Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nat Immunol ; 23(7): 1031-1041, 2022 07.
Article in English | MEDLINE | ID: mdl-35761082

ABSTRACT

The immune checkpoint receptor lymphocyte activation gene 3 protein (LAG3) inhibits T cell function upon binding to major histocompatibility complex class II (MHC class II) or fibrinogen-like protein 1 (FGL1). Despite the emergence of LAG3 as a target for next-generation immunotherapies, we have little information describing the molecular structure of the LAG3 protein or how it engages cellular ligands. Here we determined the structures of human and murine LAG3 ectodomains, revealing a dimeric assembly mediated by Ig domain 2. Epitope mapping indicates that a potent LAG3 antagonist antibody blocks interactions with MHC class II and FGL1 by binding to a flexible 'loop 2' region in LAG3 domain 1. We also defined the LAG3-FGL1 interface by mapping mutations onto structures of LAG3 and FGL1 and established that FGL1 cross-linking induces the formation of higher-order LAG3 oligomers. These insights can guide LAG3-based drug development and implicate ligand-mediated LAG3 clustering as a mechanism for disrupting T cell activation.


Subject(s)
Antigens, CD/metabolism , Lymphocyte Activation , Animals , Antibodies , Fibrinogen , Histocompatibility Antigens Class II/metabolism , Humans , Immunotherapy , Ligands , Mice , Receptors, Immunologic , Lymphocyte Activation Gene 3 Protein
2.
Immunity ; 54(6): 1154-1167.e7, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33979578

ABSTRACT

Blockade of the inhibitory receptor TIM-3 shows efficacy in cancer immunotherapy clinical trials. TIM-3 inhibits production of the chemokine CXCL9 by XCR1+ classical dendritic cells (cDC1), thereby limiting antitumor immunity in mammary carcinomas. We found that increased CXCL9 expression by splenic cDC1s upon TIM-3 blockade required type I interferons and extracellular DNA. Chemokine expression as well as combinatorial efficacy of TIM-3 blockade and paclitaxel chemotherapy were impaired by deletion of Cgas and Sting. TIM-3 blockade increased uptake of extracellular DNA by cDC1 through an endocytic process that resulted in cytoplasmic localization. DNA uptake and efficacy of TIM-3 blockade required DNA binding by HMGB1, while galectin-9-induced cell surface clustering of TIM-3 was necessary for its suppressive function. Human peripheral blood cDC1s also took up extracellular DNA upon TIM-3 blockade. Thus, TIM-3 regulates endocytosis of extracellular DNA and activation of the cytoplasmic DNA sensing cGAS-STING pathway in cDC1s, with implications for understanding the mechanisms underlying TIM-3 immunotherapy.


Subject(s)
DNA/metabolism , Dendritic Cells/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction/physiology , Animals , Biological Transport/physiology , Cell Line , Cell Line, Tumor , Chemokines/metabolism , Cytoplasm/metabolism , Endocytosis/physiology , Female , HEK293 Cells , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL
3.
Nature ; 587(7834): 499-504, 2020 11.
Article in English | MEDLINE | ID: mdl-32698187

ABSTRACT

The G-protein-coupled bile acid receptor (GPBAR) conveys the cross-membrane signalling of a vast variety of bile acids and is a signalling hub in the liver-bile acid-microbiota-metabolism axis1-3. Here we report the cryo-electron microscopy structures of GPBAR-Gs complexes stabilized by either the high-affinity P3954 or the semisynthesized bile acid derivative INT-7771,3 at 3 Å resolution. These structures revealed a large oval pocket that contains several polar groups positioned to accommodate the amphipathic cholic core of bile acids, a fingerprint of key residues to recognize diverse bile acids in the orthosteric site, a putative second bile acid-binding site with allosteric properties and structural features that contribute to bias properties. Moreover, GPBAR undertakes an atypical mode of activation and G protein coupling that features a different set of key residues connecting the ligand-binding pocket to the Gs-coupling site, and a specific interaction motif that is localized in intracellular loop 3. Overall, our study not only reveals unique structural features of GPBAR that are involved in bile acid recognition and allosteric effects, but also suggests the presence of distinct connecting mechanisms between the ligand-binding pocket and the G-protein-binding site in the G-protein-coupled receptor superfamily.


Subject(s)
Bile Acids and Salts/metabolism , Cryoelectron Microscopy , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/ultrastructure , Allosteric Regulation/drug effects , Bile Acids and Salts/chemistry , Binding Sites/drug effects , Cholic Acids/chemistry , Cholic Acids/pharmacology , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , Humans , Ligands , Models, Molecular , Protein Binding , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Substrate Specificity
4.
Nat Chem Biol ; 19(1): 9-17, 2023 01.
Article in English | MEDLINE | ID: mdl-36050494

ABSTRACT

The Notch pathway regulates cell fate decisions and is an emerging target for regenerative and cancer therapies. Recombinant Notch ligands are attractive candidates for modulating Notch signaling; however, their intrinsically low receptor-binding affinity restricts their utility in biomedical applications. To overcome this limitation, we evolved variants of the ligand Delta-like 4 with enhanced affinity and cross-reactivity. A consensus variant with maximized binding affinity, DeltaMAX, binds human and murine Notch receptors with 500- to 1,000-fold increased affinity compared with wild-type human Delta-like 4. DeltaMAX also potently activates Notch in plate-bound, bead-bound and cellular formats. When administered as a soluble decoy, DeltaMAX inhibits Notch in reporter and neuronal differentiation assays, highlighting its dual utility as an agonist or antagonist. Finally, we demonstrate that DeltaMAX stimulates increased proliferation and expression of effector mediators in T cells. Taken together, our data define DeltaMAX as a versatile tool for broad-spectrum activation or inhibition of Notch signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Intercellular Signaling Peptides and Proteins , Humans , Animals , Mice , Ligands , Adaptor Proteins, Signal Transducing/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Calcium-Binding Proteins/metabolism , Signal Transduction/physiology , Receptors, Notch/metabolism
5.
Immunity ; 42(5): 815-25, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25992858

ABSTRACT

Interleukin-2 (IL-2) is a pleiotropic cytokine that regulates immune cell homeostasis and has been used to treat a range of disorders including cancer and autoimmune disease. IL-2 signals via interleukin-2 receptor-ß (IL-2Rß):IL-2Rγ heterodimers on cells expressing high (regulatory T cells, Treg) or low (effector cells) amounts of IL-2Rα (CD25). When complexed with IL-2, certain anti-cytokine antibodies preferentially stimulate expansion of Treg (JES6-1) or effector (S4B6) cells, offering a strategy for targeted disease therapy. We found that JES6-1 sterically blocked the IL-2:IL-2Rß and IL-2:IL-2Rγ interactions, but also allosterically lowered the IL-2:IL-2Rα affinity through a "triggered exchange" mechanism favoring IL-2Rα(hi) Treg cells, creating a positive feedback loop for IL-2Rα(hi) cell activation. Conversely, S4B6 sterically blocked the IL-2:IL-2Rα interaction, while also conformationally stabilizing the IL-2:IL-2Rß interaction, thus stimulating all IL-2-responsive immune cells, particularly IL-2Rß(hi) effector cells. These insights provide a molecular blueprint for engineering selectively potentiating therapeutic antibodies.


Subject(s)
Antibodies/immunology , Interleukin-2/metabolism , Models, Molecular , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , Antibodies/chemistry , Antibodies/pharmacology , Autoimmune Diseases/immunology , Binding, Competitive/drug effects , Cell Proliferation/drug effects , Disease Progression , Flow Cytometry , Gene Expression Regulation/immunology , Humans , Interleukin-2/chemistry , Interleukin-2/genetics , Interleukin-2/immunology , Mice , Mice, Inbred BALB C , Protein Binding/drug effects , Protein Structure, Tertiary , Signal Transduction/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
6.
Nature ; 545(7654): 360-364, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28489825

ABSTRACT

The Notch signalling pathway mediates cell fate decisions and is tumour suppressive or oncogenic depending on the context. During lung development, Notch pathway activation inhibits the differentiation of precursor cells to a neuroendocrine fate. In small-cell lung cancer, an aggressive neuroendocrine lung cancer, loss-of-function mutations in NOTCH genes and the inhibitory effects of ectopic Notch activation indicate that Notch signalling is tumour suppressive. Here we show that Notch signalling can be both tumour suppressive and pro-tumorigenic in small-cell lung cancer. Endogenous activation of the Notch pathway results in a neuroendocrine to non-neuroendocrine fate switch in 10-50% of tumour cells in a mouse model of small-cell lung cancer and in human tumours. This switch is mediated in part by Rest (also known as Nrsf), a transcriptional repressor that inhibits neuroendocrine gene expression. Non-neuroendocrine Notch-active small-cell lung cancer cells are slow growing, consistent with a tumour-suppressive role for Notch, but these cells are also relatively chemoresistant and provide trophic support to neuroendocrine tumour cells, consistent with a pro-tumorigenic role. Importantly, Notch blockade in combination with chemotherapy suppresses tumour growth and delays relapse in pre-clinical models. Thus, small-cell lung cancer tumours generate their own microenvironment via activation of Notch signalling in a subset of tumour cells, and the presence of these cells may serve as a biomarker for the use of Notch pathway inhibitors in combination with chemotherapy in select patients with small-cell lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Receptors, Notch/metabolism , Signal Transduction , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Microenvironment , Animals , Cell Differentiation , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Lung Neoplasms/drug therapy , Male , Mice , Neoplasm Recurrence, Local/prevention & control , Receptors, Notch/agonists , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/deficiency , Repressor Proteins/metabolism , Small Cell Lung Carcinoma/drug therapy
7.
Nature ; 545(7653): 238-242, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28467820

ABSTRACT

The canonical Wnt/ß-catenin signalling pathway governs diverse developmental, homeostatic and pathological processes. Palmitoylated Wnt ligands engage cell-surface frizzled (FZD) receptors and LRP5 and LRP6 co-receptors, enabling ß-catenin nuclear translocation and TCF/LEF-dependent gene transactivation. Mutations in Wnt downstream signalling components have revealed diverse functions thought to be carried out by Wnt ligands themselves. However, redundancy between the 19 mammalian Wnt proteins and 10 FZD receptors and Wnt hydrophobicity have made it difficult to attribute these functions directly to Wnt ligands. For example, individual mutations in Wnt ligands have not revealed homeostatic phenotypes in the intestinal epithelium-an archetypal canonical, Wnt pathway-dependent, rapidly self-renewing tissue, the regeneration of which is fueled by proliferative crypt Lgr5+ intestinal stem cells (ISCs). R-spondin ligands (RSPO1-RSPO4) engage distinct LGR4-LGR6, RNF43 and ZNRF3 receptor classes, markedly potentiate canonical Wnt/ß-catenin signalling, and induce intestinal organoid growth in vitro and Lgr5+ ISCs in vivo. However, the interchangeability, functional cooperation and relative contributions of Wnt versus RSPO ligands to in vivo canonical Wnt signalling and ISC biology remain unknown. Here we identify the functional roles of Wnt and RSPO ligands in the intestinal crypt stem-cell niche. We show that the default fate of Lgr5+ ISCs is to differentiate, unless both RSPO and Wnt ligands are present. However, gain-of-function studies using RSPO ligands and a new non-lipidated Wnt analogue reveal that these ligands have qualitatively distinct, non-interchangeable roles in ISCs. Wnt proteins are unable to induce Lgr5+ ISC self-renewal, but instead confer a basal competency by maintaining RSPO receptor expression that enables RSPO ligands to actively drive and specify the extent of stem-cell expansion. This functionally non-equivalent yet cooperative interaction between Wnt and RSPO ligands establishes a molecular precedent for regulation of mammalian stem cells by distinct priming and self-renewal factors, with broad implications for precise control of tissue regeneration.


Subject(s)
Cell Self Renewal , Intestines/cytology , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Thrombospondins/metabolism , Wnt Proteins/metabolism , Animals , Cell Lineage , Cell Proliferation , Female , Humans , Ligands , Male , Mice , Organoids/cytology , Organoids/growth & development , Single-Cell Analysis , Stem Cell Niche , Transcriptome , Ubiquitin-Protein Ligases/metabolism , beta Catenin/metabolism
8.
Nat Chem Biol ; 14(1): 65-71, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29176671

ABSTRACT

Notch is a cell-surface receptor that controls cell-fate decisions and is regulated by O-glycans attached to epidermal growth factor-like (EGF) repeats in its extracellular domain. Protein O-fucosyltransferase 1 (Pofut1) modifies EGF repeats with O-fucose and is essential for Notch signaling. Constitutive activation of Notch signaling has been associated with a variety of human malignancies. Therefore, tools that inhibit Notch activity are being developed as cancer therapeutics. To this end, we screened L-fucose analogs for their effects on Notch signaling. Two analogs, 6-alkynyl and 6-alkenyl fucose, were substrates of Pofut1 and were incorporated directly into Notch EGF repeats in cells. Both analogs were potent inhibitors of binding to and activation of Notch1 by Notch ligands Dll1 and Dll4, but not by Jag1. Mutagenesis and modeling studies suggest that incorporation of the analogs into EGF8 of Notch1 markedly reduces the ability of Delta ligands to bind and activate Notch1.


Subject(s)
EGF Family of Proteins/metabolism , Fucose/analogs & derivatives , Fucose/pharmacology , Fucosyltransferases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Fucose/chemistry , Fucose/genetics , Fucosyltransferases/genetics , HEK293 Cells , Humans , Ligands , Protein Binding
9.
J Virol ; 90(23): 10499-10512, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27630236

ABSTRACT

While natural hepatitis C virus (HCV) infection results in highly diverse quasispecies of related viruses over time, mutations accumulate more slowly in tissue culture, in part because of the inefficiency of replication in cells. To create a highly diverse population of HCV particles in cell culture and identify novel growth-enhancing mutations, we engineered a library of infectious HCV with all codons represented at most positions in the ectodomain of the E2 gene. We identified many putative growth-adaptive mutations and selected nine highly represented E2 mutants for further study: Q412R, T416R, S449P, T563V, A579R, L619T, V626S, K632T, and L644I. We evaluated these mutants for changes in particle-to-infectious-unit ratio, sensitivity to neutralizing antibody or CD81 large extracellular loop (CD81-LEL) inhibition, entry factor usage, and buoyant density profiles. Q412R, T416R, S449P, T563V, and L619T were neutralized more efficiently by anti-E2 antibodies and T416R, T563V, and L619T by CD81-LEL. Remarkably, all nine variants showed reduced dependence on scavenger receptor class B type I (SR-BI) for infection. This shift from SR-BI usage did not correlate with a change in the buoyant density profiles of the variants, suggesting an altered E2-SR-BI interaction rather than changes in the virus-associated lipoprotein-E2 interaction. Our results demonstrate that residues influencing SR-BI usage are distributed across E2 and support the development of large-scale mutagenesis studies to identify viral variants with unique functional properties. IMPORTANCE: Characterizing variant viruses can reveal new information about the life cycle of HCV and the roles played by different viral genes. However, it is difficult to recapitulate high levels of diversity in the laboratory because of limitations in the HCV culture system. To overcome this limitation, we engineered a library of mutations into the E2 gene in the context of an infectious clone of the virus. We used this library of viruses to identify nine mutations that enhance the growth rate of HCV. These growth-enhancing mutations reduced the dependence on a key entry receptor, SR-BI. By generating a highly diverse library of infectious HCV, we mapped regions of the E2 protein that influence a key virus-host interaction and provide proof of principle for the generation of large-scale mutant libraries for the study of pathogens with great sequence variability.


Subject(s)
Hepacivirus/genetics , Mutation , Scavenger Receptors, Class B/physiology , Viral Envelope Proteins/genetics , Adaptation, Physiological/genetics , Amino Acid Substitution , Antibodies, Viral , CRISPR-Cas Systems , Cell Line , Gene Library , Genetic Engineering , Genome, Viral , Hepacivirus/growth & development , Hepacivirus/physiology , Hepatitis C/immunology , Hepatitis C/prevention & control , Hepatitis C/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Models, Molecular , Mutagenesis, Site-Directed , Tetraspanin 28 , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
10.
J Virol ; 87(2): 818-28, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115296

ABSTRACT

St. Louis encephalitis virus (SLEV) is a mosquito-borne flavivirus responsible for several human encephalitis outbreaks over the last 80 years. Mature flavivirus virions are coated with dimeric envelope (E) proteins that mediate attachment and fusion with host cells. E is a class II fusion protein, the hallmark of which is a distinct dimer-to-trimer rearrangement that occurs upon endosomal acidification and insertion of hydrophobic fusion peptides into the endosomal membrane. Herein, we report the crystal structure of SLEV E in the posfusion trimer conformation. The structure revealed specific features that differentiate SLEV E from trimers of related flavi- and alphaviruses. SLEV E fusion loops have distinct intermediate spacing such that they are positioned further apart than previously observed in flaviviruses but closer together than Semliki Forest virus, an alphavirus. Domains II and III (DII and DIII) of SLEV E also adopt different angles relative to DI, which suggests that the DI-DII joint may accommodate spheroidal motions. However, trimer interfaces are well conserved among flaviviruses, so it is likely the differences observed represent structural features specific to SLEV function. Analysis of surface potentials revealed a basic platform underneath flavivirus fusion loops that may interact with the anionic lipid head groups found in membranes. Taken together, these results highlight variations in E structure and assembly that may direct virus-specific interactions with host determinants to influence pathogenesis.


Subject(s)
Encephalitis Virus, St. Louis/chemistry , Viral Fusion Proteins/chemistry , Virus Internalization , Crystallography, X-Ray , Encephalitis Virus, St. Louis/physiology , Humans , Models, Molecular , Protein Multimerization , Protein Structure, Quaternary , Viral Fusion Proteins/metabolism
11.
Nat Commun ; 15(1): 465, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238313

ABSTRACT

The Notch signaling pathway has fundamental roles in embryonic development and in the nervous system. The current model of receptor activation involves initiation via a force-induced conformational change. Here, we define conditions that reveal pulling force-independent Notch activation using soluble multivalent constructs. We treat neuroepithelial stem-like cells with molecularly precise ligand nanopatterns displayed from solution using DNA origami. Notch signaling follows with clusters of Jag1, and with chimeric structures where most Jag1 proteins are replaced by other binders not targeting Notch. Our data rule out several confounding factors and suggest a model where Jag1 activates Notch upon prolonged binding without appearing to need a pulling force. These findings reveal a distinct mode of activation of Notch and lay the foundation for the development of soluble agonists.


Subject(s)
Receptors, Notch , Signal Transduction , Receptors, Notch/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Signal Transduction/physiology , Calcium-Binding Proteins/metabolism
12.
Dev Cell ; 59(9): 1110-1131.e22, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38569552

ABSTRACT

The developmental origin of blood-forming hematopoietic stem cells (HSCs) is a longstanding question. Here, our non-invasive genetic lineage tracing in mouse embryos pinpoints that artery endothelial cells generate HSCs. Arteries are transiently competent to generate HSCs for 2.5 days (∼E8.5-E11) but subsequently cease, delimiting a narrow time frame for HSC formation in vivo. Guided by the arterial origins of blood, we efficiently and rapidly differentiate human pluripotent stem cells (hPSCs) into posterior primitive streak, lateral mesoderm, artery endothelium, hemogenic endothelium, and >90% pure hematopoietic progenitors within 10 days. hPSC-derived hematopoietic progenitors generate T, B, NK, erythroid, and myeloid cells in vitro and, critically, express hallmark HSC transcription factors HLF and HOXA5-HOXA10, which were previously challenging to upregulate. We differentiated hPSCs into highly enriched HLF+ HOXA+ hematopoietic progenitors with near-stoichiometric efficiency by blocking formation of unwanted lineages at each differentiation step. hPSC-derived HLF+ HOXA+ hematopoietic progenitors could avail both basic research and cellular therapies.


Subject(s)
Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells , Pluripotent Stem Cells , Animals , Humans , Mice , Endothelial Cells/metabolism , Endothelial Cells/cytology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism
13.
J Virol ; 86(4): 2337-46, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22156523

ABSTRACT

Japanese encephalitis virus (JEV) is the leading global cause of viral encephalitis. The JEV envelope protein (E) facilitates cellular attachment and membrane fusion and is the primary target of neutralizing antibodies. We have determined the 2.1-Å resolution crystal structure of the JEV E ectodomain refolded from bacterial inclusion bodies. The E protein possesses the three domains characteristic of flavivirus envelopes and epitope mapping of neutralizing antibodies onto the structure reveals determinants that correspond to the domain I lateral ridge, fusion loop, domain III lateral ridge, and domain I-II hinge. While monomeric in solution, JEV E assembles as an antiparallel dimer in the crystal lattice organized in a highly similar fashion as seen in cryo-electron microscopy models of mature flavivirus virions. The dimer interface, however, is remarkably small and lacks many of the domain II contacts observed in other flavivirus E homodimers. In addition, uniquely conserved histidines within the JEV serocomplex suggest that pH-mediated structural transitions may be aided by lateral interactions outside the dimer interface in the icosahedral virion. Our results suggest that variation in dimer structure and stability may significantly influence the assembly, receptor interaction, and uncoating of virions.


Subject(s)
Encephalitis Virus, Japanese/chemistry , Membrane Glycoproteins/chemistry , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Cell Line , Crystallography, X-Ray , Dimerization , Encephalitis Virus, Japanese/genetics , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/virology , Epitope Mapping , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
14.
Trends Pharmacol Sci ; 44(12): 934-948, 2023 12.
Article in English | MEDLINE | ID: mdl-37891017

ABSTRACT

The Notch pathway regulates a diverse array of cell fate decisions, making it an enticing target in cancer therapy and regenerative medicine. During the early stages of Notch drug development, off-target toxicity precluded the approval of Notch inhibitors for the treatment of cancer. However, recent advances in our understanding of Notch structure and signaling have led to the development of several innovative Notch-based biotechnologies. In addition to new classes of inhibitors, pharmacological Notch activators have been shown to enhance osteogenesis and various aspects of T cell function. Furthermore, the mechanosensitive negative regulatory region (NRR) of the Notch receptor has been converted into synthetic Notch (synNotch) receptors with fully customizable signaling circuits. We review emergent Notch-based compounds, biologics, and cell therapies while highlighting the challenges and opportunities they face on the path to clinical development.


Subject(s)
Neoplasms , Receptors, Notch , Humans , Receptors, Notch/metabolism , Receptors, Notch/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction/physiology , Biotechnology
15.
Nat Commun ; 14(1): 891, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36797229

ABSTRACT

The atypical cadherins Fat and Dachsous are key regulators of cell growth and animal development. In contrast to classical cadherins, which form homophilic interactions to segregate cells, Fat and Dachsous cadherins form heterophilic interactions to induce cell polarity within tissues. Here, we determine the co-crystal structure of the human homologs Fat4 and Dachsous1 (Dchs1) to establish the molecular basis for Fat-Dachsous interactions. The binding domains of Fat4 and Dchs1 form an extended interface along extracellular cadherin (EC) domains 1-4 of each protein. Biophysical measurements indicate that Fat4-Dchs1 affinity is among the highest reported for cadherin superfamily members, which is attributed to an extensive network of salt bridges not present in structurally similar protocadherin homodimers. Furthermore, modeling suggests that unusual extracellular phosphorylation modifications directly modulate Fat-Dachsous binding by introducing charged contacts across the interface. Collectively, our analyses reveal how the molecular architecture of Fat4-Dchs1 enables them to form long-range, high-affinity interactions to maintain planar cell polarity.


Subject(s)
Cadherins , Cell Polarity , Tumor Suppressor Proteins , Humans , Cadherins/chemistry , Tumor Suppressor Proteins/chemistry , Cadherin Related Proteins/chemistry
16.
Nat Cancer ; 4(2): 222-239, 2023 02.
Article in English | MEDLINE | ID: mdl-36690875

ABSTRACT

Immunotherapy efficacy is limited in melanoma, and combinations of immunotherapies with other modalities have yielded limited improvements but also adverse events requiring cessation of treatment. In addition to ineffective patient stratification, efficacy is impaired by paucity of intratumoral immune cells (itICs); thus, effective strategies to safely increase itICs are needed. We report that dietary administration of L-fucose induces fucosylation and cell surface enrichment of the major histocompatibility complex (MHC)-II protein HLA-DRB1 in melanoma cells, triggering CD4+ T cell-mediated increases in itICs and anti-tumor immunity, enhancing immune checkpoint blockade responses. Melanoma fucosylation and fucosylated HLA-DRB1 associate with intratumoral T cell abundance and anti-programmed cell death protein 1 (PD1) responder status in patient melanoma specimens, suggesting the potential use of melanoma fucosylation as a strategy for stratifying patients for immunotherapies. Our findings demonstrate that fucosylation is a key mediator of anti-tumor immunity and, importantly, suggest that L-fucose is a powerful agent for safely increasing itICs and immunotherapy efficacy in melanoma.


Subject(s)
Fucose , Melanoma , Humans , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/metabolism , Fucose/metabolism , Melanoma/drug therapy , Immunotherapy , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology
17.
J Virol ; 85(14): 7005-19, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21543495

ABSTRACT

The E2 glycoprotein of hepatitis C virus (HCV) mediates viral attachment and entry into target hepatocytes and elicits neutralizing antibodies in infected patients. To characterize the structural and functional basis of HCV neutralization, we generated a novel panel of 78 monoclonal antibodies (MAbs) against E2 proteins from genotype 1a and 2a HCV strains. Using high-throughput focus-forming reduction or luciferase-based neutralization assays with chimeric infectious HCV containing structural proteins from both genotypes, we defined eight MAbs that significantly inhibited infection of the homologous HCV strain in cell culture. Two of these bound E2 proteins from strains representative of HCV genotypes 1 to 6, and one of these MAbs, H77.39, neutralized infection of strains from five of these genotypes. The three most potent neutralizing MAbs in our panel, H77.16, H77.39, and J6.36, inhibited infection at an early postattachment step. Receptor binding studies demonstrated that H77.39 inhibited binding of soluble E2 protein to both CD81 and SR-B1, J6.36 blocked attachment to SR-B1 and modestly reduced binding to CD81, and H77.16 blocked attachment to SR-B1 only. Using yeast surface display, we localized epitopes for the neutralizing MAbs on the E2 protein. Two of the strongly inhibitory MAbs, H77.16 and J6.36, showed markedly reduced binding when amino acids within hypervariable region 1 (HVR1) and at sites ∼100 to 200 residues away were changed, suggesting binding to a discontinuous epitope. Collectively, these studies help to define the structural and functional complexity of antibodies against HCV E2 protein with neutralizing potential.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Epitopes/metabolism , Hepacivirus/immunology , Viral Envelope Proteins/immunology , Animals , Base Sequence , CHO Cells , Cell Line , Cricetinae , Cricetulus , DNA Primers , Hepacivirus/genetics , Humans , Protein Binding , Viral Envelope Proteins/metabolism
18.
ACS Synth Biol ; 11(10): 3343-3353, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36107643

ABSTRACT

The Notch pathway converts receptor-ligand interactions at the cell surface into a transcriptional response in the receiver cell. In recent years, synthetic Notch systems (synNotch) that respond to different inputs and transduce different transcriptional responses have been engineered. One class of synNotch systems uses antibody-antigen interactions at the cell surface to induce the proteolytic cleavage cascade of the endogenous Notch autoregulatory core and the consequent release of a synNotch intracellular domain (ICD), converting surface antigen detection into a cellular response. While the activation of endogenous Notch requires ubiquitylation and subsequent endocytosis of the ligand ICD, these synNotch systems do not seem to have such a requirement because the synNotch ligands completely lack an ICD. This observation raises questions about existing models for the synNotch activation mechanism. Here, we test how different structural and biochemical factors affect the dependence of endogenous and synthetic Notch activation on ligand ICD. We compare the behavior of antibody-antigen synNotch (aa-synNotch) to that of endogenous Notch, and to a synNotch system that uses rapamycin induced dimerization of FK506 binding protein (FKBP) and FKBP rapamycin binding (FRB) domaindimerization domains (ff-synNotch), which still requires a ligand ICD. We found that differences in receptor-ligand affinity, in the identity of the transmembrane domain, or in the presence or absence of extracellular epidermal growth factor repeats cannot explain the differences in ligand ICD requirement that distinguishes aa-synNotch from endogenous Notch or ff-synNotch. We also found that unlike endogenous Notch and ff-synNotch, the aa-synNotch system does not exhibit trans-endocytosis of the receptor extracellular domain into the sender cell. These findings suggest that the aa-synNotch systems bypass the ligand ICD requirement because antigen-antibody pairs are able to promote other adhesive cell-cell interactions that provide the mechanical tension needed for ligand activation.


Subject(s)
Epidermal Growth Factor , Signal Transduction , Ligands , Tacrolimus Binding Proteins , Sirolimus , Antigens, Surface
19.
J Med Chem ; 64(16): 12109-12131, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34382808

ABSTRACT

Aberrant activation of Wnt/ß-catenin signaling is strongly associated with many diseases including cancer invasion and metastasis. Small-molecule targeting of the central signaling node of this pathway, ß-catenin, is a biologically rational approach to abolish hyperactivation of ß-catenin signaling but has been demonstrated to be a difficult task. Herein, we report a drug-like small molecule, ZW4864, that binds with ß-catenin and selectively disrupts the protein-protein interaction (PPI) between B-cell lymphoma 9 (BCL9) and ß-catenin while sparing the ß-catenin/E-cadherin PPI. ZW4864 dose-dependently suppresses ß-catenin signaling activation, downregulates oncogenic ß-catenin target genes, and abrogates invasiveness of ß-catenin-dependent cancer cells. More importantly, ZW4864 shows good pharmacokinetic properties and effectively suppresses ß-catenin target gene expression in the patient-derived xenograft mouse model. This study offers a selective chemical probe to explore ß-catenin-related biology and a drug-like small-molecule ß-catenin/BCL9 disruptor for future drug development.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Piperidines/therapeutic use , Protein Binding/drug effects , Transcription Factors/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Movement/drug effects , Drug Design , Female , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Molecular Structure , Piperidines/chemical synthesis , Piperidines/pharmacokinetics , Structure-Activity Relationship , Transcription Factors/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
20.
PLoS One ; 15(1): e0226928, 2020.
Article in English | MEDLINE | ID: mdl-31914456

ABSTRACT

Secreted R-spondin1-4 proteins (RSPO1-4) orchestrate stem cell renewal and tissue homeostasis by potentiating Wnt/ß-catenin signaling. RSPOs induce the turnover of negative Wnt regulators RNF43 and ZNRF3 through a process that requires RSPO interactions with Leucine-rich repeat-containing G-protein coupled receptors (LGRs), or through an LGR-independent mechanism that is enhanced by RSPO binding to heparin sulfate proteoglycans (HSPGs). Here, we describe the engineering of 'surrogate RSPOs' that function independently of LGRs to potentiate Wnt signaling on cell types expressing a target surface marker. These bispecific proteins were generated by fusing an RNF43- or ZNRF3-specific single chain antibody variable fragment (scFv) to the immune cytokine IL-2. Surrogate RSPOs mimic the function of natural RSPOs by crosslinking the extracellular domain (ECD) of RNF43 or ZNRF3 to the ECD of the IL-2 receptor CD25, which sequesters the complex and results in highly selective amplification of Wnt signaling on CD25+ cells. Furthermore, surrogate RSPOs were able substitute for wild type RSPO in a colon organoid growth assay when intestinal stem cells were transduced to express CD25. Our results provide proof-of-concept for a technology that may be adapted for use on a broad range of cell- or tissue-types and will open new avenues for the development of Wnt-based therapeutics for regenerative medicine.


Subject(s)
Colon/growth & development , Single-Chain Antibodies/metabolism , Thrombospondins/metabolism , Wnt Signaling Pathway , Binding Sites , Colon/metabolism , HEK293 Cells , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Organ Culture Techniques , Organ Specificity , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL