aza Compounds/chemical synthesis , Aza Compounds/chemistry , Chemistry, Organic/methods , Lactams/chemical synthesis , Lactams/chemistry , Molecular Conformation , Molecular Structure , Oligopeptides/chemistry , Peptidomimetics
16.
J Control Release ; 163(2): 256-65, 2012 Oct 28.
Article in English | MEDLINE | ID: mdl-22922050

ABSTRACT

The discovery of cell-penetrating peptide opened up new promising avenues for the non-invasive delivery of non-permeable biomolecules within the intracellular compartment. However, some setbacks such as possible toxic effects or unexpected immunological responses have limited their use in clinic. To overcome these obstacles, we investigated the use of novel cell-penetrating peptides (CPPs) derived from the endogenous neuropeptide Pituitary adenylate cyclase-activating polypeptide (PACAP). First, we demonstrated the propensity of native PACAP isoforms (PACAP27 and PACAP38) to efficiently deliver a large and non-permeable molecule, i.e. streptavidin, into cells. An inactive modified fragment of PACAP38, i.e. [Arg(17)]PACAP(11-38), with preserved cell-penetrating physico-chemical properties, was also synthesized and successfully use for the intracellular delivery of various cargoes such as small molecules, peptides, proteins, and polynucleotides. Especially, its effectiveness as a transfection agent was comparable to Lipofectamine 2000 while being non-toxic for cells. Uptake mechanism studies demonstrated that direct translocation, caveolae-dependent endocytosis and macropinocytosis were involved in the internalization of [Arg(17)]PACAP(11-38). This study not only opened up a new aspect in the usefulness of PACAP and its derivatives for therapeutic application but also contributed to the identification of new members of the CPP family. As such, inactive PACAP-related analogs could represent excellent vectors for in vitro and in vivo applications.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Transfection/methods , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Design , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Plasmids
17.
Biochim Biophys Acta ; 1823(4): 940-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22343001

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP), a hypophysiotropic neurohormone, participates in the regulation of pleiotropic functions. The recent discovery of intracellular PACAP receptors in the brain and the testis as well as the physico-chemical characteristics of PACAP, i.e. extended α-helix containing basic residues, prompted us to evaluate the propensity of PACAP to cross the plasma membrane in a receptor-independent manner. Using confocal microscopy and flow cytometry, we demonstrated the ability of FITC-conjugated PACAP to efficiently penetrate into the internal cell compartment by direct translocation and endocytosis through clathrin-coated pits and macropinocytosis. Our study also revealed that, once inside the cells, PACAP38 is not entirely degraded by intracellular enzymes and that a significant amount of intact PACAP38 is also able to exit cells. Moreover, using binding assay on rat nuclear fractions from various tissues, PACAP nuclear receptors were identified. We also found that PACAP stimulates calcium release in rat testis nuclei. Interestingly, PACAP27 and PACAP38 but not VIP were able to upregulate de novo DNA synthesis in testis nuclei and that this effect was abolished by PACAP(6-38). These results support the presence of PAC1 receptors at the nuclear membrane and raise questions about their role in the biological activity of the peptide. These findings contribute to the characterization of PACAP as an intracrine factor and suggest that these intracellular PAC1 binding sites, probably associated with specific biological activities, should be taken into account during the development of PACAP-based drugs.


Subject(s)
Endocytosis , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Survival , Cytosol/metabolism , Fluorescein-5-isothiocyanate/metabolism , Humans , Male , Mass Spectrometry , Molecular Sequence Data , Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Testis/cytology , Testis/metabolism , Transcription, Genetic
18.
Biochimie ; 93(4): 669-77, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21185349

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts many crucial biological functions through the interaction with its specific PAC1 receptor (PAC1-R), a class B G protein-coupled receptor (GPCR). To identify the binding sites of PACAP in the PAC1-R, three peptide derivatives containing a photoreactive p-benzoyl-phenylalanine (Bpa) residue were developed. These photosensitive PACAP analogs were fully biologically active and competent to displace radiolabeled Ac-PACAP27 from the PAC1-R. Subsequently, the (125)I-labeled photoprobes were used to anchor the PAC1-R expressed in Chinese hamster ovary cells. Photolabeling led to the formation of two protein complexes of 76 and 67 kDa, representing different glycosylated forms of the receptor. Proteinase and chemical cleavages of the peptide-receptor complexes revealed that (125)I[Bpa(0), Nle(17)]PACAP27, (125)I[Bpa(6), Nle(17)]PACAP27 and (125)I[Nle(17), Bpa(22)]PACAP27 covalently labeled the Ser(98) - Met(111) segment, the Ser(124) - Glu(125) dipeptide and the Ser(141) - Met(172) fragment, respectively. Taking into account the topology of the PAC1-R, these segments are mainly located within the extracellular N-terminal domain, indicating that this PAC1-R domain is the major binding site of PACAP27. The present study constitutes the first characterization of the binding domains of PACAP to its specific receptor and suggests heterogeneity within the binding mode of peptide ligands to class B GPCRs.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Amino Acid Sequence , Animals , Benzophenones/analysis , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Glycosylation , Humans , Iodine Isotopes/chemistry , Molecular Sequence Data , Phenylalanine/analogs & derivatives , Phenylalanine/analysis , Photoaffinity Labels/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/analysis , Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Protein Binding/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics
19.
Biochem Pharmacol ; 81(4): 552-61, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21114961

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide that exerts a large array of actions in the central nervous system and periphery. Through the activation of PAC1 and VPAC1, PACAP is able to exert neuroprotective, as well as anti-inflammatory effects, two phenomena involved in the pathogenesis and the progression of neurodegenerative diseases. The aim of the current study was to provide insights into the molecular arrangement of the amino terminus of PACAP and to develop new potent and selective PAC1/VPAC1 agonists promoting neuronal survival. We have synthesized a series of PACAP derivatives and measured their binding affinity and their ability to induce intracellular calcium mobilization for each receptor, i.e. PAC1, VPAC1, and VPAC2. Ultimately, analogs with an improved pharmacological profile were evaluated in an in vitro model of neuronal loss. Results showed that introduction of a hydroxyproline or an alanine moiety, respectively, at position 2 or 7 generated derivatives without significant VPAC2 agonistic activity. Moreover, the structure-activity relationship study suggests the presence of common (Asx-turn like) and distinct (different N-capping type) secondary structures that might be responsible for receptor recognition, selectivity and activation. Finally, evaluation of the neuroprotective activity of [Ala(7)]PACAP27 and [Hyp(2)]PACAP27 demonstrated their ability to protect potently human dopaminergic SH-SY5Y neuroblasts against the toxicity of MPP(+), in pre- and co-treatment experiments. These new pharmacological and structural data should prove useful for the rational design of PACAP-derived compounds that could be putative therapeutic agents for the treatment of neurodegenerative diseases.


Subject(s)
Neuroprotective Agents/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/agonists , Receptors, Vasoactive Intestinal Polypeptide, Type I/agonists , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Design , Humans , Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide , Structure-Activity Relationship , Transfection
20.
J Comb Chem ; 10(1): 44-51, 2008.
Article in English | MEDLINE | ID: mdl-18067269

ABSTRACT

The Suzuki-Miyaura (SM) cross-coupling reaction has recently become one of the most efficient methods for C-C bond construction opening a wide range of opportunities in organic synthesis. This study focused on the evaluation of the use of the SM reaction to modify peptides using a solid-phase synthesis approach, an avenue that was still not investigated intensively. We used as a peptide model [Ala (1,2,3), Leu (8)]Enk linked to a polystyrene support on which it was previously assembled. The aromatic residues Tyr (4) and Phe (7) of [Ala (1,2,3), Leu (8)]Enk were respectively substituted with p-iodo-Phe, and an SM-related strategy was developed. Results indicated that the reaction conditions involving K 3PO 4 or Na 2CO 3 (base), DMF (solvent), Pd(PPh 3) 4 (catalyst), and temperatures ranging from 50 to 80 degrees C during 20 h were found as optimal. Finally applying those optimal conditions, a series of [Ala (1,2,3), Leu (8)]Enk analogs modified at Tyr (4) or Phe (7) positions was synthesized using diverse boronic acid derivatives.


Subject(s)
Combinatorial Chemistry Techniques , Peptides , Catalysis , Chromatography, High Pressure Liquid , Combinatorial Chemistry Techniques/methods , Cross-Linking Reagents/chemistry , Palladium , Peptides/chemical synthesis , Peptides/chemistry , Polystyrenes/chemistry , Protein Conformation , Solvents/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...