Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Chem Biol ; 18(8): 850-858, 2022 08.
Article in English | MEDLINE | ID: mdl-35654846

ABSTRACT

The growing appreciation of immune cell-cell interactions within disease environments has led to extensive efforts to develop immunotherapies. However, characterizing complex cell-cell interfaces in high resolution remains challenging. Thus, technologies leveraging therapeutic-based modalities to profile intercellular environments offer opportunities to study cell-cell interactions with molecular-level insight. We introduce photocatalytic cell tagging (PhoTag) for interrogating cell-cell interactions using single-domain antibodies (VHHs) conjugated to photoactivatable flavin-based cofactors. Following irradiation with visible light, the flavin photocatalyst generates phenoxy radical tags for targeted labeling. Using this technology, we demonstrate selective synaptic labeling across the PD-1/PD-L1 axis in antigen-presenting cell-T cell systems. In combination with multiomics single-cell sequencing, we monitored interactions between peripheral blood mononuclear cells and Raji PD-L1 B cells, revealing differences in transient interactions with specific T cell subtypes. The utility of PhoTag in capturing cell-cell interactions will enable detailed profiling of intercellular communication across different biological systems.


Subject(s)
B7-H1 Antigen , Leukocytes, Mononuclear , Cell Communication , Flavins , Immunotherapy
2.
Chembiochem ; 21(24): 3555-3562, 2020 12 11.
Article in English | MEDLINE | ID: mdl-32749732

ABSTRACT

Despite the growing use of visible-light photochemistry in both chemistry and biology, no general low-heat photoreactor for use across these different disciplines exists. Herein, we describe the design and use of a standardized photoreactor for visible-light-driven activation and photocatalytic chemical transformations. Using this single benchtop photoreactor, we performed photoredox reactions across multiple visible light wavelengths, a high-throughput photocatalytic cross-coupling reaction, and in vitro labeling of proteins and live cells. Given the success of this reactor in all tested applications, we envision that this multi-use photoreactor will be widely used in biology, chemical biology, and medicinal chemistry settings.


Subject(s)
Biotin/analysis , Light , Photobioreactors , Tyramine/chemistry , Catalysis , Cell Line, Tumor , Equipment Design , Humans , Molecular Structure , Photochemical Processes , Tyramine/analogs & derivatives , Tyramine/chemical synthesis
3.
Curr Microbiol ; 76(4): 398-409, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30603964

ABSTRACT

Cell wall hydrolases are enzymes that cleave bacterial cell walls by hydrolyzing specific bonds within peptidoglycan and other portions of the envelope. Two major sources of hydrolases in nature are from hosts and microbes. This study specifically investigated whether cell wall hydrolytic enzymes could be employed as exogenous reagents to augment the efficacy of antimicrobial agents against mycobacteria. Mycobacterium smegmatis cultures were treated with ten conventional antibiotics and six anti-tuberculosis drugs-alone or in combination with cell wall hydrolases. Culture turbidity, colony-forming units (CFUs), vital staining, and oxygen consumption were all monitored. The majority of antimicrobial agents tested alone only had minimal inhibitory effects on bacterial growth. However, the combination of cell wall hydrolases and most of the antimicrobial agents tested, revealed a synergistic effect that resulted in significant enhancement of bactericidal activity. Vital staining showed increased cellular damage when M. smegmatis and Mycobacterium bovis bacillus Calmette-Guérin (M. bovis BCG) were treated with both drug and lysozyme. Respiration analysis revealed stress responses when cells were treated with lysozyme and drugs individually, and an acute increase in oxygen consumption when treated with both drug and lysozyme. Similar trends were also observed for the other three enzymes (hydrolase-30, RipA-His6 and RpfE-His6) evaluated. These findings demonstrated that cell wall hydrolytic enzymes, as a group of biological agents, have the capability to improve the potency of many current antimicrobial drugs and render ineffective antibiotics effective in killing mycobacteria. This combinatorial approach may represent an important strategy to eliminate drug-resistant bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Wall/enzymology , Hydrolases/metabolism , Mycobacterium/drug effects , Antitubercular Agents/pharmacology , Colony Count, Microbial , Drug Synergism , Microbial Viability/drug effects , Mycobacterium/enzymology , Mycobacterium/growth & development , Mycobacterium/metabolism , Mycobacterium bovis/drug effects , Mycobacterium bovis/growth & development , Mycobacterium bovis/metabolism , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/growth & development , Mycobacterium smegmatis/metabolism , Oxygen/metabolism
4.
Nat Chem Biol ; 10(9): 760-767, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25038787

ABSTRACT

Kinases are principal components of signal transduction pathways and the focus of intense basic and drug discovery research. Irreversible inhibitors that covalently modify non-catalytic cysteines in kinase active sites have emerged as valuable probes and approved drugs. Many protein classes, however, have functional cysteines, and therefore understanding the proteome-wide selectivity of covalent kinase inhibitors is imperative. Here, we accomplish this objective using activity-based protein profiling coupled with quantitative MS to globally map the targets, both specific and nonspecific, of covalent kinase inhibitors in human cells. Many of the specific off-targets represent nonkinase proteins that, notably, have conserved active site cysteines. We define windows of selectivity for covalent kinase inhibitors and show that, when these windows are exceeded, rampant proteome-wide reactivity and kinase target-independent cell death conjointly occur. Our findings, taken together, provide an experimental road map to illuminate opportunities and surmount challenges for the development of covalent kinase inhibitors.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proteome/genetics , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Cell Line, Tumor , Cell Survival/drug effects , Cysteine/chemistry , Genes, erbB-1/genetics , Humans , Kinetics , Piperidines , Protein Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
5.
Org Biomol Chem ; 14(26): 6179-83, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27216142

ABSTRACT

Proof of drug-target engagement in physiologically-relevant contexts is a key pillar of successful therapeutic target validation. We developed two orthogonal technologies, the cellular thermal shift assay (CETSA) and a covalent chemical probe reporter approach (harnessing sulfonyl fluoride tyrosine labeling and subsequent click chemistry) to measure the occupancy of the mRNA-decapping scavenger enzyme DcpS by a small molecule inhibitor in live cells. Enzyme affinity determined using isothermal dose response fingerprinting (ITDRFCETSA) and the concentration required to occupy 50% of the enzyme (OC50) using the chemical probe reporter assay were very similar. In this case, the chemical probe method worked well due to the long offset kinetics of the reversible inhibitor (determined using a fluorescent dye-tagged probe). This work suggests that CETSA could become the first choice assay to determine in-cell target engagement due to its simplicity.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/chemistry , Quinazolines/pharmacology , Temperature , Click Chemistry , Dose-Response Relationship, Drug , Endoribonucleases/metabolism , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Molecular Structure , Quinazolines/chemistry , Sulfinic Acids/chemistry , Tyrosine/chemistry
6.
Proc Natl Acad Sci U S A ; 110(24): 9932-7, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23716698

ABSTRACT

Bacterial toxins have evolved successful strategies for coopting host proteins to access the cytosol of host cells. Anthrax lethal factor (LF) enters the cytosol through pores in the endosomal membrane formed by anthrax protective antigen. Although in vitro models using planar lipid bilayers have shown that translocation can occur in the absence of cellular factors, recent studies using intact endosomes indicate that host factors are required for translocation in the cellular environment. In this study, we describe a high-throughput shRNA screen to identify host factors required for anthrax lethal toxin-induced cell death. The cytosolic chaperonin complex chaperonin containing t-complex protein 1 (CCT) was identified, and subsequent studies showed that CCT is required for efficient delivery of LF and related fusion proteins into the cytosol. We further show that knockdown of CCT inhibits the acid-induced delivery of LF and the fusion protein LFN-Bla (N terminal domain of LF fused to ß-lactamase) across the plasma membrane of intact cells. Together, these results suggest that CCT is required for efficient delivery of enzymatically active toxin to the cytosol and are consistent with a direct role for CCT in translocation of LF through the protective antigen pore.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Chaperonin Containing TCP-1/metabolism , Cytosol/metabolism , Animals , Bacillus anthracis/physiology , Blotting, Western , Cell Line , Chaperonin Containing TCP-1/genetics , Cytosol/microbiology , Endosomes/metabolism , Host-Pathogen Interactions , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology , Mice , Protein Transport/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , beta-Lactamases/genetics , beta-Lactamases/metabolism
7.
Nat Chem Biol ; 9(6): 398-405, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23603659

ABSTRACT

Formation of the inflammasome, a scaffolding complex that activates caspase-1, is important in numerous diseases. Pyroptotic cell death induced by anthrax lethal toxin (LT) is a model for inflammasome-mediated caspase-1 activation. We discovered 7-desacetoxy-6,7-dehydrogedunin (7DG) in a phenotypic screen as a small molecule that protects macrophages from LT-induced death. Using chemical proteomics, we identified protein kinase R (PKR) as the target of 7DG and show that RNAi knockdown of PKR phenocopies treatment with 7DG. Further, we show that PKR's role in ASC assembly and caspase-1 activation induced by several different inflammasome stimuli is independent of PKR's kinase activity, demonstrating that PKR has a previously uncharacterized role in caspase-1 activation and pyroptosis that is distinct from its reported kinase-dependent roles in apoptosis and inflammasome formation in lipopolysaccharide-primed cells. Remarkably, PKR has different roles in two distinct cell death pathways and has a broad role in inflammasome function relevant in other diseases.


Subject(s)
Cell Death , eIF-2 Kinase/chemistry , Animals , Bacillus anthracis/enzymology , Caspase 1/metabolism , Catalytic Domain , Cell Line , Enzyme-Linked Immunosorbent Assay , HSP90 Heat-Shock Proteins/metabolism , Hydrogen-Ion Concentration , Inflammation , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Models, Biological , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Conformation
8.
Cell Chem Biol ; 30(10): 1313-1322.e7, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37499664

ABSTRACT

Identifying virus-host interactions on the cell surface can improve our understanding of viral entry and pathogenesis. SARS-CoV-2, the causative agent of the COVID-19 disease, uses ACE2 as a receptor to enter cells. Yet the full repertoire of cell surface proteins that contribute to viral entry is unknown. We developed a photocatalyst-based viral-host protein microenvironment mapping platform (ViraMap) to probe the molecular neighborhood of the SARS-CoV-2 spike protein on the human cell surface. Application of ViraMap to ACE2-expressing cells captured ACE2, the established co-receptor NRP1, and several novel cell surface proteins. We systematically analyzed the relevance of these candidate proteins to SARS-CoV-2 entry by knockdown and overexpression approaches in pseudovirus and authentic infection models and identified PTGFRN and EFNB1 as bona fide viral entry factors. Our results highlight additional host targets that participate in SARS-CoV-2 infection and showcase ViraMap as a powerful platform for defining viral interactions on the cell surface.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Angiotensin-Converting Enzyme 2 , Spike Glycoprotein, Coronavirus , Viral Proteins/metabolism , Protein Binding
9.
Bioeng Transl Med ; 8(5): e10542, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37693049

ABSTRACT

Cyclic peptides are poised to target historically difficult to drug intracellular protein-protein interactions, however, their general cell impermeability poses a challenge for characterizing function. Recent advances in microfluidics have enabled permeabilization of the cytoplasmic membrane by physical cell deformation (i.e., mechanoporation), resulting in intracellular delivery of impermeable macromolecules in vector- and electrophoretic-free approaches. However, the number of payloads (e.g., peptides) and/or concentrations delivered via microfluidic mechanoporation is limited by having to pre-mix cells and payloads, a manually intensive process. In this work, we show that cells are momentarily permeable (t 1/2 = 1.1-2.8 min) after microfluidic vortex shedding (µVS) and that lower molecular weight macromolecules can be cytosolically delivered upon immediate exposure after cells are processed/permeabilized. To increase the ability to screen peptides, we built a system, dispensing-microfluidic vortex shedding (DµVS), that integrates a µVS chip with inline microplate-based dispensing. To do so, we synced an electronic pressure regulator, flow sensor, on/off dispense valve, and an x-y motion platform in a software-driven feedback loop. Using this system, we were able to deliver low microliter-scale volumes of transiently mechanoporated cells to hundreds of wells on microtiter plates in just several minutes (e.g., 96-well plate filled in <2.5 min). We validated the delivery of an impermeable peptide directed at MDM2, a negative regulator of the tumor suppressor p53, using a click chemistry- and NanoBRET-based cell permeability assay in 96-well format, with robust delivery across the full plate. Furthermore, we demonstrated that DµVS could be used to identify functional, low micromolar, cellular activity of otherwise cell-inactive MDM2-binding peptides using a p53 reporter cell assay in 96- and 384-well format. Overall, DµVS can be combined with downstream cell assays to investigate intracellular target engagement in a high-throughput manner, both for improving structure-activity relationship efforts and for early proof-of-biology of non-optimized peptide (or potentially other macromolecular) tools.

10.
PLoS Pathog ; 6(7): e1001020, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20686708

ABSTRACT

Bacterial cell growth and division require coordinated cell wall hydrolysis and synthesis, allowing for the removal and expansion of cell wall material. Without proper coordination, unchecked hydrolysis can result in cell lysis. How these opposing activities are simultaneously regulated is poorly understood. In Mycobacterium tuberculosis, the resuscitation-promoting factor B (RpfB), a lytic transglycosylase, interacts and synergizes with Rpf-interacting protein A (RipA), an endopeptidase, to hydrolyze peptidoglycan. However, it remains unclear what governs this synergy and how it is coordinated with cell wall synthesis. Here we identify the bifunctional peptidoglycan-synthesizing enzyme, penicillin binding protein 1 (PBP1), as a RipA-interacting protein. PBP1, like RipA, localizes both at the poles and septa of dividing cells. Depletion of the ponA1 gene, encoding PBP1 in M. smegmatis, results in a severe growth defect and abnormally shaped cells, indicating that PBP1 is necessary for viability and cell wall stability. Finally, PBP1 inhibits the synergistic hydrolysis of peptidoglycan by the RipA-RpfB complex in vitro. These data reveal a post-translational mechanism for regulating cell wall hydrolysis and synthesis through protein-protein interactions between enzymes with antagonistic functions.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/enzymology , Cytokines/metabolism , Endopeptidases/metabolism , Mycobacterium/enzymology , Peptidoglycan Glycosyltransferase/metabolism , Hydrolysis
12.
Cell Rep ; 38(8): 110399, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35139367

ABSTRACT

Follicular helper T (Tfh) cells promote, whereas follicular regulatory T (Tfr) cells restrain, germinal center (GC) reactions. However, the precise roles of these cells in the complex GC reaction remain poorly understood. Here, we perturb Tfh or Tfr cells after SARS-CoV-2 spike protein vaccination in mice. We find that Tfh cells promote the frequency and somatic hypermutation (SHM) of Spike-specific GC B cells and regulate clonal diversity. Tfr cells similarly control SHM and clonal diversity in the GC but do so by limiting clonal competition. In addition, deletion of Tfh or Tfr cells during primary vaccination results in changes in SHM after vaccine boosting. Aged mice, which have altered Tfh and Tfr cells, have lower GC responses, presenting a bimodal distribution of SHM. Together, these data demonstrate that GC responses to SARS-CoV-2 spike protein vaccines require a fine balance of positive and negative follicular T cell help to optimize humoral immunity.


Subject(s)
COVID-19/prevention & control , Germinal Center/immunology , Spike Glycoprotein, Coronavirus/administration & dosage , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Aging , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , COVID-19/virology , Germinal Center/cytology , Germinal Center/metabolism , Immunity, Humoral , Mice , Mice, Inbred C57BL , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Vaccination , Vaccines, Subunit/immunology
13.
Proc Natl Acad Sci U S A ; 105(1): 264-9, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18172199

ABSTRACT

Tuberculosis remains a major global health problem that kills up to 2 million people annually. Central to the success of Mycobacterium tuberculosis (Mtb) as a pathogen is its ability to evade host immunity and to establish a chronic infection. Although its primary intracellular niche is within macrophages, the underlying molecular mechanisms are poorly understood. Here we show that Rv2224c, a cell envelope-associated predicted protease, is critical for Mtb virulence. Disruption of Rv2224c led to prolonged survival of infected mice and highly reduced lung pathology. Absence of Rv2224c enhanced host innate immune responses, compromised the intracellular survival of Mtb in macrophages, and increased its susceptibility to lysozyme. We provide insights into the molecular basis for Rv2224c function by showing that Rv2224c activity promotes processing and extracellular release of the Mtb protein, GroEL2. Inhibition of Rv2224c and its targets offers opportunities for therapeutic interventions and immune-modulatory strategies.


Subject(s)
Immune System/physiology , Macrophages/microbiology , Mycobacterium tuberculosis/metabolism , Tuberculosis/microbiology , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cell Survival , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Muramidase/metabolism , Mycobacterium bovis/metabolism , Plasmids/metabolism , Time Factors
14.
RSC Chem Biol ; 2(1): 30-46, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-34458775

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the current coronavirus disease 2019 (COVID-19) pandemic that has led to a global economic disruption and collapse. With several ongoing efforts to develop vaccines and treatments for COVID-19, understanding the molecular interaction between the coronavirus, host cells, and the immune system is critical for effective therapeutic interventions. Greater insight into these mechanisms will require the contribution and combination of multiple scientific disciplines including the techniques and strategies that have been successfully deployed by chemical biology to tease apart complex biological pathways. We highlight in this review well-established strategies and methods to study coronavirus-host biophysical interactions and discuss the impact chemical biology will have on understanding these interactions at the molecular level.

15.
PLoS Pathog ; 4(2): e1000001, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18463693

ABSTRACT

The final stage of bacterial cell division requires the activity of one or more enzymes capable of degrading the layers of peptidoglycan connecting two recently developed daughter cells. Although this is a key step in cell division and is required by all peptidoglycan-containing bacteria, little is known about how these potentially lethal enzymes are regulated. It is likely that regulation is mediated, at least partly, through protein-protein interactions. Two lytic transglycosylases of mycobacteria, known as resuscitation-promoting factor B and E (RpfB and RpfE), have previously been shown to interact with the peptidoglycan-hydrolyzing endopeptidase, Rpf-interacting protein A (RipA). These proteins may form a complex at the septum of dividing bacteria. To investigate the function of this potential complex, we generated depletion strains in M. smegmatis. Here we show that, while depletion of rpfB has no effect on viability or morphology, ripA depletion results in a marked decrease in growth and formation of long, branched chains. These growth and morphological defects could be functionally complemented by the M. tuberculosis ripA orthologue (rv1477), but not by another ripA-like orthologue (rv1478). Depletion of ripA also resulted in increased susceptibility to the cell wall-targeting beta-lactams. Furthermore, we demonstrate that RipA has hydrolytic activity towards several cell wall substrates and synergizes with RpfB. These data reveal the unusual essentiality of a peptidoglycan hydrolase and suggest a novel protein-protein interaction as one way of regulating its activity.


Subject(s)
Bacterial Proteins/physiology , Cell Division/physiology , Cytokines/physiology , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , N-Acetylmuramoyl-L-alanine Amidase/physiology , Aconitate Hydratase/physiology , Anti-Bacterial Agents/pharmacology , Cell Wall/enzymology , Drug Synergism , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Gene Silencing , Genes, Bacterial , Microbial Sensitivity Tests , Microbial Viability , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/genetics , Recombinant Proteins/biosynthesis
16.
Sci Rep ; 10(1): 5321, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210258

ABSTRACT

Recent reports show that colorectal tumors contain microbiota that are distinct from those that reside in a 'normal' colon environment, and that these microbiota can contribute to cancer progression. Fusobacterium nucleatum is the most commonly observed species in the colorectal tumor microenvironment and reportedly influences disease progression through numerous mechanisms. However, a detailed understanding of the role of this organism in cancer progression is limited, in part due to challenges in maintaining F. nucleatum viability under standard aerobic cell culture conditions. Herein we describe the development of a 3-dimensional (3D) tumor spheroid model that can harbor and promote the growth of anaerobic bacteria. Bacteria-tumor cell interactions and metabolic crosstalk were extensively studied by measuring the kinetics of bacterial growth, cell morphology and lysis, cancer-related gene expression, and metabolomics. We observed that viable F. nucleatum assembles biofilm-like structures in the tumor spheroid microenvironment, whereas heat-killed F. nucleatum is internalized and sequestered in the cancer cells. Lastly, we use the model to co-culture 28 Fusobacterium clinical isolates and demonstrate that the model successfully supports co-culture with diverse fusobacterial species. This bacteria-spheroid co-culture model enables mechanistic investigation of the role of anaerobic bacteria in the tumor microenvironment.


Subject(s)
Cell Culture Techniques/methods , Colorectal Neoplasms/microbiology , Spheroids, Cellular/metabolism , Bacteria, Anaerobic , Cell Line, Tumor , Coculture Techniques/methods , Colorectal Neoplasms/pathology , Disease Progression , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/metabolism , Fusobacterium nucleatum/pathogenicity , Humans , Models, Biological , Tumor Microenvironment/physiology
17.
Science ; 367(6482): 1091-1097, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32139536

ABSTRACT

Many disease pathologies can be understood through the elucidation of localized biomolecular networks, or microenvironments. To this end, enzymatic proximity labeling platforms are broadly applied for mapping the wider spatial relationships in subcellular architectures. However, technologies that can map microenvironments with higher precision have long been sought. Here, we describe a microenvironment-mapping platform that exploits photocatalytic carbene generation to selectively identify protein-protein interactions on cell membranes, an approach we term MicroMap (µMap). By using a photocatalyst-antibody conjugate to spatially localize carbene generation, we demonstrate selective labeling of antibody binding targets and their microenvironment protein neighbors. This technique identified the constituent proteins of the programmed-death ligand 1 (PD-L1) microenvironment in live lymphocytes and selectively labeled within an immunosynaptic junction.


Subject(s)
B7-H1 Antigen/metabolism , Cell Membrane/metabolism , Cellular Microenvironment , Lymphocytes/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Catalysis , Cell Membrane/radiation effects , Energy Transfer , Humans , Jurkat Cells , Lymphocytes/radiation effects , Methane/analogs & derivatives , Methane/chemistry , Methane/radiation effects , Photochemical Processes , Ultraviolet Rays
18.
Sci Signal ; 11(541)2018 07 31.
Article in English | MEDLINE | ID: mdl-30065029

ABSTRACT

Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin ß receptor (LTßR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTßR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Osteosarcoma/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cyclins/metabolism , Gene Expression Profiling , High-Throughput Screening Assays , Humans , Indoles/pharmacology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Propionates/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteome , Signal Transduction , TWEAK Receptor/antagonists & inhibitors , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Tumor Cells, Cultured , NF-kappaB-Inducing Kinase
19.
ACS Chem Biol ; 12(8): 2015-2020, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28718624

ABSTRACT

Irreversible enzyme inhibitors and covalent chemical biology probes often utilize the reaction of a protein cysteine residue with an appropriately positioned electrophile (e.g., acrylamide) on the ligand template. However, cysteine residues are not always available for site-specific protein labeling, and therefore new approaches are needed to expand the toolkit of appropriate electrophiles ("warheads") that target alternative amino acids. We previously described the rational targeting of tyrosine residues in the active site of a protein (the mRNA decapping scavenger enzyme, DcpS) using inhibitors armed with a sulfonyl fluoride electrophile. These inhibitors subsequently enabled the development of clickable probe technology to measure drug-target occupancy in live cells. Here we describe a fluorosulfate-containing inhibitor (aryl fluorosulfate probe (FS-p1)) with excellent chemical and metabolic stability that reacts selectively with a noncatalytic serine residue in the same active site of DcpS as confirmed by peptide mapping experiments. Our results suggest that noncatalytic serine targeting using fluorosulfate electrophilic warheads could be a suitable strategy for the development of covalent inhibitor drugs and chemical probes.


Subject(s)
Enzyme Inhibitors/chemistry , Fluorides/chemistry , Serine/chemistry , Sulfuric Acids/chemistry , Animals , Catalytic Domain , Cell Line , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Enzyme Stability , Humans
20.
ACS Comb Sci ; 18(10): 611-615, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27494431

ABSTRACT

Small molecule selectivity is an essential component of candidate drug selection and target validation. New technologies are required to better understand off-target effects, with particular emphasis needed on broad protein profiling. Here, we describe the use of a tritiated chemical probe and a 9000 human protein microarray to discern the binding selectivity of an inhibitor of the mRNA decapping scavenger enzyme DcpS. An immobilized m7GTP resin was also used to assess the selectivity of a DcpS inhibitor against mRNA cap-associated proteins in whole cell extracts. These studies confirm the exquisite selectivity of diaminoquinazoline DcpS inhibitors, and highlight the utility of relatively simple protein microarray and affinity enrichment technologies in drug discovery and chemical biology.


Subject(s)
Endoribonucleases/analysis , Molecular Probes/chemistry , Quinazolines/chemistry , RNA Cap-Binding Proteins/analysis , Catalysis , Cells, Cultured , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Humans , Leukocytes, Mononuclear/chemistry , Protein Array Analysis , RNA, Messenger/genetics , Survival of Motor Neuron 2 Protein/analysis , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL