Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 1378, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33654081

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) and GITR ligand (GITRL) are members of the tumor necrosis superfamily that play a role in immune cell signaling, activation, and survival. GITR is a therapeutic target for directly activating effector CD4 and CD8 T cells, or depleting GITR-expressing regulatory T cells (Tregs), thereby promoting anti-tumor immune responses. GITR activation through its native ligand is important for understanding immune signaling, but GITR structure has not been reported. Here we present structures of human and mouse GITR receptors bound to their cognate ligands. Both species share a receptor-ligand interface and receptor-receptor interface; the unique C-terminal receptor-receptor enables higher order structures on the membrane. Human GITR-GITRL has potential to form a hexameric network of membrane complexes, while murine GITR-GITRL complex forms a linear chain due to dimeric interactions. Mutations at the receptor-receptor interface in human GITR reduce cell signaling with in vitro ligand binding assays and minimize higher order membrane structures when bound by fluorescently labeled ligand in cell imaging experiments.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/chemistry , Tumor Necrosis Factors/metabolism , Animals , Biophysical Phenomena , Cell Line , Cell Membrane/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Humans , Mice , Models, Molecular , Protein Binding , Reproducibility of Results , Tumor Necrosis Factors/chemistry
2.
Mol Cancer Ther ; 19(10): 2105-2116, 2020 10.
Article in English | MEDLINE | ID: mdl-32847983

ABSTRACT

Development of antagonistic mAbs that specifically target the immune checkpoint receptor, programmed cell death protein-1 (PD-1), is of great interest for cancer immunotherapy. Here, we report the biophysical characteristics and nonclinical antagonistic activities of sasanlimab (PF-06801591), a humanized anti-PD-1 antibody of IgG4 isotype. We show that sasanlimab binds selectively and with similar high potency to human and cynomolgus monkey PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, with no detectable Fc-dependent effector function. The binding of sasanlimab to human and cynomolgus PD-1 is associated with the formation of a stable complex, which is likely to be the main driver of this high-affinity interaction. In vitro, sasanlimab significantly augmented T-cell proliferation and cytokine production in mixed lymphocyte reaction and superantigen stimulation assays. In vivo, sasanlimab accelerated the incidence of GvHD by enhancing T-cell proliferation and cytokine secretion in a xenogeneic model of acute GvHD and halted the growth of MC-38 colon adenocarcinoma tumors in human PD-1 knock-in mice. Pharmacokinetic and toxicokinetic findings from cynomolgus monkey showed that sasanlimab was active and well-tolerated. Taken together, the data presented here support the clinical development of sasanlimab for the treatment of patients with advanced cancers as a single agent or in combination with other immunotherapies.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Humans , Immune Checkpoint Inhibitors/pharmacology , Mice
3.
J Mol Biol ; 431(15): 2869-2883, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31082439

ABSTRACT

Store-Operated Calcium Entry (SOCE) plays key roles in cell proliferation, muscle contraction, immune responses, and memory formation. The coordinated interactions of a number of proteins from the plasma and endoplasmic reticulum membranes control SOCE to replenish internal Ca2+ stores and generate intracellular Ca2+ signals. SARAF, an endoplasmic reticulum resident component of the SOCE pathway having no homology to any characterized protein, serves as an important brake on SOCE. Here, we describe the X-ray crystal structure of the SARAF luminal domain, SARAFL. This domain forms a novel 10-stranded ß-sandwich fold that includes a set of three conserved disulfide bonds, denoted the "SARAF-fold." The structure reveals a domain-swapped dimer in which the last two ß-strands (ß9 and ß10) are exchanged forming a region denoted the "SARAF luminal switch" that is essential for dimerization. Sequence comparisons reveal that the SARAF-fold is highly conserved in vertebrates and in a variety of pathologic fungi. Förster resonance energy transfer experiments using full-length SARAF validate the formation of the domain-swapped dimer in cells and demonstrate that dimerization is reversible. A designed variant lacking the SARAF luminal switch shows that the domain swapping is essential to function and indicates that the SARAF dimer accelerates SOCE inactivation.


Subject(s)
Calcium/metabolism , Intracellular Calcium-Sensing Proteins/metabolism , Membrane Proteins/metabolism , Calcium Signaling , Crystallography, X-Ray , HEK293 Cells , Humans , Intracellular Calcium-Sensing Proteins/chemistry , Membrane Proteins/chemistry , Models, Molecular , Protein Conformation, beta-Strand , Protein Domains , Protein Folding , Protein Multimerization
4.
Nat Commun ; 9(1): 4679, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30410017

ABSTRACT

4-1BB (CD137, TNFRSF9) is an inducible costimulatory receptor expressed on activated T cells. Clinical trials of two agonist antibodies, utomilumab (PF-05082566) and urelumab (BMS-663513), are ongoing in multiple cancer indications, and both antibodies demonstrate distinct activities in the clinic. To understand these differences, we solved structures of the human 4-1BB/4-1BBL complex, the 4-1BBL trimer alone, and 4-1BB bound to utomilumab or urelumab. The 4-1BB/4-1BBL complex displays a unique interaction between receptor and ligand when compared with other TNF family members. Furthermore, our ligand-only structure differs from previously published data. Utomilumab, a ligand-blocking antibody, binds 4-1BB between CRDs 3 and 4. In contrast, urelumab binds 4-1BB CRD-1, away from the ligand binding site. Finally, cell-based assays demonstrate utomilumab is a milder agonist than urelumab. Collectively, our data provide a deeper understanding of the 4-1BB signaling complex, providing a template for future development of next generation 4-1BB targeted biologics.


Subject(s)
4-1BB Ligand/chemistry , 4-1BB Ligand/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Antibodies, Monoclonal, Humanized , Binding Sites , HEK293 Cells , Humans , Jurkat Cells , Models, Molecular , Protein Domains
5.
PLoS Pathog ; 8(9): e1002916, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028316

ABSTRACT

Filoviruses, including Marburg virus (MARV) and Ebola virus (EBOV), cause fatal hemorrhagic fever in humans and non-human primates. All filoviruses encode a unique multi-functional protein termed VP35. The C-terminal double-stranded (ds)RNA-binding domain (RBD) of VP35 has been implicated in interferon antagonism and immune evasion. Crystal structures of the VP35 RBD from two ebolaviruses have previously demonstrated that the viral protein caps the ends of dsRNA. However, it is not yet understood how the expanses of dsRNA backbone, between the ends, are masked from immune surveillance during filovirus infection. Here, we report the crystal structure of MARV VP35 RBD bound to dsRNA. In the crystal structure, molecules of dsRNA stack end-to-end to form a pseudo-continuous oligonucleotide. This oligonucleotide is continuously and completely coated along its sugar-phosphate backbone by the MARV VP35 RBD. Analysis of dsRNA binding by dot-blot and isothermal titration calorimetry reveals that multiple copies of MARV VP35 RBD can indeed bind the dsRNA sugar-phosphate backbone in a cooperative manner in solution. Further, MARV VP35 RBD can also cap the ends of the dsRNA in solution, although this arrangement was not captured in crystals. Together, these studies suggest that MARV VP35 can both coat the backbone and cap the ends, and that for MARV, coating of the dsRNA backbone may be an essential mechanism by which dsRNA is masked from backbone-sensing immune surveillance molecules.


Subject(s)
Immune Evasion , Interferons/antagonists & inhibitors , Marburgvirus/chemistry , Marburgvirus/immunology , RNA, Double-Stranded/metabolism , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism , Cell Line , Crystallography, X-Ray , Ebolavirus/chemistry , Ebolavirus/genetics , Ebolavirus/immunology , Ebolavirus/metabolism , HEK293 Cells , Humans , Marburgvirus/genetics , Marburgvirus/metabolism , Models, Molecular , Protein Binding , Protein Structure, Tertiary , RNA, Double-Stranded/chemistry , RNA-Binding Proteins/metabolism
6.
Nat Struct Mol Biol ; 19(6): 633-41, 2012 May 06.
Article in English | MEDLINE | ID: mdl-22562138

ABSTRACT

The Ciona intestinalis voltage-sensing phosphatase (Ci-VSP) couples a voltage-sensing domain (VSD) to a lipid phosphatase that is similar to the tumor suppressor PTEN. How the VSD controls enzyme function has been unclear. Here, we present high-resolution crystal structures of the Ci-VSP enzymatic domain that reveal conformational changes in a crucial loop, termed the 'gating loop', that controls access to the active site by a mechanism in which residue Glu411 directly competes with substrate. Structure-based mutations that restrict gating loop conformation impair catalytic function and demonstrate that Glu411 also contributes to substrate selectivity. Structure-guided mutations further define an interaction between the gating loop and linker that connects the phosphatase to the VSD for voltage control of enzyme activity. Together, the data suggest that functional coupling between the gating loop and the linker forms the heart of the regulatory mechanism that controls voltage-dependent enzyme activation.


Subject(s)
Ciona intestinalis/enzymology , Glutamic Acid/chemistry , PTEN Phosphohydrolase/chemistry , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Amino Acid Sequence , Animals , Ciona intestinalis/chemistry , Ciona intestinalis/genetics , Crystallography, X-Ray , Enzyme Activation , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Molecular Sequence Data , Mutation , PTEN Phosphohydrolase/metabolism , Phosphoric Monoester Hydrolases/genetics , Protein Conformation , Protein Structure, Tertiary
7.
Curr Opin Virol ; 2(2): 151-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22482712

ABSTRACT

The innate immune system is one of the first lines of defense against invading pathogens. Pathogens have, in turn, evolved different strategies to counteract these responses. Recent studies have illuminated how the hemorrhagic fever viruses Ebola and Lassa fever prevent host sensing of double-stranded RNA (dsRNA), a key hallmark of viral infection. The ebolavirus protein VP35 adopts a unique bimodal configuration to mask key cellular recognition sites on dsRNA. Conversely, the Lassa fever virus nucleoprotein actually digests the dsRNA signature. Collectively, these structural and functional studies shed new light on the mechanisms of pathogenesis of these viruses and provide new targets for therapeutic intervention.


Subject(s)
Ebolavirus/immunology , Hemorrhagic Fevers, Viral/immunology , Immune Evasion , Immunity, Innate , Lassa virus/immunology , Animals , Ebolavirus/genetics , Hemorrhagic Fevers, Viral/virology , Humans , Lassa virus/genetics , Viral Proteins/genetics , Viral Proteins/immunology
8.
Proc Natl Acad Sci U S A ; 108(6): 2396-401, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262835

ABSTRACT

Lassa fever virus, a member of the family Arenaviridae, is a highly endemic category A pathogen that causes 300,000-500,000 infections per year in Western Africa. The arenaviral nucleoprotein NP has been implicated in suppression of the host innate immune system, but the mechanism by which this occurs has remained elusive. Here we present the crystal structure at 1.5 Å of the immunosuppressive C-terminal portion of Lassa virus NP and illustrate that, unexpectedly, its 3D fold closely mimics that of the DEDDh family of exonucleases. Accompanying biochemical experiments illustrate that NP indeed has a previously unknown, bona fide exonuclease activity, with strict specificity for double-stranded RNA substrates. We further demonstrate that this exonuclease activity is essential for the ability of NP to suppress translocation of IFN regulatory factor 3 and block activation of the innate immune system. Thus, the nucleoprotein is a viral exonuclease with anti-immune activity, and this work provides a unique opportunity to combat arenaviral infections.


Subject(s)
Exoribonucleases/chemistry , Lassa virus/enzymology , Nucleoproteins/chemistry , RNA, Double-Stranded/chemistry , Viral Proteins/chemistry , Cell Line , Crystallography, X-Ray , Exoribonucleases/immunology , Humans , Immunity, Innate , Interferon Regulatory Factor-3/immunology , Lassa virus/immunology , Nucleoproteins/immunology , Protein Structure, Tertiary , RNA, Double-Stranded/immunology , Viral Proteins/immunology
9.
Proc Natl Acad Sci U S A ; 107(1): 314-9, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20018665

ABSTRACT

Ebolavirus causes a severe hemorrhagic fever and is divided into five distinct species, of which Reston ebolavirus is uniquely nonpathogenic to humans. Disease caused by ebolavirus is marked by early immunosuppression of innate immune signaling events, involving silencing and sequestration of double-stranded RNA (dsRNA) by the viral protein VP35. Here we present unbound and dsRNA-bound crystal structures of the dsRNA-binding domain of Reston ebolavirus VP35. The structures show that VP35 forms an unusual, asymmetric dimer on dsRNA binding, with each of the monomers binding dsRNA in a different way: one binds the backbone whereas the other caps the terminus. Additional SAXS, DXMS, and dsRNA-binding experiments presented here support a model of cooperative dsRNA recognition in which binding of the first monomer assists binding of the next monomer of the oligomeric VP35 protein. This work illustrates how ebolavirus VP35 could mask key recognition sites of molecules such as RIG-I, MDA-5, and Dicer to silence viral dsRNA in infection.


Subject(s)
Immunity, Innate/physiology , RNA, Double-Stranded/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Humans , Macromolecular Substances , Mass Spectrometry/methods , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation , Protein Multimerization , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , Scattering, Small Angle , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/genetics
10.
J Virol ; 80(11): 5168-78, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16698997

ABSTRACT

The Ebola virus (EBOV) VP35 protein blocks the virus-induced phosphorylation and activation of interferon regulatory factor 3 (IRF-3), a transcription factor critical for the induction of alpha/beta interferon (IFN-alpha/beta) expression. However, the mechanism(s) by which this blockage occurs remains incompletely defined. We now provide evidence that VP35 possesses double-stranded RNA (dsRNA)-binding activity. Specifically, VP35 bound to poly(rI) . poly(rC)-coated Sepharose beads but not control beads. In contrast, two VP35 point mutants, R312A and K309A, were found to be greatly impaired in their dsRNA-binding activity. Competition assays showed that VP35 interacted specifically with poly(rI) . poly(rC), poly(rA) . poly(rU), or in vitro-transcribed dsRNAs derived from EBOV sequences, and not with single-stranded RNAs (ssRNAs) or double-stranded DNA. We then screened wild-type and mutant VP35s for their ability to target different components of the signaling pathways that activate IRF-3. These experiments indicate that VP35 blocks activation of IRF-3 induced by overexpression of RIG-I, a cellular helicase recently implicated in the activation of IRF-3 by either virus or dsRNA. Interestingly, the VP35 mutants impaired for dsRNA binding have a decreased but measurable IFN antagonist activity in these assays. Additionally, wild-type and dsRNA-binding-mutant VP35s were found to have equivalent abilities to inhibit activation of the IFN-beta promoter induced by overexpression of IPS-1, a recently identified signaling molecule downstream of RIG-I, or by overexpression of the IRF-3 kinases IKKepsilon and TBK-1. These data support the hypothesis that dsRNA binding may contribute to VP35 IFN antagonist function. However, additional mechanisms of inhibition, at a point proximal to the IRF-3 kinases, most likely also exist.


Subject(s)
DEAD-box RNA Helicases/physiology , Ebolavirus/chemistry , RNA, Double-Stranded/drug effects , Signal Transduction/drug effects , Viral Proteins/pharmacology , Animals , Cell Line , Chlorocebus aethiops , DEAD Box Protein 58 , Ebolavirus/metabolism , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , RNA, Double-Stranded/metabolism , Receptors, Immunologic , Signal Transduction/physiology , Vero Cells , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...