Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 78
Filtrer
1.
Nucl Med Biol ; 134-135: 108913, 2024.
Article de Anglais | MEDLINE | ID: mdl-38703588

RÉSUMÉ

INTRODUCTION: Single domain antibody fragments (sdAbs) are an appealing scaffold for radiopharmaceutical development due to their small size (~15 kDa), high solubility, high stability, and excellent tumor penetration. Previously, we developed NB7 sdAb, which has very high affinity for an epitope on PSMA that is different from those targeted by small molecule PSMA inhibitors. Herein, we evaluated NB7 after radioiodination using [*I]SGMIB (1,3,4-isomer) and iso-[*I]SGMIB (1,3,5-isomer), as well as their 211At-labeled analogues. METHODS: [*I]SGMIB, iso-[*I]SGMIB, [211At]SAGMB, and iso-[211At]SAGMB conjugates of NB7 sdAb were synthesized and their binding affinity, cell uptake and internalization were assessed in PSMA+ PC3 PIP and PSMA- PC3 flu cells. Biodistribution studies were performed in mice bearing PSMA+ PC3 PIP xenografts. First, a single-label experiment evaluated the tissue distribution of a NB7 bearing a His6-tag (NB7H6) and labeled with iso-[125I]SGMIB. Three paired-label experiments then were performed to compare: a) NB7 labeled using [*I]SGMIB and iso-[*I]SGMIB, b) 131I- vs 211At-labeled NB7 conjugates and c) [125I]SGMIB-NB7H6 to the small molecule PSMA inhibitor [131I]YF2. RESULTS: All NB7 radioconjugates bound specifically to PSMA with dissociation constants, Kd, in the low nM range (1.4-6.4 nM). An initial biodistribution study demonstrated good tumor uptake for iso-[125I]SGMIB-NB7H6 (7.2 ± 1.5 % ID/g at 1 h) and no deleterious effect of the His6-tag on renal activity levels, which declined to 3.1 ± 1.1 % ID/g by 4 h. Paired-label biodistribution found no distinction between the two SGMIB isomer NB7 conjugates with the [131I]SGMIB-NB7-to-iso-[125I]SGMIB-NB7 tumor uptake ratios not significantly different from unity: 1.06 ± 0.08 at 1 h, 1.04 ± 0.12 at 4 h, and 1.07 ± 0.09 at 24 h. Both isomer conjugates cleared rapidly from normal tissues and exhibited very low uptake in thyroid, lacrimal and salivary glands. Paired-label biodistribution of [131I]SGMIB-NB7H6 and [211At]SAGMB-NB7H6 demonstrated similar tumor uptake and kidney clearance for the two radioconjugates. However, levels of 211At in thyroid, stomach, salivary and lacrimal glands were significantly higher (P < 0.05) that those for 131I suggesting greater dehalogenation for [211At]SAGMB-NB7H6. Finally, co-administration of [125I]SGMIB-NB7H6 and [131I]YF2 demonstrated good tumor uptake for both with considerably more rapid renal clearance for the NB7 radioconjugate. CONCLUSION: NB7 radioconjugates exhibited good accumulation in PSMA-positive xenografts with rapid clearance from kidney and other normal tissues. We conclude that NB7 is a potentially useful scaffold for developing PSMA-targeted theranostics with different characteristics than current small molecule and antibody-based approaches.


Sujet(s)
Antigènes de surface , Glutamate carboxypeptidase II , Tumeurs de la prostate , Anticorps à domaine unique , Mâle , Humains , Animaux , Souris , Tumeurs de la prostate/imagerie diagnostique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Glutamate carboxypeptidase II/immunologie , Glutamate carboxypeptidase II/antagonistes et inhibiteurs , Glutamate carboxypeptidase II/métabolisme , Anticorps à domaine unique/composition chimique , Anticorps à domaine unique/immunologie , Antigènes de surface/métabolisme , Antigènes de surface/immunologie , Lignée cellulaire tumorale , Distribution tissulaire , Transformation cellulaire néoplasique
2.
J Control Release ; 364: 246-260, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37879441

RÉSUMÉ

Sustained drug-release systems prolong the retention of therapeutic drugs within target tissues to alleviate the need for repeated drug administration. Two major caveats of the current systems are that the release rate and the timing cannot be predicted or fine-tuned because they rely on uncontrolled environmental conditions and that the system must be redesigned for each drug and treatment regime because the drug is bound via interactions that are specific to its structure and composition. We present a controlled and universal sustained drug-release system, which comprises minute spherical particles in which a therapeutic protein is affinity-bound to alginate sulfate (AlgS) through one or more short heparin-binding peptide (HBP) sequence repeats. Employing post-myocardial infarction (MI) heart remodeling as a case study, we show that the release of C9-a matrix metalloproteinase-9 (MMP-9) inhibitor protein that we easily bound to AlgS by adding one, two, or three HBP repeats to its sequence-can be directly controlled by modifying the number of HBP repeats. In an in vivo study, we directly injected AlgS particles, which were bound to C9 through three HBP repeats, into the left ventricular myocardium of mice following MI. We found that the particles substantially reduced post-MI remodeling, attesting to the sustained, local release of the drug within the tissue. As the number of HBP repeats controls the rate of drug release from the AlgS particles, and since C9 can be easily replaced with almost any protein, our tunable sustained-release system can readily accommodate a wide range of protein-based treatments.


Sujet(s)
Matrix metalloproteinase 9 , Infarctus du myocarde , Souris , Animaux , Matrix metalloproteinase 9/métabolisme , Préparations à action retardée/usage thérapeutique , Remodelage ventriculaire , Fonction ventriculaire gauche/physiologie , Infarctus du myocarde/thérapie , Myocarde/métabolisme
3.
Mol Oncol ; 17(11): 2337-2355, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37609678

RÉSUMÉ

Targeted therapies for prostate, breast, and ovarian cancers are based on their activity against primary tumors rather than their anti-metastatic activity. Consequently, there is an urgent need for new agents targeting the metastatic process. Emerging evidence correlates in vitro and in vivo cancer invasion and metastasis with increased activity of the proteases mesotrypsin (prostate and breast cancer) and kallikrein 6 (KLK6; ovarian cancer). Thus, mesotrypsin and KLK6 are attractive putative targets for therapeutic intervention. As potential therapeutics for advanced metastatic prostate, breast, and ovarian cancers, we report novel mesotrypsin- and KLK6-based therapies, based on our previously developed mutants of the human amyloid ß-protein precursor Kunitz protease inhibitor domain (APPI). These mutants, designated APPI-3M (prostate and breast cancer) and APPI-4M (ovarian cancer), demonstrated significant accumulation in tumors and therapeutic efficacy in orthotopic preclinical models, with the advantages of long retention times in vivo, high affinity and favorable pharmacokinetic properties. The applicability of the APPIs, as a novel therapy and for imaging purposes, is supported by their good safety profile and their controlled and scalable manufacturability in bioreactors.


Sujet(s)
Tumeurs du sein , Tumeurs de l'ovaire , Mâle , Humains , Femelle , Inhibiteurs de la sérine protéinase/usage thérapeutique , Peptides bêta-amyloïdes/usage thérapeutique , Prostate/anatomopathologie , Précurseur de la protéine bêta-amyloïde/pharmacologie , Précurseur de la protéine bêta-amyloïde/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Kallicréines/génétique
4.
ACS Chem Neurosci ; 14(18): 3385-3397, 2023 09 20.
Article de Anglais | MEDLINE | ID: mdl-37579500

RÉSUMÉ

An amyloid precursor protein inhibitor (APPI) and amyloid beta 42 (Aß42) are both subdomains of the human transmembrane amyloid precursor protein (APP). In the brains of patients with Alzheimer's disease (AD), Aß42 oligomerizes into aggregates of various sizes, with intermediate, low-molecular-weight Aß42 oligomers currently being held to be the species responsible for the most neurotoxic effects associated with the disease. Strategies to ameliorate the toxicity of these intermediate Aß42 oligomeric species include the use of short, Aß42-interacting peptides that either inhibit the formation of the Aß42 oligomeric species or promote their conversion to high-molecular-weight aggregates. We therefore designed such an Aß42-interacting peptide that is based on the ß-hairpin amino acid sequence of the APPI, which exhibits high similarity to the ß-sheet-like aggregation site of Aß42. Upon tight binding of this 20-mer cyclic peptide to Aß42 (in a 1:1 molar ratio), the formation of Aß42 aggregates was enhanced, and consequently, Aß42-mediated cell toxicity was ameliorated. We showed that in the presence of the cyclic peptide, interactions of Aß42 with both plasma and mitochondrial membranes and with phospholipid vesicles that mimic these membranes were inhibited. Specifically, the cyclic peptide inhibited Aß42-mediated mitochondrial membrane depolarization and reduced Aß42-mediated apoptosis and cell death. We suggest that the cyclic peptide modulates Aß42 aggregation by enhancing the formation of large aggregates─as opposed to low-molecular-weight intermediates─and as such has the potential for further development as an AD therapeutic.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Humains , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde , Peptides cycliques/pharmacologie , Fragments peptidiques/métabolisme , Maladie d'Alzheimer/métabolisme
5.
Sci Rep ; 13(1): 11923, 2023 07 24.
Article de Anglais | MEDLINE | ID: mdl-37488176

RÉSUMÉ

Cancer progression is enhanced by the interaction of programmed death-ligand 1 (PDL1), which is associated with inhibition of the immune response against tumors, and vascular endothelial growth factor (VEGF), which inhibits immune cell activity while inducing angiogenesis and proliferation of cancer cells. Dual inhibition of PDL1 and VEGF may therefore confer a synergistic anti-cancer therapeutic effect. We present a novel strategy for developing a therapeutic that simultaneously binds and inhibits both PDL1 and VEGF. We generated a bi-specific protein, designated DuRan-Bis, comprising a single chain variable fragment (scFv)-based inhibitor of PDL1 fused to an scFv-based inhibitor of VEGF, with the latter being attached to an Fc fragment. We found that DuRan-Bis binds to both PDL1 and VEGF with high affinity. Compared to treatments with mono-specific proteins, alone or in combination, the DuRan-Bis chimera showed superior inhibition of the proliferation of glioblastoma cells. In comparison to treatment with immune cells alone, a combination of immune cells with DuRan-Bis decreased the viability of head and neck cancer cells. To the best of our knowledge, this study is the first to use a single polypeptide chain scFv-scFv-Fc scaffold for engineering a high-affinity bi-specific inhibitor of PDL1 and VEGF.


Sujet(s)
Glioblastome , Anticorps à chaîne unique , Humains , Facteur de croissance endothéliale vasculaire de type A , Antigène CD274 , Inhibiteurs de l'angiogenèse
6.
Sci Rep ; 13(1): 5186, 2023 03 30.
Article de Anglais | MEDLINE | ID: mdl-36997589

RÉSUMÉ

Matrix metalloproteinases (MMPs) regulate the degradation of extracellular matrix (ECM) components in biological processes. MMP activity is controlled by natural tissue inhibitors of metalloproteinases (TIMPs) that non-selectively inhibit the function of multiple MMPs via interaction with the MMPs' Zn2+-containing catalytic pocket. Recent studies suggest that TIMPs engineered to confer MMP specificity could be exploited for therapeutic purposes, but obtaining specific TIMP-2 inhibitors has proved to be challenging. Here, in an effort to improve MMP specificity, we incorporated the metal-binding non-canonical amino acids (NCAAs), 3,4-dihydroxyphenylalanine (L-DOPA) and (8-hydroxyquinolin-3-yl)alanine (HqAla), into the MMP-inhibitory N-terminal domain of TIMP2 (N-TIMP2) at selected positions that interact with the catalytic Zn2+ ion (S2, S69, A70, L100) or with a structural Ca2+ ion (Y36). Evaluation of the inhibitory potency of the NCAA-containing variants towards MMP-2, MMP-9 and MMP-14 in vitro revealed that most showed a significant loss of inhibitory activity towards MMP-14, but not towards MMP-2 and MMP-9, resulting in increased specificity towards the latter proteases. Substitutions at S69 conferred the best improvement in selectivity for both L-DOPA and HqAla variants. Molecular modeling provided an indication of how MMP-2 and MMP-9 are better able to accommodate the bulky NCAA substituents at the intermolecular interface with N-TIMP2. The models also showed that, rather than coordinating to Zn2+, the NCAA side chains formed stabilizing polar interactions at the intermolecular interface with MMP-2 and MMP-9. Our findings illustrate how incorporation of NCAAs can be used to probe-and possibly exploit-differential tolerance for substitution within closely related protein-protein complexes as a means to improve specificity.


Sujet(s)
Matrix metalloproteinase 2 , Inhibiteur tissulaire de métalloprotéinase-2 , Inhibiteur tissulaire de métalloprotéinase-2/génétique , Inhibiteur tissulaire de métalloprotéinase-2/métabolisme , Matrix metalloproteinase 2/génétique , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 9/génétique , Matrix metalloproteinase 14 , Lévodopa , Inhibiteur tissulaire des métalloprotéinases/génétique
7.
Res Sq ; 2023 Jan 16.
Article de Anglais | MEDLINE | ID: mdl-36712032

RÉSUMÉ

Matrix metalloproteinases (MMPs) regulate the degradation of extracellular matrix (ECM) components in biological processes. MMP activity is controlled by natural tissue inhibitors of metalloproteinases (TIMPs) that non-selectively inhibit the function of multiple MMPs via interaction with the MMPs' Zn 2+ -containing catalytic pocket. Recent studies suggest that TIMPs engineered to confer MMP specificity could be exploited for therapeutic purposes, but obtaining specific TIMP-2 inhibitors has proved to be challenging. Here, in an effort to improve MMP specificity, we incorporated the metal-binding non-canonical amino acids (NCAAs), 3,4-dihydroxyphenylalanine (L-DOPA) and (8-hydroxyquinolin-3-yl)alanine (HqAla), into the MMP-inhibitory N-terminal domain of TIMP2 (N-TIMP2) at selected positions that interact with the catalytic Zn 2+ ion (S2, S69, A70, L100) or with a structural Ca 2+ ion (Y36). Evaluation of the inhibitory potency of the NCAA-containing variants towards MMP-2, MMP-9 and MMP-14 in vitro revealed that most showed a significant loss of inhibitory activity towards MMP-14, but not towards MMP-2 and MMP-9, resulting in increased specificity towards the latter proteases. Substitutions at S69 conferred the best improvement in selectivity for both L-DOPA and HqAla variants. Molecular modeling revealed how MMP-2 and MMP-9 are better able to accommodate the bulky NCAA substituents at the intermolecular interface with N-TIMP2. The models also showed that, rather than coordinating to Zn 2+ , the NCAA side chains formed stabilizing polar interactions at the intermolecular interface with MMP-2 and MMP-9. The findings illustrate how incorporation of NCAAs can be used to probe and exploit differential tolerance for substitution within closely related protein-protein complexes to achieve improved specificity.

8.
Sci Rep ; 12(1): 8657, 2022 05 23.
Article de Anglais | MEDLINE | ID: mdl-35606532

RÉSUMÉ

The immense potential of G protein-coupled receptors (GPCRs) as targets for drug discovery is not fully realized due to the enormous difficulties associated with structure elucidation of these profoundly unstable membrane proteins. The existing methods of GPCR stability-engineering are cumbersome and low-throughput; in addition, the scope of GPCRs that could benefit from these techniques is limited. Here, we present a yeast-based screening platform for a single-step isolation of GRCR variants stable in the presence of short-chain detergents, a feature essential for their successful crystallization using vapor diffusion method. The yeast detergent-resistant cell wall presents a unique opportunity for compartmentalization, to physically link the receptor's phenotype to its encoding DNA, and thus enable discovery of stable GPCR variants with unprecedent efficiency. The scope of mutations identified by the method reveals a surprising amenability of the GPCR scaffold to stabilization, and suggests an intriguing possibility of amending the stability properties of GPCR by varying the structural status of the C-terminus.


Sujet(s)
Récepteurs couplés aux protéines G , Saccharomyces cerevisiae , Découverte de médicament , Protéines membranaires/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme
9.
Cell Mol Life Sci ; 79(6): 312, 2022 May 23.
Article de Anglais | MEDLINE | ID: mdl-35604495

RÉSUMÉ

The human signaling molecules Tie1 and Tie2 receptor tyrosine kinases (RTKs) play important pathophysiological roles in many diseases, including different cancers. The activity of Tie1 is mediated mainly through the downstream angiopoietin-1 (Ang1)-dependent activation of Tie2, rendering both Tie 1 and the Tie1/Tie2/Ang1 axis attractive putative targets for therapeutic intervention. However, the development of inhibitors that target Tie1 and an understanding of their effect on Tie2 and on the Tie1/Tie2/Ang1 axis remain unfulfilled tasks, due, largely, to the facts that Tie1 is an orphan receptor and is difficult to produce and use in the quantities required for immune antibody library screens. In a search for a selective inhibitor of this orphan receptor, we sought to exploit the advantages (e.g., small size that allows binding to hidden epitopes) of non-immune nanobodies and to simultaneously overcome their limitations (i.e., low expression and stability). We thus performed expression, stability, and affinity screens of yeast-surface-displayed naïve and predesigned synthetic (non-immune) nanobody libraries against the Tie1 extracellular domain. The screens yielded a nanobody with high expression and good affinity and specificity for Tie1, thereby yielding preferential binding for Tie1 over Tie2. The stability, selectivity, potency, and therapeutic potential of this synthetic nanobody were profiled using in vitro and cell-based assays. The nanobody triggered Tie1-dependent inhibition of RTK (Tie2, Akt, and Fak) phosphorylation and angiogenesis in endothelial cells, as well as suppression of human glioblastoma cell viability and migration. This study opens the way to developing nanobodies as therapeutics for different cancers associated with Tie1 activation.


Sujet(s)
Tumeurs , Anticorps à domaine unique , Angiopoïétine-1 , Cellules endothéliales/métabolisme , Humains , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Phosphorylation , Récepteur TIE-1/métabolisme , Récepteur TIE-2/génétique , Récepteur TIE-2/métabolisme , Anticorps à domaine unique/pharmacologie
10.
Bioconjug Chem ; 33(5): 795-806, 2022 05 18.
Article de Anglais | MEDLINE | ID: mdl-35446024

RÉSUMÉ

Tissue inhibitors of metalloproteinases (TIMPs) are natural inhibitors of the matrix metalloproteinase (MMP) family of proteins, whose members are key regulators of the proteolysis of extracellular matrix components and hence of multiple biological processes. In particular, imbalanced activity of matrix metalloproteinase-14 (MMP-14) may lead to the development of cancer and cardiovascular and other diseases. This study aimed to engineer TIMP2, one of the four homologous TIMPs, as a potential therapeutic by virtue of its ability to bind to the active-site Zn2+ of MMP-14. However, the susceptibility to degradation of TIMP2 and its small size, which results in a short circulation half-life, limit its use as a therapeutic. PEGylation was thus used to improve the pharmacokinetic profile of TIMP2. PEGylation of the MMP-targeting N-terminal domain of TIMP2 (N-TIMP2), via either cysteine or lysine residues, resulted in a significant decrease in N-TIMP2 affinity toward MMP-14 or multisite conjugation and conjugate heterogeneity, respectively. Our strategy designed to address this problem was based on incorporating a noncanonical amino acid (NCAA) into N-TIMP2 to enable site-specific mono-PEGylation. The first step was to incorporate the NCAA propargyl lysine (PrK) at position S31 in N-TIMP2, which does not interfere with the N-TIMP2-MMP-14 binding interface. Thereafter, site-specific PEGylation was achieved via a click chemistry reaction between N-TIMP2-S31PrK and PEG-azide-20K. Inhibition studies showed that PEGylated N-TIMP2-S31PrK did indeed retain its inhibitory activity toward MMP-14. The modified protein also showed improved serum stability vs non-PEGylated N-TIMP2. In vivo pharmacokinetic studies in mice revealed a significant 8-fold increase in the elimination half-life of PEGylated N-TIMP2 vs the non-PEGylated protein. This study shows that site-specific bioorthogonal mono-PEGylation extends the half-life of N-TIMP2 without impairing its biological activity, thereby highlighting the advantage of this strategy for generating potent PEGylated proteins.


Sujet(s)
Lysine , Matrix metalloproteinase 14 , Inhibiteur tissulaire de métalloprotéinase-2 , Animaux , Période , Lysine/métabolisme , Matrix metalloproteinase 14/métabolisme , Matrix metalloproteinases , Souris , Polyéthylène glycols/composition chimique , Inhibiteur tissulaire de métalloprotéinase-2/métabolisme
11.
Protein Eng Des Sel ; 352022 02 17.
Article de Anglais | MEDLINE | ID: mdl-35174858

RÉSUMÉ

LDL-receptor (LDLR)-mediated uptake of LDL-C into hepatocytes is impaired by lysosomal degradation of LDLR, which is promoted by proprotein convertase subtilisin/kexin type 9 (PCSK9). Cell surface binding of PCSK9 to LDLR produces a complex that translocates to an endosome, where the acidic pH strengthens the binding affinity of PCSK9 to LDLR, preventing LDLR recycling to the cell membrane. We present a new approach to inhibit PCSK9-mediated LDLR degradation, namely, targeting the PCSK9/LDLR interface with a PCSK9-antagonist, designated Flag-PCSK9PH, which prevents access of WT PCSK9 to LDLR. In HepG2 cells, Flag-PCSK9PH, a truncated version (residues 53-451) of human WT PCSK9, strongly bound LDLR at the neutral pH of the cell surface but dissociated from it in the endosome (acidic pH), allowing LDLR to exit the lysosomes intact and recycle to the cell membrane. Flag-PCSK9PH thus significantly enhanced cell-surface LDLR levels and the ability of LDLR to take up extracellular LDL-C.


Sujet(s)
Hépatocytes , Proprotéine convertase 9 , Récepteurs aux lipoprotéines LDL/métabolisme , Cholestérol LDL , Cellules HepG2 , Humains
12.
Cell Mol Life Sci ; 79(2): 113, 2022 Jan 31.
Article de Anglais | MEDLINE | ID: mdl-35099616

RÉSUMÉ

Induction of bone formation by Wnt ligands is inhibited when sclerostin (Scl), an osteocyte-produced antagonist, binds to its receptors, the low-density lipoprotein receptor-related proteins 5 or 6 (LRP5/6). Recently, it was shown that enhanced inhibition is achieved by Scl binding to the co-receptor LRP4. However, it is not clear if the binding of Scl to LRP4 facilitates Scl binding to LRP5/6 or inhibits the Wnt pathway in an LRP5/6-independent manner. Here, using the yeast display system, we demonstrate that Scl exhibits a stronger binding affinity for LRP4 than for LRP6. Moreover, we found stronger Scl binding to LRP6 in the presence of LRP4. We further show that a Scl mutant (SclN93A), which tightly binds LRP4 but not LRP6, does not inhibit the Wnt pathway on its own. We demonstrate that SclN93A competes with Scl for a common binding site on LRP4 and antagonizes Scl inhibition of the Wnt signaling pathway in osteoblasts in vitro. Finally, we demonstrate that 2 weeks of bi-weekly subcutaneous injections of SclN93A fused to the fragment crystallizable (Fc) domain of immunoglobulin (SclN93AFc), which retains the antagonistic activity of the mutant, significantly increases bone formation rate and enhances trabecular volumetric bone fraction, trabecular number, and bone length in developing mice. Our data show that LRP4 serves as an anchor that facilitates Scl-LRP6 binding and that inhibition of the Wnt pathway by Scl depends on its prior binding to LRP4. We further provide evidence that compounds that inhibit Scl-LRP4 interactions offer a potential strategy to promote anabolic bone functions.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines apparentées au récepteur LDL/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Protéines recombinantes/pharmacologie , Protéines adaptatrices de la transduction du signal/composition chimique , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Fixation compétitive/effets des médicaments et des substances chimiques , Fixation compétitive/génétique , Cellules cultivées , Femelle , Cellules HEK293 , Humains , Protéines apparentées au récepteur LDL/antagonistes et inhibiteurs , Protéines apparentées au récepteur LDL/composition chimique , Protéines apparentées au récepteur LDL/génétique , Souris , Souris de lignée C57BL , Protéines mutantes/composition chimique , Protéines mutantes/pharmacologie , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/physiologie , Ostéogenèse/génétique , Liaison aux protéines/effets des médicaments et des substances chimiques , Liaison aux protéines/génétique , Motifs et domaines d'intéraction protéique/effets des médicaments et des substances chimiques , Motifs et domaines d'intéraction protéique/génétique , Petit ARN interférent/pharmacologie , Protéines recombinantes/composition chimique
13.
Proteins ; 90(1): 45-57, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34293212

RÉSUMÉ

Deep mutational scanning provides unprecedented wealth of quantitative data regarding the functional outcome of mutations in proteins. A single experiment may measure properties (eg, structural stability) of numerous protein variants. Leveraging the experimental data to gain insights about unexplored regions of the mutational landscape is a major computational challenge. Such insights may facilitate further experimental work and accelerate the development of novel protein variants with beneficial therapeutic or industrially relevant properties. Here we present a novel, machine learning approach for the prediction of functional mutation outcome in the context of deep mutational screens. Using sequence (one-hot) features of variants with known properties, as well as structural features derived from models thereof, we train predictive statistical models to estimate the unknown properties of other variants. The utility of the new computational scheme is demonstrated using five sets of mutational scanning data, denoted "targets": (a) protease specificity of APPI (amyloid precursor protein inhibitor) variants; (b-d) three stability related properties of IGBPG (immunoglobulin G-binding ß1 domain of streptococcal protein G) variants; and (e) fluorescence of GFP (green fluorescent protein) variants. Performance is measured by the overall correlation of the predicted and observed properties, and enrichment-the ability to predict the most potent variants and presumably guide further experiments. Despite the diversity of the targets the statistical models can generalize variant examples thereof and predict the properties of test variants with both single and multiple mutations.


Sujet(s)
Analyse de mutations d'ADN/méthodes , Séquençage nucléotidique à haut débit/méthodes , Apprentissage machine , Mutation/génétique , Protéines , Algorithmes , Biologie informatique/méthodes , Modèles statistiques , Cartes d'interactions protéiques , Protéines/composition chimique , Protéines/génétique , Protéines/métabolisme
14.
PLoS One ; 16(11): e0249616, 2021.
Article de Anglais | MEDLINE | ID: mdl-34847142

RÉSUMÉ

Although myriad protein-protein interactions in nature use polyvalent binding, in which multiple ligands on one entity bind to multiple receptors on another, to date an affinity advantage of polyvalent binding has been demonstrated experimentally only in cases where the target receptor molecules are clustered prior to complex formation. Here, we demonstrate cooperativity in binding affinity (i.e., avidity) for a protein complex in which an engineered dimer of the amyloid precursor protein inhibitor (APPI), possessing two fully functional inhibitory loops, interacts with mesotrypsin, a soluble monomeric protein that does not self-associate or cluster spontaneously. We found that each inhibitory loop of the purified APPI homodimer was over three-fold more potent than the corresponding loop in the monovalent APPI inhibitor. This observation is consistent with a suggested mechanism whereby the two APPI loops in the homodimer simultaneously and reversibly bind two corresponding mesotrypsin monomers to mediate mesotrypsin dimerization. We propose a simple model for such dimerization that quantitatively explains the observed cooperativity in binding affinity. Binding cooperativity in this system reveals that the valency of ligands may affect avidity in protein-protein interactions including those of targets that are not surface-anchored and do not self-associate spontaneously. In this scenario, avidity may be explained by the enhanced concentration of ligand binding sites in proximity to the monomeric target, which may favor rebinding of the multiple ligand binding sites with the receptor molecules upon dissociation of the protein complex.


Sujet(s)
Modèles moléculaires , Liaison aux protéines , Sites de fixation , Domaine catalytique , Multimérisation de protéines , Trypsine/métabolisme
15.
ACS Chem Neurosci ; 12(24): 4554-4563, 2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34806861

RÉSUMÉ

Aggregation of the ß-Amyloid (Aß) peptide in brain tissues is the hallmark of Alzheimer's disease (AD). While Aß is presumed to be insidiously involved in the disease's pathophysiology, concrete mechanisms accounting for the role of Aß in AD are yet to be deciphered. While Aß has been primarily identified in the extracellular space, the peptide also accumulates in cellular compartments such as mitochondria and lysosomes and impairs cellular functions. Here, we show that prominent proapoptotic peptides associated with the mitochondrial outer membrane, the Bcl-2-homology-only peptides BID, PUMA, and NOXA, exert significant and divergent effects upon aggregation, cytotoxicity, and membrane interactions of Aß42, the main Aß homolog. Interestingly, we show that BID and PUMA accelerated aggregation of Aß42, reduced Aß42-induced toxicity and mitochondrial disfunction, and inhibited Aß42-membrane interactions. In contrast, NOXA exhibited opposite effects, reducing Aß42 fibril formation, affecting more pronounced apoptotic effects and mitochondrial disfunction, and enhancing membrane interactions of Aß42. The effects of BID, PUMA, and NOXA upon the Aß42 structure and toxicity may be linked to its biological properties and affect pathophysiological features of AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Peptides bêta-amyloïdes/toxicité , Humains , Mitochondries , Fragments peptidiques
16.
Cell Mol Life Sci ; 78(23): 7185-7198, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34643743

RÉSUMÉ

A review of the multidisciplinary scientific literature reveals a large variety of amyloid-ß (Aß) oligomeric species, differing in molecular weight, conformation and morphology. These species, which may assemble via either on- or off-aggregation pathways, exhibit differences in stability, function and neurotoxicity, according to different experimental settings. The conformations of the different Aß species are stabilized by intra- and inter-molecular hydrogen bonds and by electrostatic and hydrophobic interactions, all depending on the chemical and physical environment (e.g., solvent, ions, pH) and interactions with other molecules, such as lipids and proteins. This complexity and the lack of a complete understanding of the relationship between the different Aß species and their toxicity is currently dictating the nature of the inhibitor (or inducer)-based approaches that are under development for interfering with (or inducing) the formation of specific species and Aß oligomerization, and for interfering with the associated downstream neurotoxic effects. Here, we review the principles that underlie the involvement of different Aß oligomeric species in neurodegeneration, both in vitro and in preclinical studies. In addition, we provide an overview of the existing inhibitors (or inducers) of Aß oligomerization that serve as potential therapeutics for neurodegenerative diseases. The review, which covers the exciting studies that have been published in the past few years, comprises three main parts: 1) on- and off-fibrillar assembly mechanisms and Aß structural polymorphism; 2) interactions of Aß with other molecules and cell components that dictate the Aß aggregation pathway; and 3) targeting the on-fibrillar Aß assembly pathway as a therapeutic approach.


Sujet(s)
Peptides bêta-amyloïdes/génétique , Peptides bêta-amyloïdes/métabolisme , Amyloïde/composition chimique , Maladies neurodégénératives/anatomopathologie , Fragments peptidiques/métabolisme , Humains , Maladies neurodégénératives/génétique , Maladies neurodégénératives/thérapie , Fragments peptidiques/génétique , Agrégation pathologique de protéines/anatomopathologie , Conformation des protéines
17.
J Am Chem Soc ; 143(41): 17261-17275, 2021 10 20.
Article de Anglais | MEDLINE | ID: mdl-34609866

RÉSUMÉ

Protein-protein interactions (PPIs) have evolved to display binding affinities that can support their function. As such, cognate and noncognate PPIs could be highly similar structurally but exhibit huge differences in binding affinities. To understand this phenomenon, we study three homologous protease-inhibitor PPIs that span 9 orders of magnitude in binding affinity. Using state-of-the-art methodology that combines protein randomization, affinity sorting, deep sequencing, and data normalization, we report quantitative binding landscapes consisting of ΔΔGbind values for the three PPIs, gleaned from tens of thousands of single and double mutations. We show that binding landscapes of the three complexes are strikingly different and depend on the PPI evolutionary optimality. We observe different patterns of couplings between mutations for the three PPIs with negative and positive epistasis appearing most frequently at hot-spot and cold-spot positions, respectively. The evolutionary trends observed here are likely to be universal to other biological complexes in the cell.


Sujet(s)
Cartographie d'interactions entre protéines
18.
Sci Rep ; 11(1): 16170, 2021 08 09.
Article de Anglais | MEDLINE | ID: mdl-34373558

RÉSUMÉ

Proteinase-activated receptor-1 (PAR1), triggered by thrombin and other serine proteinases such as tissue kallikrein-4 (KLK4), is a key driver of inflammation, tumor invasiveness and tumor metastasis. The PAR1 transmembrane G-protein-coupled receptor therefore represents an attractive target for therapeutic inhibitors. We thus used a computational design to develop a new PAR1 antagonist, namely, a catalytically inactive human KLK4 that acts as a proteinase substrate-capture reagent, preventing receptor cleavage (and hence activation) by binding to and occluding the extracellular R41-S42 canonical PAR1 proteolytic activation site. On the basis of in silico site-saturation mutagenesis, we then generated KLK4S207A,L185D, a first-of-a-kind 'decoy' PAR1 inhibitor, by mutating the S207A and L185D residues in wild-type KLK4, which strongly binds to PAR1. KLK4S207A,L185D markedly inhibited PAR1 cleavage, and PAR1-mediated MAPK/ERK activation as well as the migration and invasiveness of melanoma cells. This 'substrate-capturing' KLK4 variant, engineered to bind to PAR1, illustrates proof of principle for the utility of a KLK4 'proteinase substrate capture' approach to regulate proteinase-mediated PAR1 signaling.


Sujet(s)
Kallicréines/métabolisme , Récepteur de type PAR-1/antagonistes et inhibiteurs , Substitution d'acide aminé , Sites de fixation , Lignée cellulaire tumorale , Simulation numérique , Conception de médicament , Humains , Kallicréines/composition chimique , Kallicréines/génétique , Cinétique , Cellules MCF-7 , Mutagenèse dirigée , Invasion tumorale/prévention et contrôle , Ingénierie des protéines , Motifs et domaines d'intéraction protéique , Protéolyse , Récepteur de type PAR-1/composition chimique , Récepteur de type PAR-1/métabolisme , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Transduction du signal , Spécificité du substrat , Thrombine/métabolisme
19.
Cancers (Basel) ; 13(16)2021 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-34439122

RÉSUMÉ

As cancer-associated factors, kallikrein-related peptidases (KLKs) are components of the tumour microenvironment, which represents a rich substrate repertoire, and considered attractive targets for the development of novel treatments. Standard-of-care therapy of pancreatic cancer shows unsatisfactory results, indicating the need for alternative therapeutic approaches. We aimed to investigate the expression of KLKs in pancreatic cancer and to inhibit the function of KLK6 in pancreatic cancer cells. KLK6, KLK7, KLK8, KLK10 and KLK11 were coexpressed and upregulated in tissues from pancreatic cancer patients compared to normal pancreas. Their high expression levels correlated with each other and were linked to shorter survival compared to low KLK levels. We then validated KLK6 mRNA and protein expression in patient-derived tissues and pancreatic cancer cells. Coexpression of KLK6 with KRT19, αSMA or CD68 was independent of tumour stage, while KLK6 was coexpressed with KRT19 and CD68 in the invasive tumour area. High KLK6 levels in tumour and CD68+ cells were linked to shorter survival. KLK6 inhibition reduced KLK6 mRNA expression, cell metabolic activity and KLK6 secretion and increased the secretion of other serine and aspartic lysosomal proteases. The association of high KLK levels and poor prognosis suggests that inhibiting KLKs may be a therapeutic strategy for precision medicine.

20.
ACS Chem Neurosci ; 12(9): 1563-1577, 2021 05 05.
Article de Anglais | MEDLINE | ID: mdl-33904703

RÉSUMÉ

It is currently believed that molecular agents that specifically bind to and neutralize the toxic proteins/peptides, amyloid ß (Aß42), tau, and the tau-derived peptide PHF6, hold the key to attenuating the progression of Alzheimer's disease (AD). We thus tested our previously developed nonaggregating Aß42 double mutant (Aß42DM) as a multispecific binder for three AD-associated molecules, wild-type Aß42, the tauK174Q mutant, and a synthetic PHF6 peptide. Aß42DM acted as a functional inhibitor of these molecules in in vitro assays and in neuronal cell-based models of AD. The double mutant bound both cytotoxic tauK174Q and synthetic PHF6 and protected neuronal cells from the accumulation of tau in cell lysates and mitochondria. Aß42DM also reduced toxic intracellular levels of calcium and the overall cell toxicity induced by overexpressed tau, synthetic PHF6, Aß42, or a combination of PHF6and Aß42. Aß42DM inhibited PHF6-induced overall mitochondrial dysfunction: In particular, Aß42DM inhibited PHF6-induced damage to submitochondrial particles (SMPs) and suppressed PHF6-induced elevation of the ζ-potential of inverted SMPs (proxy for the inner mitochondrial membrane, IMM). PHF6 reduced the lipid fluidity of cardiolipin/DOPC vesicles (that mimic the IMM) but not DOPC (which mimics the outer mitochondrial membrane), and this effect was inhibited by Aß42DM. This inhibition may be explained by the conformational changes in PHF6 induced by Aß42DM in solution and in membrane mimetics. On this basis, the paper presents a mechanistic explanation for the inhibitory activity of Aß42DM against Aß42- and tau-induced membrane permeability and cell toxicity and provides confirmatory evidence for its protective function in neuronal cells.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Peptides bêta-amyloïdes/toxicité , Humains , Membrane artificielle , Mitochondries , Fragments peptidiques/toxicité , Protéines tau
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...