Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cell ; 151(7): 1557-68, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23260142

ABSTRACT

HLA-DR molecules bind microbial peptides in an endosomal compartment and present them on the cell surface for CD4 T cell surveillance. HLA-DM plays a critical role in the endosomal peptide selection process. The structure of the HLA-DM-HLA-DR complex shows major rearrangements of the HLA-DR peptide-binding groove. Flipping of a tryptophan away from the HLA-DR1 P1 pocket enables major conformational changes that position hydrophobic HLA-DR residues into the P1 pocket. These conformational changes accelerate peptide dissociation and stabilize the empty HLA-DR peptide-binding groove. Initially, incoming peptides have access to only part of the HLA-DR groove and need to compete with HLA-DR residues for access to the P2 site and the hydrophobic P1 pocket. This energetic barrier creates a rapid and stringent selection process for the highest-affinity binders. Insertion of peptide residues into the P2 and P1 sites reverses the conformational changes, terminating selection through DM dissociation.


Subject(s)
HLA-D Antigens/chemistry , HLA-D Antigens/metabolism , HLA-DR1 Antigen/chemistry , HLA-DR1 Antigen/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Interaction Domains and Motifs , Sequence Alignment
2.
Nat Immunol ; 14(7): 699-705, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23666294

ABSTRACT

Activating and inhibitory receptors on natural killer (NK) cells have a crucial role in innate immunity, although the basis of the engagement of activating NK cell receptors is unclear. The activating receptor Ly49H confers resistance to infection with murine cytomegalovirus by binding to the 'immunoevasin' m157. We found that m157 bound to the helical stalk of Ly49H, whereby two m157 monomers engaged the Ly49H dimer. The helical stalks of Ly49H lay centrally across the m157 platform, whereas its lectin domain was not required for recognition. Instead, m157 targeted an 'aromatic peg motif' present in stalks of both activating and inhibitory receptors of the Ly49 family, and substitution of this motif abrogated binding. Furthermore, ligation of m157 to Ly49H or Ly49C resulted in intracellular signaling. Accordingly, m157 has evolved to 'tackle the legs' of a family of NK cell receptors.


Subject(s)
Herpesviridae Infections/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Innate/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology , Amino Acid Motifs/immunology , Amino Acid Sequence , Animals , Crystallography, X-Ray , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Signal Transduction/immunology , Specific Pathogen-Free Organisms , Surface Plasmon Resonance
3.
Mol Cell ; 68(4): 659-672.e9, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149594

ABSTRACT

Certain BH3-only proteins transiently bind and activate Bak and Bax, initiating their oligomerization and the permeabilization of the mitochondrial outer membrane, a pivotal step in the mitochondrial pathway to apoptosis. Here we describe the first crystal structures of an activator BH3 peptide bound to Bak and illustrate their use in the design of BH3 derivatives capable of inhibiting human Bak on mitochondria. These BH3 derivatives compete for the activation site at the canonical groove, are the first engineered inhibitors of Bak activation, and support the role of key conformational transitions associated with Bak activation.


Subject(s)
Apoptosis/drug effects , Bcl-2-Like Protein 11 , Mitochondria , Peptides , bcl-2 Homologous Antagonist-Killer Protein , Animals , Bcl-2-Like Protein 11/chemistry , Bcl-2-Like Protein 11/pharmacology , Cell Line, Transformed , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Structure-Activity Relationship , bcl-2 Homologous Antagonist-Killer Protein/chemistry , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism
4.
Proc Natl Acad Sci U S A ; 119(48): e2208058119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36409917

ABSTRACT

The B cell and T cell antigen receptors (BCR and TCR) share a common architecture in which variable dimeric antigen-binding modules assemble with invariant dimeric signaling modules to form functional receptor complexes. In the TCR, a highly conserved T cell receptor αß (TCRαß) transmembrane (TM) interface forms a rigid structure around which its three dimeric signaling modules assemble through well-characterized polar interactions. Noting that the key features stabilizing this TCRαß TM interface also appear with high evolutionary conservation in the TM sequences of the membrane immunoglobulin (mIg) heavy chains that form the BCR's homodimeric antigen-binding module, we asked whether the BCR contained an analogous TM structure. Using an unbiased biochemical and computational modeling approach, we found that the mouse IgM BCR forms a core TM structure that is remarkably similar to that of the TCR. This structure is reinforced by a network of interhelical hydrogen bonds, and our model is nearly identical to the arrangement observed in the just-released cryo-electron microscopy (cryo-EM) structures of intact human BCRs. Our biochemical analysis shows that the integrity of this TM structure is vital for stable assembly with the BCR signaling module CD79AB in the B cell endoplasmic reticulum, and molecular dynamics simulations indicate that BCRs of all five isotypes can form comparable structures. These results demonstrate that, despite their many differences in composition, complexity, and ligand type, TCRs and BCRs rely on a common core TM structure that has been shaped by evolution for optimal receptor assembly and stability in the cell membrane.


Subject(s)
Receptors, Antigen, B-Cell , T-Lymphocytes , Humans , Mice , Animals , Receptors, Antigen, B-Cell/metabolism , T-Lymphocytes/metabolism , Cryoelectron Microscopy , Receptors, Antigen, T-Cell/metabolism , Cell Membrane/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism
5.
Immunol Cell Biol ; 101(10): 923-935, 2023.
Article in English | MEDLINE | ID: mdl-37721869

ABSTRACT

The emergence of large language models (LLMs) and assisted artificial intelligence (AI) technologies have revolutionized the way in which we interact with technology. A recent symposium at the Walter and Eliza Hall Institute explored the current practical applications of LLMs in medical research and canvassed the emerging ethical, legal and social implications for the use of AI-assisted technologies in the sciences. This paper provides an overview of the symposium's key themes and discussions delivered by diverse speakers, including early career researchers, group leaders, educators and policy-makers highlighting the opportunities and challenges that lie ahead for scientific researchers and educators as we continue to explore the potential of this cutting-edge and emerging technology.


Subject(s)
Artificial Intelligence , Biomedical Research , Technology
6.
Nat Immunol ; 12(1): 54-61, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21131964

ABSTRACT

The mechanisms of HLA-DM-catalyzed peptide exchange remain uncertain. Here we found that all stages of the interaction of HLA-DM with HLA-DR were dependent on the occupancy state of the peptide-binding groove. High-affinity peptides were protected from removal by HLA-DM through two mechanisms: peptide binding induced the dissociation of a long-lived complex of empty HLA-DR and HLA-DM, and high-affinity HLA-DR-peptide complexes bound HLA-DM only very slowly. Nonbinding covalent HLA-DR-peptide complexes were converted into efficient HLA-DM binders after truncation of an N-terminal peptide segment that emptied the P1 pocket and disrupted conserved hydrogen bonds to HLA-DR. HLA-DM thus binds only to HLA-DR conformers in which a critical part of the binding site is already vacant because of spontaneous peptide motion.


Subject(s)
HLA-D Antigens/metabolism , HLA-DR2 Antigen/metabolism , Mutant Proteins/metabolism , Peptide Fragments/metabolism , Animals , Antigen Presentation , CHO Cells , Catalysis , Cricetinae , Cricetulus , HLA-D Antigens/chemistry , HLA-D Antigens/genetics , HLA-DR2 Antigen/chemistry , HLA-DR2 Antigen/genetics , Humans , Models, Chemical , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Binding , Surface Plasmon Resonance , Transgenes/genetics
7.
J Biol Chem ; 297(1): 100900, 2021 07.
Article in English | MEDLINE | ID: mdl-34157285

ABSTRACT

Immune-stimulatory ligands, such as major histocompatibility complex molecules and the T-cell costimulatory ligand CD86, are central to productive immunity. Endogenous mammalian membrane-associated RING-CHs (MARCH) act on these and other targets to regulate antigen presentation and activation of adaptive immunity, whereas virus-encoded homologs target the same molecules to evade immune responses. Substrate specificity is encoded in or near the membrane-embedded domains of MARCHs and the proteins they regulate, but the exact sequences that distinguish substrates from nonsubstrates are poorly understood. Here, we examined the requirements for recognition of the costimulatory ligand CD86 by two different MARCH-family proteins, human MARCH1 and Kaposi's sarcoma herpesvirus modulator of immune recognition 2 (MIR2), using deep mutational scanning. We identified a highly specific recognition surface in the hydrophobic core of the CD86 transmembrane (TM) domain (TMD) that is required for recognition by MARCH1 and prominently features a proline at position 254. In contrast, MIR2 requires no specific sequences in the CD86 TMD but relies primarily on an aspartic acid at position 244 in the CD86 extracellular juxtamembrane region. Surprisingly, MIR2 recognized CD86 with a TMD composed entirely of valine, whereas many different single amino acid substitutions in the context of the native TM sequence conferred MIR2 resistance. These results show that the human and viral proteins evolved completely different recognition modes for the same substrate. That some TM sequences are incompatible with MIR2 activity, even when no specific recognition motif is required, suggests a more complicated mechanism of immune modulation via CD86 than was previously appreciated.


Subject(s)
B7-2 Antigen/chemistry , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Cell Membrane/metabolism , Down-Regulation , HEK293 Cells , HeLa Cells , Humans , Mutation , Protein Domains , Protein Transport
8.
Blood ; 135(4): 287-292, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31697803

ABSTRACT

The single transmembrane domain (TMD) of the human thrombopoietin receptor (TpoR/myeloproliferative leukemia [MPL] protein), encoded by exon 10 of the MPL gene, is a hotspot for somatic mutations associated with myeloproliferative neoplasms (MPNs). Approximately 6% and 14% of JAK2 V617F- essential thrombocythemia and primary myelofibrosis patients, respectively, have "canonical" MPL exon 10 driver mutations W515L/K/R/A or S505N, which generate constitutively active receptors and consequent loss of Tpo dependence. Other "noncanonical" MPL exon 10 mutations have also been identified in patients, both alone and in combination with canonical mutations, but, in almost all cases, their functional consequences and relevance to disease are unknown. Here, we used a deep mutational scanning approach to evaluate all possible single amino acid substitutions in the human TpoR TMD for their ability to confer cytokine-independent growth in Ba/F3 cells. We identified all currently recognized driver mutations and 7 novel mutations that cause constitutive TpoR activation, and a much larger number of second-site mutations that enhance S505N-driven activation. We found examples of both of these categories in published and previously unpublished MPL exon 10 sequencing data from MPN patients, demonstrating that some, if not all, of the new mutations reported here represent likely drivers or modifiers of myeloproliferative disease.


Subject(s)
Amino Acid Substitution , Myeloproliferative Disorders/genetics , Receptors, Thrombopoietin/genetics , Animals , Cell Line , Exons , Humans , Mice , Models, Molecular , Mutation , Protein Domains , Receptors, Thrombopoietin/chemistry
9.
J Biol Chem ; 294(7): 2470-2485, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30554144

ABSTRACT

The membrane-associated RING-CH (MARCH) family of membrane-bound E3 ubiquitin ligases regulates the levels of cell-surface membrane proteins, many of which are involved in immune responses. Although their role in ubiquitin-dependent endocytosis and degradation of cell-surface proteins is extensively documented, the features of MARCH proteins and their substrates that drive the molecular recognition events leading to ubiquitin transfer remain poorly defined. In this study, we sought to determine the features of human MARCH9 that are required for regulating the surface levels of its substrate proteins. Consistent with previous studies of other MARCH proteins, we found that susceptibility to MARCH9 activity is encoded in the transmembrane (TM) domains of its substrates. Accordingly, substitutions at specific residues and motifs within MARCH9's TM domains resulted in varying degrees of functional impairment. Most notably, a single serine-to-alanine substitution in the first of its two TM domains rendered MARCH9 completely unable to alter the surface levels of two different substrates: the major histocompatibility class I molecule HLA-A2 and the T-cell co-receptor CD4. Solution NMR analysis of a MARCH9 fragment encompassing the two TM domains and extracellular connecting loop revealed that the residues contributing most to MARCH9 activity are located in the α-helical portions of TM1 and TM2 that are closest to the extracellular face of the lipid bilayer. This observation defines a key region required for substrate regulation. In summary, our biochemical and structural findings demonstrate that specific sequences in the α-helical MARCH9 TM domains make crucial contributions to its ability to down-regulate its protein substrates.


Subject(s)
Down-Regulation , Gene Expression Regulation, Enzymologic , Membrane Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , CD4 Antigens/chemistry , CD4 Antigens/genetics , CD4 Antigens/metabolism , HEK293 Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Domains , Protein Structure, Secondary , Serine/chemistry , Serine/genetics , Serine/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
10.
Proc Natl Acad Sci U S A ; 114(28): E5645-E5654, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28652325

ABSTRACT

Many activating immunoreceptors associate with signaling adaptor molecules like FcεR1γ or CD247. FcεR1γ and CD247 share high sequence homology and form disulphide-linked homodimers that contain a pair of acidic aspartic acid residues in their transmembrane (TM) domains that mediate assembly, via interaction with an arginine residue at a similar register to these aspartic acids, with the activating immunoreceptors. However, this model cannot hold true for receptors like CD16A, whose TM domains do not contain basic residues. We have carried out an extensive site-directed mutagenesis analysis of the CD16A receptor complex and now report that the association of receptor with the signaling adaptor depends on a network of polar and aromatic residues along the length of the TM domain. Molecular modeling indicates that CD16A TM residues F202, D205, and T206 form the core of the membrane-embedded trimeric interface by establishing highly favorable contacts to the signaling modules through rearrangement of a hydrogen bond network previously identified in the CD247 TM dimer solution NMR structure. Strikingly, the amino acid D205 also regulates the turnover and surface expression of CD16A in the absence of FcεR1γ or CD247. Modeling studies indicate that similar features underlie the association of other activating immune receptors, including CD64 and FcεR1α, with signaling adaptor molecules, and we confirm experimentally that equivalent F, D, and T residues in the TM domain of FcεR1α markedly influence the biology of this receptor and its association with FcεR1γ.


Subject(s)
CD3 Complex/metabolism , Cell Membrane/metabolism , Receptors, IgG/metabolism , Amino Acid Motifs , Animals , Cell Line , GPI-Linked Proteins/metabolism , Glycosylation , HEK293 Cells , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Mice , Mutagenesis, Site-Directed , Protein Domains , Protein Multimerization , Receptors, IgE/metabolism , Receptors, Immunologic/metabolism , Signal Transduction
11.
Int J Mol Sci ; 21(19)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33050044

ABSTRACT

The impressive success of chimeric antigen receptor (CAR)-T cell therapies in treating advanced B-cell malignancies has spurred a frenzy of activity aimed at developing CAR-T therapies for other cancers, particularly solid tumors, and optimizing engineered T cells for maximum clinical benefit in many different disease contexts. A rapidly growing body of design work is examining every modular component of traditional single-chain CARs as well as expanding out into many new and innovative engineered immunoreceptor designs that depart from this template. New approaches to immune cell and receptor engineering are being reported with rapidly increasing frequency, and many recent high-quality reviews (including one in this special issue) provide comprehensive coverage of the history and current state of the art in CAR-T and related cellular immunotherapies. In this review, we step back to examine our current understanding of the structure-function relationships in natural and engineered lymphocyte-activating receptors, with an eye towards evaluating how well the current-generation CAR designs recapitulate the most desirable features of their natural counterparts. We identify key areas that we believe are under-studied and therefore represent opportunities to further improve our grasp of form and function in natural and engineered receptors and to rationally design better therapeutics.


Subject(s)
Cell Engineering/methods , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Cell Communication/immunology , Humans , Neoplasms/therapy , Protein Domains , Receptors, Chimeric Antigen/chemistry
12.
Langmuir ; 35(25): 8344-8356, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31122018

ABSTRACT

For evolving biological and biomedical applications of hybrid protein?lipid materials, understanding the behavior of the protein within the lipid mesophase is crucial. After more than two decades since the invention of the in meso crystallization method, a protein-eye view of its mechanism is still lacking. Numerous structural studies have suggested that integral membrane proteins preferentially partition at localized flat points on the bilayer surface of the cubic phase with crystal growth occurring from a local fluid lamellar L? phase conduit. However, studies to date have, by necessity, focused on structural transitions occurring in the lipid mesophase. Here, we demonstrate using small-angle neutron scattering that the lipid bilayer of monoolein (the most commonly used lipid for in meso crystallization) can be contrast-matched using deuteration, allowing us to isolate scattering from encapsulated peptides during the crystal growth process for the first time. During in meso crystallization, a clear decrease in form factor scattering intensity of the peptides was observed and directly correlated with crystal growth. A transient fluid lamellar L? phase was observed, providing direct evidence for the proposed mechanism for this technique. This suggests that the peptide passes through a transition from the cubic QII phase, via an L? phase to the lamellar crystalline Lc phase with similar layered spacing. When high protein loading was possible, the lamellar crystalline Lc phase of the peptide in the single crystals was observed. These findings show the mechanism of in meso crystallization for the first time from the perspective of integral membrane proteins.


Subject(s)
Crystallization/methods , Lipid Bilayers/chemistry , Glycerides/chemistry , X-Ray Diffraction
13.
Proc Natl Acad Sci U S A ; 113(43): E6649-E6658, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27791034

ABSTRACT

The T-cell antigen receptor (TCR) is an assembly of eight type I single-pass membrane proteins that occupies a central position in adaptive immunity. Many TCR-triggering models invoke an alteration in receptor complex structure as the initiating event, but both the precise subunit organization and the pathway by which ligand-induced alterations are transferred to the cytoplasmic signaling domains are unknown. Here, we show that the receptor complex transmembrane (TM) domains form an intimately associated eight-helix bundle organized by a specific interhelical TCR TM interface. The salient features of this core structure are absolutely conserved between αß and γδ TCR sequences and throughout vertebrate evolution, and mutations at key interface residues caused defects in the formation of stable TCRαß:CD3δε:CD3γε:ζζ complexes. These findings demonstrate that the eight TCR-CD3 subunits form a compact and precisely organized structure within the membrane and provide a structural basis for further investigation of conformationally regulated models of transbilayer TCR signaling.


Subject(s)
CD3 Complex/chemistry , Cell Membrane/ultrastructure , Protein Subunits/chemistry , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, gamma-delta/chemistry , Amino Acid Sequence , Binding Sites , CD3 Complex/genetics , CD3 Complex/immunology , Cell Membrane/chemistry , Cell Membrane/immunology , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Subunits/genetics , Protein Subunits/immunology , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , T-Lymphocytes/chemistry , T-Lymphocytes/immunology
14.
Biophys J ; 114(5): 1030-1035, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29395047

ABSTRACT

T cell receptors (TCRs) are octameric assemblies of type-I membrane proteins in which a receptor heterodimer (αß, δγ, or pre-Tαß) is associated with three dimeric signaling modules (CD3δε, CD3γε, and ζζ) at the T cell or pre-T cell surface. In the human αßTCR, the α and ß transmembrane (TM) domains form a specific structure that acts as a hub for assembly with the signaling modules inside the lipid bilayer. Conservation of key polar contacts across the C-terminal half of this TM interface suggests that the structure is a common feature of all TCR types. In this study, using molecular dynamics simulations in explicit lipid bilayers, we show that human δγ and pre-Tαß TM domains also adopt stable αß-like interfaces, yet each displays unique features that modulate the stability of the interaction and are related to sequences that are conserved within TCR types, but are distinct from the αß sequences. We also performed simulations probing effects of previously reported mutations in the human αß TM interface, and observed that the most disruptive mutations caused substantial departures from the wild-type TM structure and increased dynamics. These simulations show a strong correlation between structural instability, increased conformational variation, and the severity of structural defects in whole-TCR complexes measured in our previous biochemical assays. These results thus support the view that the stability of the core TM structure is a key determinant of TCR structural integrity and suggest that the interface has been evolutionarily optimized for different forms of TCRs.


Subject(s)
Cell Membrane/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Amino Acid Sequence , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Protein Multimerization , Protein Structure, Quaternary
15.
J Biol Chem ; 290(42): 25307-21, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26324715

ABSTRACT

Plasmodium falciparum parasites must invade red blood cells to survive within humans. Entry into red blood cells is governed by interactions between parasite adhesins and red blood cell receptors. Previously we identified that P. falciparum reticulocyte binding protein-like homologue 4 (PfRh4) binds to complement receptor 1 (CR1) to mediate entry of malaria parasites into human red blood cells. In this report we characterize a collection of anti-PfRh4 monoclonal antibodies and CR1 protein fragments that modulate the interaction between PfRh4 and CR1. We identify an anti-PfRh4 monoclonal that blocks PfRh4-CR1 interaction in vitro, inhibits PfRh4 binding to red blood cells, and as a result abolishes the PfRh4-CR1 invasion pathway in P. falciparum. Epitope mapping of anti-PfRh4 monoclonal antibodies identified distinct functional regions within PfRh4 involved in modulating its interaction with CR1. Furthermore, we designed a set of protein fragments based on extensive mutagenesis analyses of the PfRh4 binding site on CR1 and determined their interaction affinities using surface plasmon resonance. These CR1 protein fragments bind tightly to PfRh4 and also function as soluble inhibitors to block PfRh4 binding to red blood cells and to inhibit the PfRh4-CR1 invasion pathway. Our findings can aid future efforts in designing specific single epitope antibodies to block P. falciparum invasion via complement receptor 1.


Subject(s)
Antibodies, Monoclonal/immunology , Erythrocytes/metabolism , Membrane Proteins/metabolism , Protozoan Proteins/metabolism , Receptors, Complement/metabolism , Animals , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/immunology
16.
Immunol Cell Biol ; 99(7): 674-676, 2021 08.
Article in English | MEDLINE | ID: mdl-34180552
17.
J Biol Chem ; 289(12): 8240-51, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24488493

ABSTRACT

In mammals, the αßT cell receptor (TCR) signaling complex is composed of a TCRαß heterodimer that is noncovalently coupled to three dimeric signaling molecules, CD3εδ, CD3εγ, and CD3ζζ. The nature of the TCR signaling complex and subunit arrangement in different species remains unclear however. Here we present a structural and biochemical analysis of the more primitive ancestral form of the TCR signaling complex found in chickens. In contrast to mammals, chickens do not express separate CD3δ and CD3γ chains but instead encode a single hybrid chain, termed CD3δ/γ, that is capable of pairing with CD3ε. The NMR structure of the chicken CD3εδ/γ heterodimer revealed a unique dimer interface that results in a heterodimer with considerable deviation from the distinct side-by-side architecture found in human and murine CD3εδ and CD3εγ. The chicken CD3εδ/γ heterodimer also contains a unique molecular surface, with the vast majority of surface-exposed, nonconserved residues being clustered to a single face of the heterodimer. Using an in vitro biochemical assay, we demonstrate that CD3εδ/γ can assemble with both chicken TCRα and TCRß via conserved polar transmembrane sites. Moreover, analogous to the human TCR signaling complex, the presence of two copies of CD3εδ/γ is required for ζζ assembly. These data provide insight into the evolution of this critical receptor signaling apparatus.


Subject(s)
Avian Proteins/chemistry , CD3 Complex/chemistry , Chickens/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Amino Acid Sequence , Animals , CD3 Complex/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Multimerization , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Sequence Alignment
18.
J Am Chem Soc ; 137(50): 15676-9, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26642914

ABSTRACT

The mechanisms of assembly and function for many important type I/II (single-pass) transmembrane (TM) receptors are proposed to involve the formation and/or alteration of specific interfaces among their membrane-embedded α-helical TM domains. The application of lipidic cubic phase (LCP) bilayer media for crystallization of single-α-helical TM complexes has the potential to provide valuable structural and mechanistic insights into many such systems. However, the fidelity of the interfaces observed in crowded crystalline arrays has been difficult to establish from the very limited number of such structures determined using X-ray diffraction data. Here we examine this issue using the glycophorin A (GpA) model system, whose homodimeric TM helix interface has been characterized by solution and solid-state NMR and biochemical techniques but never crystallographically. We report that a GpA-TM peptide readily crystallized in a monoolein cubic phase bilayer, yielding a dimeric α-helical structure that is in excellent agreement with previously reported NMR measurements made in several different types of host media. These results provide compelling support for the wider application of LCP techniques to enable X-ray crystallographic analysis of single-pass TM interactions.


Subject(s)
Glycophorins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Lipid Bilayers , Molecular Sequence Data , Protein Conformation
19.
Clin Transl Immunology ; 12(3): e1440, 2023.
Article in English | MEDLINE | ID: mdl-36890859

ABSTRACT

Objectives: Glioblastoma is a highly aggressive and fatal brain malignancy, and effective targeted therapies are required. The combination of standard treatments including surgery, chemotherapy and radiotherapy is not curative. Chimeric antigen receptor (CAR) T cells are known to cross the blood-brain barrier, mediating antitumor responses. A tumor-expressed deletion mutant of the epidermal growth factor receptor (EGFRvIII) is a robust CAR T cell target in glioblastoma. Here, we show our de novo generated, high-affinity EGFRvIII-specific CAR; GCT02, demonstrating curative efficacy in human orthotopic glioblastoma models. Methods: The GCT02 binding epitope was predicted using Deep Mutational Scanning (DMS). GCT02 CAR T cell cytotoxicity was investigated in three glioblastoma models in vitro using the IncuCyte platform, and cytokine secretion with a cytometric bead array. GCT02 in vivo functionality was demonstrated in two NSG orthotopic glioblastoma models. The specificity profile was generated by measuring T cell degranulation in response to coculture with primary human healthy cells. Results: The GCT02 binding location was predicted to be located at a shared region of EGFR and EGFRvIII; however, the in vitro functionality remained exquisitely EGFRvIII specific. A single CAR T cell infusion generated curative responses in two orthotopic models of human glioblastoma in NSG mice. The safety analysis further validated the specificity of GCT02 for mutant-expressing cells. Conclusion: This study demonstrates the preclinical functionality of a highly specific CAR targeting EGFRvIII on human cells. This CAR could be an effective treatment for glioblastoma and warrants future clinical investigation.

20.
Elife ; 112022 05 04.
Article in English | MEDLINE | ID: mdl-35506657

ABSTRACT

De novo-designed receptor transmembrane domains (TMDs) present opportunities for precise control of cellular receptor functions. We developed a de novo design strategy for generating programmed membrane proteins (proMPs): single-pass α-helical TMDs that self-assemble through computationally defined and crystallographically validated interfaces. We used these proMPs to program specific oligomeric interactions into a chimeric antigen receptor (CAR) that we expressed in mouse primary T cells and found that both in vitro CAR T cell cytokine release and in vivo antitumor activity scaled linearly with the oligomeric state encoded by the receptor TMD, from monomers up to tetramers. All programmed CARs stimulated substantially lower T cell cytokine release relative to the commonly used CD28 TMD, which we show elevated cytokine release through lateral recruitment of the endogenous T cell costimulatory receptor CD28. Precise design using orthogonal and modular TMDs thus provides a new way to program receptor structure and predictably tune activity for basic or applied synthetic biology.


Subject(s)
CD28 Antigens , Receptors, Chimeric Antigen , Animals , CD28 Antigens/metabolism , Cytokines/metabolism , Mice , Protein Domains , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL