Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3259, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627419

ABSTRACT

The heterogeneity inherent in today's biotherapeutics, especially as a result of heavy glycosylation, can affect a molecule's safety and efficacy. Characterizing this heterogeneity is crucial for drug development and quality assessment, but existing methods are limited in their ability to analyze intact glycoproteins or other heterogeneous biotherapeutics. Here, we present an approach to the molecular assessment of biotherapeutics that uses proton-transfer charge-reduction with gas-phase fractionation to analyze intact heterogeneous and/or glycosylated proteins by mass spectrometry. The method provides a detailed landscape of the intact molecular weights present in biotherapeutic protein preparations in a single experiment. For glycoproteins in particular, the method may offer insights into glycan composition when coupled with a suitable bioinformatic strategy. We tested the approach on various biotherapeutic molecules, including Fc-fusion, VHH-fusion, and peptide-bound MHC class II complexes to demonstrate efficacy in measuring the proteoform-level diversity of biotherapeutics. Notably, we inferred the glycoform distribution for hundreds of molecular weights for the eight-times glycosylated fusion drug IL22-Fc, enabling correlations between glycoform sub-populations and the drug's pharmacological properties. Our method is broadly applicable and provides a powerful tool to assess the molecular heterogeneity of emerging biotherapeutics.


Subject(s)
Glycoproteins , Polysaccharides , Glycosylation , Glycoproteins/metabolism , Mass Spectrometry/methods , Polysaccharides/metabolism
2.
Anal Chem ; 95(49): 17957-17961, 2023 12 12.
Article in English | MEDLINE | ID: mdl-38084380

ABSTRACT

Biotransformation leading to single residue modifications (e.g., deamidation, oxidation) can contribute to decreased efficacy/potency, poor pharmacokinetics, and/or toxicity/immunogenicity for protein therapeutics. Identifying and characterizing such liabilities in vivo are emerging needs for biologics drug discovery. In vitro stress assays involving PBS for deamidation or AAPH for oxidation are commonly used for predicting liabilities in manufacturing and storage and are sometimes considered a predictive tool for in vivo liabilities. However, reports discussing their in vivo translatability are limited. Herein, we introduce a mass spectrometry workflow that characterizes in vivo oxidation and deamidation in pharmacokinetically relevant compartments for diverse protein therapeutic modalities. The workflow has low bias of <10% in quantitating degradation in the relevant pharmacokinetic concentration range for monkey and rabbit serum/plasma (1-100 µg/mL) and allows for high sequence coverage (∼85%) for discovery/monitoring of amino acid modifications. For oxidation and deamidation, the assay was precise, with percent coefficient of variation of <8% at 1-100 µg/mL and ≤6% method-induced artifacts. A high degree of in vitro and in vivo correlation was observed for deamidation on the six diverse protein therapeutics (seven liability sites) tested. In vivo translatability for oxidation liabilities were not observed for the 11 molecules tested using in vitro AAPH stress. One of the molecules dosed in eyes resulted in a false positive and a false negative prediction for in vivo oxidation following AAPH stress. Finally, peroxide stress was also tested but resulted in limited success (1 out of 4 molecules) in predicting oxidation liabilities.


Subject(s)
Oxidation-Reduction , Animals , Rabbits , Biotransformation
3.
Cell Chem Biol ; 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38056465

ABSTRACT

Selective and precise activation of signaling transduction cascades is key for cellular reprogramming and tissue regeneration. However, the development of small- or large-molecule agonists for many signaling pathways has remained elusive and is rate limiting to realize the full clinical potential of regenerative medicine. Focusing on the Wnt pathway, here we describe a series of disulfide-constrained peptides (DCPs) that promote Wnt signaling activity by modulating the cell surface levels of ZNRF3, an E3 ubiquitin ligase that controls the abundance of the Wnt receptor complex FZD/LRP at the plasma membrane. Mechanistically, monomeric DCPs induce ZNRF3 ubiquitination, leading to its cell surface clearance, ultimately resulting in FZD stabilization. Furthermore, we engineered multimeric DCPs that induce expansive growth of human intestinal organoids, revealing a dependence between valency and ZNRF3 clearance. Our work highlights a strategy for the development of potent, biologically active Wnt signaling pathway agonists via targeting of ZNRF3.

4.
Transl Vis Sci Technol ; 11(10): 27, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36255358

ABSTRACT

Purpose: Diabetic macular edema (DME) is the leading cause of vision loss and blindness among working-age adults. Although current intravitreal anti-vascular endothelial growth factor (VEGF) therapies improve vision for many patients with DME, approximately half do not achieve the visual acuity required to drive. We therefore sought additional approaches to resolve edema and improve vision for these patients. Methods: We explored direct agonists of Tie2, a receptor known to stabilize vasculature and prevent leakage. We identified a multivalent PEG-Fab conjugate, Tie2.1-hexamer, that oligomerizes Tie2 and drives receptor activation and characterized its activities in vitro and in vivo. Results: Tie2.1-hexamer normalized and stabilized intercellular junctions of stressed endothelial cell monolayers in vitro, suppressed vascular leak in mice under conditions where anti-VEGF alone was ineffective, and demonstrated extended ocular exposure and robust pharmacodynamic responses in non-human primates. Conclusions: Tie2.1-hexamer directly activates the Tie2 pathway, reduces vascular leak, and is persistent within the vitreal humor. Translational Relevance: Our study presents a promising potential therapeutic for the treatment of DME.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , Mice , Animals , Macular Edema/drug therapy , Macular Edema/etiology , Diabetic Retinopathy/drug therapy , Endothelial Growth Factors/therapeutic use , Visual Acuity , Vision Disorders/complications , Vision Disorders/drug therapy , Blindness/complications
5.
MAbs ; 14(1): 2115213, 2022.
Article in English | MEDLINE | ID: mdl-36206404

ABSTRACT

T cell-engaging bispecific antibodies (TCEs) are clinically effective treatments for hematological cancers. While the utility of TCEs in solid malignancies is being explored, toxicities arising from antigen expression on normal tissues have slowed or halted several clinical trials. Here, we describe the development of TCEs that preferentially drive T cell-mediated death against target cells co-expressing two tumor-associated antigens. We show that Ly6E and B7-H4 are simultaneously expressed on approximately 50% of breast cancers, whereas normal tissue expression is limited and mostly orthogonal. Traditional bispecific TCEs targeting a singular antigen, either Ly6E or B7-H4, are active when paired with high-affinity CD3-engagers, but normal tissue expression presents a toxicity risk. Treatment with a murine cross-reactive B7-H4-TCE results in rapid and severe weight loss in mice along with damage to B7-H4-expressing tissues. To overcome on-target toxicity, we designed trispecific antibodies co-targeting Ly6E, B7-H4, and CD3 and characterized the impact of dual-antigen binding and the relative placement of each binding domain on tumor killing in vitro and in vivo. In vitro killing of tumor cells co-expressing both antigens correlates to the placement of the higher affinity B7-H4 binding domain, with only modest enhancements seen upon addition of Ly6E binding. In xenograft models, avid binding of appropriately designed trispecific TCEs enables tumor growth inhibition while evading the poor tolerability seen with active bispecific TCEs. Collectively these data highlight the potential for dual-antigen targeting to improve safety and efficacy, and expand the scope of tumors that may effectively be treated by TCEs.Abbreviations: Chimeric antigen receptor T cells (CAR-Ts), dual-antigen targeted T cell engagers (DAT-TCE), Fragment antigen-binding (Fab), Hematoxylin and eosin (H&E), Institutional Animal Care and Use Committee (IACUC), Immunoglobulin G (IgG), immunohistochemistry (IHC), NOD SCID gamma (NSG), peripheral blood mononuclear cells (PBMCs), surface plasmon resonance (SPR), T cell-engagers (TCEs).


Subject(s)
Antibodies, Bispecific , Receptors, Chimeric Antigen , Animals , Antibodies, Bispecific/pharmacology , Cell Line, Tumor , Eosine Yellowish-(YS) , Hematoxylin , Humans , Immunoglobulin G , Leukocytes, Mononuclear , Mice , Mice, SCID , T-Lymphocytes , Xenograft Model Antitumor Assays
6.
Nature ; 610(7930): 182-189, 2022 10.
Article in English | MEDLINE | ID: mdl-36131013

ABSTRACT

Most current therapies that target plasma membrane receptors function by antagonizing ligand binding or enzymatic activities. However, typical mammalian proteins comprise multiple domains that execute discrete but coordinated activities. Thus, inhibition of one domain often incompletely suppresses the function of a protein. Indeed, targeted protein degradation technologies, including proteolysis-targeting chimeras1 (PROTACs), have highlighted clinically important advantages of target degradation over inhibition2. However, the generation of heterobifunctional compounds binding to two targets with high affinity is complex, particularly when oral bioavailability is required3. Here we describe the development of proteolysis-targeting antibodies (PROTABs) that tether cell-surface E3 ubiquitin ligases to transmembrane proteins, resulting in target degradation both in vitro and in vivo. Focusing on zinc- and ring finger 3 (ZNRF3), a Wnt-responsive ligase, we show that this approach can enable colorectal cancer-specific degradation. Notably, by examining a matrix of additional cell-surface E3 ubiquitin ligases and transmembrane receptors, we demonstrate that this technology is amendable for 'on-demand' degradation. Furthermore, we offer insights on the ground rules governing target degradation by engineering optimized antibody formats. In summary, this work describes a strategy for the rapid development of potent, bioavailable and tissue-selective degraders of cell-surface proteins.


Subject(s)
Antibodies , Antibody Specificity , Membrane Proteins , Proteolysis , Ubiquitin-Protein Ligases , Animals , Antibodies/immunology , Antibodies/metabolism , Colorectal Neoplasms/metabolism , Ligands , Membrane Proteins/immunology , Membrane Proteins/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Substrate Specificity , Ubiquitin-Protein Ligases/immunology , Ubiquitin-Protein Ligases/metabolism
7.
Mol Pharm ; 19(5): 1540-1547, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35393854

ABSTRACT

Treatment of age-related macular degeneration (AMD) with anti-vascular endothelial growth factor (VEGF) biologic agents has been shown to restore and maintain visual acuity for many patients afflicted with wet AMD. These agents are usually administered via intravitreal injection at a dosing interval of 4-8 weeks. Employment of long-acting delivery (LAD) technologies could improve the therapeutic outcome, ensure timely treatment, and reduce burden on patients, caregivers, and the health care system. Development of LAD approaches requires thorough testing in pre-clinical species; however, therapeutic proteins of human origin may not be well tolerated during testing in non-human species due to immunogenicity. Here, we have engineered a surrogate porcine antibody Fab fragment (pigG6.31) from a human antibody for testing ocular LAD technologies in a porcine model. The engineered Fab retains the VEGF-A-binding and inhibition properties of the parental human Fab and has stability properties suitable for LAD evaluation. Upon intravitreal injection in minipigs, pigG6.31 showed first-order clearance from the ocular compartments with vitreal elimination rates consistent with other molecules of this size. Application of the surrogate molecule in an in vivo evaluation in minipigs of a prototype of the port delivery (PD) platform indicated continuous ocular delivery from the implant, with release kinetics consistent with both the results from in vitro release studies and the efficacy observed in human clinical studies of the PD system with ranibizumab (PDS). Anti-drug antibodies in the serum against pigG6.31 were not detected over exposure durations up to 16 weeks, suggesting that this molecule has low porcine immunogenicity.


Subject(s)
Angiogenesis Inhibitors , Wet Macular Degeneration , Animals , Humans , Immunoglobulin Fab Fragments/therapeutic use , Intravitreal Injections , Protein Engineering , Ranibizumab/therapeutic use , Swine , Swine, Miniature/metabolism , Technology , Vascular Endothelial Growth Factor A/metabolism , Wet Macular Degeneration/drug therapy
8.
MAbs ; 13(1): 1862452, 2021.
Article in English | MEDLINE | ID: mdl-33382956

ABSTRACT

Early success with brentuximab vedotin in treating classical Hodgkin lymphoma spurred an influx of at least 20 monomethyl auristatin E (MMAE) antibody-drug conjugates (ADCs) into clinical trials. While three MMAE-ADCs have been approved, most of these conjugates are no longer being investigated in clinical trials. Some auristatin conjugates show limited or no efficacy at tolerated doses, but even for drugs driving initial remissions, tumor regrowth and metastasis often rapidly occur. Here we describe the development of second-generation therapeutic ADCs targeting Lymphocyte antigen 6E (Ly6E) where the tubulin polymerization inhibitor MMAE (Compound 1) is replaced with DNA-damaging agents intended to drive increased durability of response. Comparison of a seco-cyclopropyl benzoindol-4-one (CBI)-dimer (compound 2) to MMAE showed increased potency, activity across more cell lines, and resistance to efflux by P-glycoprotein, a drug transporter commonly upregulated in tumors. Both anti-Ly6E-CBI and -MMAE conjugates drove single-dose efficacy in xenograft and patient-derived xenograft models, but seco-CBI-dimer conjugates showed reduced tumor outgrowth following multiple weeks of treatment, suggesting that they are less susceptible to developing resistance. In parallel, we explored approaches to optimize the targeting antibody. In contrast to immunization with recombinant Ly6E or Ly6E DNA, immunization with virus-like particles generated a high-affinity anti-Ly6E antibody. Conjugates to this antibody improve efficacy versus a previous clinical candidate both in vitro and in vivo with multiple cytotoxics. Conjugation of compound 2 to the second-generation antibody results in a substantially improved ADC with promising preclinical efficacy.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Surface/immunology , Antineoplastic Agents/immunology , Immunoconjugates/immunology , Oligopeptides/immunology , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Female , GPI-Linked Proteins/immunology , HEK293 Cells , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Mice, SCID , Rats, Sprague-Dawley , Tumor Burden/drug effects , Tumor Burden/immunology
9.
MAbs ; 12(1): 1818436, 2020.
Article in English | MEDLINE | ID: mdl-32936727

ABSTRACT

Treatment of ocular disease is hindered by the presence of the blood-retinal barrier, which restricts access of systemic drugs to the eye. Intravitreal injections bypass this barrier, delivering high concentrations of drug to the targeted tissue. However, the recommended dosing interval for approved biologics is typically 6-12 weeks, and frequent travel to the physician's office poses a substantial burden for elderly patients with poor vision. Real-world data suggest that many patients are under-treated. Here, we investigate IgMs as a novel platform for treating ocular disease. We show that IgMs are well-suited to ocular administration due to moderate viscosity, long ocular exposure, and rapid systemic clearance. The complement-dependent cytotoxicity of IgMs can be readily removed with a P436G mutation, reducing safety liabilities. Furthermore, dodecavalent binding of IgM hexamers can potently activate pathways implicated in the treatment of progressive blindness, including the Tie2 receptor tyrosine kinase signaling pathway for the treatment of diabetic macular edema, or the death receptor 4 tumor necrosis family receptor pathway for the treatment of wet age-related macular degeneration. Collectively, these data demonstrate the promise of IgMs as therapeutic agonists for treating progressive blindness.


Subject(s)
Drug Delivery Systems , Immunoglobulin M/pharmacology , Macular Degeneration , Vitreous Body/metabolism , Animals , CHO Cells , Cricetulus , Humans , Intravitreal Injections , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Rats
10.
MAbs ; 10(7): 1073-1083, 2018 10.
Article in English | MEDLINE | ID: mdl-30130444

ABSTRACT

Developability assessment of therapeutic antibody candidates assists drug discovery by enabling early identification of undesirable instabilities. Rapid chemical stability screening of antibody variants can accelerate the identification of potential solutions. We describe here the development of a high-throughput assay to characterize asparagine deamidation. We applied the assay to identify a mutation that unexpectedly stabilizes a critical asparagine. Ninety antibody variants were incubated under thermal stress in order to induce deamidation and screened for both affinity and total binding capacity. Surprisingly, a mutation five residues downstream from the unstable asparagine greatly reduced deamidation. Detailed assessment by LC-MS analysis confirmed the predicted improvement. This work describes both a high-throughput method for antibody stability screening during the early stages of antibody discovery and highlights the value of broad searches of antibody sequence space.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies/chemistry , Asparagine/chemistry , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Amides/chemistry , Animals , Antibody Affinity , Drug Discovery , High-Throughput Screening Assays , Humans , Mutation/genetics , Protein Binding , Protein Stability
11.
Org Lett ; 18(22): 5888-5891, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27802043

ABSTRACT

A scalable and efficient synthesis of the GPR40 agonist MK-8666 was developed from a simple pyridine building block. The key step to set the stereochemistry at two centers relied on an enzymatic dynamic kinetic reduction of an unactivated ketone. Directed evolution was leveraged to generate an optimized ketoreductase that provided the desired trans alcohol in >30:1 dr and >99% ee. Further, it was demonstrated that all four diastereomers of this hydroxy-ester could be prepared in high yield and selectivity. Subsequently, a challenging intramolecular displacement was carried out to form the cyclopropane ring system with perfect control of endo/exo selectivity. The endgame coupling strategy relied on a Pd-catalyzed C-O coupling to join the headpiece chloropyridine with the benzylic alcohol tailpiece.

12.
Nat Chem Biol ; 10(11): 969-76, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25262416

ABSTRACT

We previously discovered a small-molecule inducer of cell death, named 1541, that noncovalently self-assembles into chemical fibrils ('chemi-fibrils') and activates procaspase-3 in vitro. We report here that 1541-induced cell death is caused by the fibrillar rather than the soluble form of the drug. A short hairpin RNA screen reveals that knockdown of genes involved in endocytosis, vesicle trafficking and lysosomal acidification causes partial 1541 resistance. We confirm the role of these pathways using pharmacological inhibitors. Microscopy shows that the fluorescent chemi-fibrils accumulate in punctae inside cells that partially colocalize with lysosomes. Notably, the chemi-fibrils bind and induce liposome leakage in vitro, suggesting they may do the same in cells. The chemi-fibrils induce extensive proteolysis including caspase substrates, yet modulatory profiling reveals that chemi-fibrils form a distinct class from existing inducers of cell death. The chemi-fibrils share similarities with proteinaceous fibrils and may provide insight into their mechanism of cellular toxicity.


Subject(s)
Benzamides/chemistry , Benzamides/pharmacology , Caspase 3/metabolism , Coumarins/chemistry , Coumarins/pharmacology , Amino Acid Sequence , Cell Death/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , K562 Cells , Lysosomes/chemistry , Lysosomes/drug effects , Lysosomes/metabolism , Models, Biological , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured
13.
J Biol Chem ; 287(40): 33781-95, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22872644

ABSTRACT

Most proteases are expressed as inactive precursors, or zymogens, that become activated by limited proteolysis. We previously identified a small molecule, termed 1541, that dramatically promotes the maturation of the zymogen, procaspase-3, to its mature form, caspase-3. Surprisingly, compound 1541 self-assembles into nanofibrils, and localization of procaspase-3 to the fibrils promotes activation. Here, we interrogate the biochemical mechanism of procaspase-3 activation on 1541 fibrils in addition to proteogenic amyloid-ß(1-40) fibrils. In contrast to previous reports, we find no evidence that procaspase-3 alone is capable of self-activation, consistent with its fate-determining role in executing apoptosis. In fact, mature caspase-3 is >10(7)-fold more active than procaspase-3, making this proenzyme a remarkably inactive zymogen. However, we also show that fibril-induced colocalization of trace amounts of caspase-3 or other initiator proteases with procaspase-3 dramatically stimulates maturation of the proenzyme in vitro. Thus, similar to known cellular signaling complexes, these synthetic or natural fibrils can serve as platforms to concentrate procaspase-3 for trans-activation by upstream proteases.


Subject(s)
Apoptosis , Caspase 3/chemistry , Amyloid beta-Peptides/chemistry , Catalysis , Dimerization , Drug Delivery Systems , Enzyme Activation , Enzyme Precursors/chemistry , Humans , Kinetics , Models, Biological , Models, Chemical , Peptide Hydrolases/chemistry , Signal Transduction , Transcriptional Activation
14.
Proc Natl Acad Sci U S A ; 109(6): 1913-8, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308409

ABSTRACT

Mass spectrometry-based proteomics is a powerful tool for identifying hundreds to thousands of posttranslational modifications in complex mixtures. However, it remains enormously challenging to simultaneously assess the intrinsic catalytic efficiencies (k(cat)/K(M)) of these modifications in the context of their natural interactors. Such fundamental enzymological constants are key to determining substrate specificity and for establishing the timing and importance of cellular signaling. Here, we report the use of selected reaction monitoring (SRM) for tracking proteolysis induced by human apoptotic caspases-3, -7, -8, and -9 in lysates and living cells. By following the appearance of the cleaved peptides in lysate as a function of time, we were able to determine hundreds of catalytic efficiencies in parallel. Remarkably, we find the rates of substrate hydrolysis for individual caspases vary greater than 500-fold indicating a sequential process. Moreover, the rank-order of substrate cutting is similar in apoptotic cells, suggesting that cellular structures do not dramatically alter substrate accessibility. Comparisons of extrinsic (TRAIL) and intrinsic (staurosporine) inducers of apoptosis revealed similar substrate profiles, suggesting the final proteolytic demolitions proceed by similarly ordered plans. Certain biological processes were rapidly targeted by the caspases, including multiple components of the endocyotic pathway and miRNA processing machinery. We believe this massively parallel and quantitative label-free approach to obtaining basic enzymological constants will facilitate the study of proteolysis and other posttranslational modifications in complex mixtures.


Subject(s)
Proteolysis , Proteomics/methods , Amino Acid Sequence , Apoptosis/drug effects , Caspases/metabolism , Cell Survival/drug effects , Endocytosis/drug effects , Enzyme Activation/drug effects , Humans , Jurkat Cells , Kinetics , MicroRNAs/metabolism , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Proteolysis/drug effects , RNA Processing, Post-Transcriptional/drug effects , Staurosporine/pharmacology , Substrate Specificity/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology
15.
Mol Cell Proteomics ; 9(5): 880-93, 2010 May.
Article in English | MEDLINE | ID: mdl-20173201

ABSTRACT

The inflammatory caspases, human caspases-1, -4, and -5, proteolytically modulate diverse physiological outcomes in response to proinflammatory signals. Surprisingly, only a few substrates are known for these enzymes, including other caspases and the interleukin-1 family of cytokines. To more comprehensively characterize inflammatory caspase substrates, we combined an enzymatic N-terminal enrichment method with mass spectrometry-based proteomics to identify newly cleaved proteins. Analysis of THP-1 monocytic cell lysates treated with recombinant purified caspases identified 82 putative caspase-1 substrates, three putative caspase-4 substrates, and no substrates for caspase-5. By contrast, inflammatory caspases activated in THP-1 cells by mimics of gout (monosodium urate), bacterial infection (lipopolysaccharide and ATP), or viral infection (poly(dA.dT)) were found to cleave only 27, 16, and 22 substrates, respectively. Quantitative stable isotope labeling with amino acids in cell culture (SILAC) comparison of these three inflammatory stimuli showed that they induced largely overlapping substrate profiles but different extents of proteolysis. Interestingly, only half of the cleavages found in response to proinflammatory stimuli were contained within our set of 82 in vitro cleavage sites. These data provide the most comprehensive set of caspase-1-cleaved products reported to date and indicate that caspases-4 and -5 have far fewer substrates. Comparisons between the in vitro and in vivo data highlight the importance of localization in regulating inflammatory caspase activity. Finally, our data suggest that inducers of inflammation may subtly alter caspase-1 substrate profiles.


Subject(s)
Caspases/metabolism , Inflammation/enzymology , Amino Acid Sequence , Apoptosis , Biotinylation , Cell Extracts , Cell Line , Enzyme Activation , Humans , Hydrophobic and Hydrophilic Interactions , Inflammation/pathology , Isotope Labeling , Molecular Sequence Data , Peptide Synthases/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Biosynthesis , Protein Processing, Post-Translational , Proteomics , Substrate Specificity , Subtilisins/metabolism , Transcription, Genetic , Transfection
16.
Proc Natl Acad Sci U S A ; 107(5): 1821-6, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080615

ABSTRACT

Chemical reactions that enable selective biomolecule labeling in living organisms offer a means to probe biological processes in vivo. Very few reactions possess the requisite bioorthogonality, and, among these, only the Staudinger ligation between azides and triarylphosphines has been employed for direct covalent modification of biomolecules with probes in the mouse, an important model organism for studies of human disease. Here we explore an alternative bioorthogonal reaction, the 1,3-dipolar cycloaddition of azides and cyclooctynes, also known as "Cu-free click chemistry," for labeling biomolecules in live mice. Mice were administered peracetylated N-azidoacetylmannosamine (Ac(4)ManNAz) to metabolically label cell-surface sialic acids with azides. After subsequent injection with cyclooctyne reagents, glycoconjugate labeling was observed on isolated splenocytes and in a variety of tissues including the intestines, heart, and liver, with no apparent toxicity. The cyclooctynes tested displayed various labeling efficiencies that likely reflect the combined influence of intrinsic reactivity and bioavailability. These studies establish Cu-free click chemistry as a bioorthogonal reaction that can be executed in the physiologically relevant context of a mouse.


Subject(s)
Azides/metabolism , Glycoconjugates/metabolism , Hexosamines/metabolism , Animals , Azides/chemistry , Cell Membrane/metabolism , Copper/chemistry , Copper/metabolism , Cyclization , Cyclooctanes/chemistry , Cyclooctanes/metabolism , Glycoconjugates/chemistry , Hexosamines/chemistry , Humans , In Vitro Techniques , Indicators and Reagents , Jurkat Cells , Mice , Models, Biological , Molecular Probes/chemistry , Protein Binding , Serum Albumin/metabolism , Spleen/cytology , Spleen/metabolism
17.
Curr Opin Chem Biol ; 13(5-6): 503-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19729334

ABSTRACT

Proteolysis is a key regulatory post-translational modification in diverse cellular processes including programed cell death, immune function, and development. Tracking proteolytic events has become a focus of researchers assessing the downstream consequences of protease activation. In this review we summarize unbiased methods for identifying protease substrates and tracking the extent of cleavage, a field termed 'degradomics'. These include one-dimensional and two-dimensional gel-based methods for identifying protease substrates, N-terminal peptide identification methods for simultaneously identifying substrates and cleavage sites, and approaches for the quantitation of cleavage events during endogenous proteolysis. Individual methods have identified more than 300 caspase-cleaved targets during apoptosis suggesting broad future applications for these technologies.


Subject(s)
Peptide Hydrolases/metabolism , Proteomics/methods , Animals , Electrophoresis , Humans , Peptide Hydrolases/chemistry , Substrate Specificity
18.
Acc Chem Res ; 42(6): 788-97, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19361192

ABSTRACT

Glycosylation is an essential form of post-translational modification that regulates intracellular and extracellular processes. Regrettably, conventional biochemical and genetic methods often fall short for the study of glycans, because their structures are often not precisely defined at the genetic level. To address this deficiency, chemists have developed technologies to perturb glycan biosynthesis, profile their presentation at the systems level, and perceive their spatial distribution. These tools have identified potential disease biomarkers and ways to monitor dynamic changes to the glycome in living organisms. Still, glycosylation remains the underexplored frontier of many biological systems. In this Account, we focus on research in our laboratory that seeks to transform the study of glycan function from a challenge to routine practice. In studies of proteins and nucleic acids, functional studies have often relied on genetic manipulations to perturb structure. Though not directly subject to mutation, we can determine glycan structure-function relationships by synthesizing defined glycoconjugates or by altering natural glycosylation pathways. Chemical syntheses of uniform glycoproteins and polymeric glycoprotein mimics have facilitated the study of individual glycoconjugates in the absence of glycan microheterogeneity. Alternatively, selective inhibition or activation of glycosyltransferases or glycosidases can define the biological roles of the corresponding glycans. Investigators have developed tools including small molecule inhibitors, decoy substrates, and engineered proteins to modify cellular glycans. Current approaches offer a precision approaching that of genetic control. Genomic and proteomic profiling form a basis for biological discovery. Glycans also present a rich matrix of information that adapts rapidly to changing environs. Glycomic and glycoproteomic analyses via microarrays and mass spectrometry are beginning to characterize alterations in glycans that correlate with disease. These approaches have already identified several cancer biomarkers. Metabolic labeling can identify recently synthesized glycans and thus directly track glycan dynamics. This approach can highlight changes in physiology or environment and may be more informative than steady-state analyses. Together, glycomic and metabolic labeling techniques provide a comprehensive description of glycosylation as a foundation for hypothesis generation. Direct visualization of proteins via the green fluorescent protein (GFP) and its congeners has revolutionized the field of protein dynamics. Similarly, the ability to perceive the spatial organization of glycans could transform our understanding of their role in development, infection, and disease progression. Fluorescent tagging in cultured cells and developing organisms has revealed important insights into the dynamics of these structures during growth and development. These results have highlighted the need for additional imaging probes.


Subject(s)
Polysaccharides/chemistry , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/metabolism , Glycosylation , Glycosyltransferases/antagonists & inhibitors , Glycosyltransferases/metabolism , Green Fluorescent Proteins/chemistry , Polysaccharides/metabolism , Protein Engineering
19.
J Am Chem Soc ; 130(34): 11486-93, 2008 Aug 27.
Article in English | MEDLINE | ID: mdl-18680289

ABSTRACT

The 1,3-dipolar cycloaddition of azides and activated alkynes has been used for site-selective labeling of biomolecules in vitro and in vivo. While copper catalysis has been widely employed to activate terminal alkynes for [3 + 2] cycloaddition, this method, often termed "click chemistry", is currently incompatible with living systems because of the toxicity of the metal. We recently reported a difluorinated cyclooctyne (DIFO) reagent that rapidly reacts with azides in living cells without the need for copper catalysis. Here we report a novel class of DIFO reagents for copper-free click chemistry that are considerably more synthetically tractable. The new analogues maintained the same elevated rates of [3 + 2] cycloaddition as the parent compound and were used for imaging glycans on live cells. These second-generation DIFO reagents should expand the use of copper-free click chemistry in the hands of biologists.


Subject(s)
Alkynes/chemistry , Azides/chemistry , Cell Membrane/metabolism , Copper/chemistry , Hydrocarbons, Cyclic/chemistry , Hydrocarbons, Fluorinated/chemistry , Polysaccharides/analysis , Alkynes/metabolism , Animals , Azides/metabolism , CHO Cells , Catalysis , Cell Membrane/chemistry , Cells, Cultured , Cricetinae , Cricetulus , Cyclization , Fluorescein-5-isothiocyanate , Humans , Hydrocarbons, Cyclic/metabolism , Hydrocarbons, Fluorinated/metabolism , Jurkat Cells , Kinetics , Polysaccharides/chemistry , Polysaccharides/metabolism , Staining and Labeling
20.
Proc Natl Acad Sci U S A ; 104(43): 16793-7, 2007 Oct 23.
Article in English | MEDLINE | ID: mdl-17942682

ABSTRACT

Dynamic imaging of proteins in live cells is routinely performed by using genetically encoded reporters, an approach that cannot be extended to other classes of biomolecules such as glycans and lipids. Here, we report a Cu-free variant of click chemistry that can label these biomolecules rapidly and selectively in living systems, overcoming the intrinsic toxicity of the canonical Cu-catalyzed reaction. The critical reagent, a substituted cyclooctyne, possesses ring strain and electron-withdrawing fluorine substituents that together promote the [3 + 2] dipolar cycloaddition with azides installed metabolically into biomolecules. This Cu-free click reaction possesses comparable kinetics to the Cu-catalyzed reaction and proceeds within minutes on live cells with no apparent toxicity. With this technique, we studied the dynamics of glycan trafficking and identified a population of sialoglycoconjugates with unexpectedly rapid internalization kinetics.


Subject(s)
Chemistry, Organic/methods , Copper/metabolism , Imaging, Three-Dimensional/methods , Animals , Biological Transport , CHO Cells , Catalysis , Cell Survival , Cricetinae , Cricetulus , Endocytosis , Humans , Hydrocarbons, Cyclic/chemical synthesis , Hydrocarbons, Cyclic/chemistry , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Jurkat Cells , Kinetics , Polysaccharides/metabolism , Proteins/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...