Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Pharmacol Res ; 170: 105735, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34146695

RESUMO

Therapeutic mAbs have dominated the class of immunotherapeutics in general and immune checkpoint inhibitors in particular. The high specificity of mAbs to the target molecule as well as their extended half-life and (or) the effector functions raised by the Fc part are some of the important aspects that contribute to the success of this class of therapeutics. Equally potential candidates are decoys and their fusions that can address some of the inherent limitations of mAbs, like immunogenicity, resistance development, low bio-availability and so on, besides maintaining the advantages of mAbs. The decoys are molecules that trap the ligands and prevent them from interacting with the signaling receptors. Although a few FDA-approved decoy immune modulators are very successful, the potential of this class of drugs is yet to be fully realized. Here, we review various strategies employed in fusion protein therapeutics with a focus on the design of decoy immunomodulators from the structural perspective and discuss how the information on protein structure and function can strategically guide the development of next-generation immune modulators.


Assuntos
Anticorpos Monoclonais/química , Desenho de Fármacos , Imunoconjugados/química , Agentes de Imunomodulação/química , Receptores Imunológicos/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Estabilidade de Medicamentos , Meia-Vida , Humanos , Imunoconjugados/imunologia , Imunoconjugados/farmacocinética , Agentes de Imunomodulação/imunologia , Agentes de Imunomodulação/farmacocinética , Ligantes , Estrutura Molecular , Estabilidade Proteica , Receptores Imunológicos/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
2.
Proc Natl Acad Sci U S A ; 114(21): E4223-E4232, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28484017

RESUMO

Rational modulation of the immune response with biologics represents one of the most promising and active areas for the realization of new therapeutic strategies. In particular, the use of function blocking monoclonal antibodies targeting checkpoint inhibitors such as CTLA-4 and PD-1 have proven to be highly effective for the systemic activation of the human immune system to treat a wide range of cancers. Ipilimumab is a fully human antibody targeting CTLA-4 that received FDA approval for the treatment of metastatic melanoma in 2011. Ipilimumab is the first-in-class immunotherapeutic for blockade of CTLA-4 and significantly benefits overall survival of patients with metastatic melanoma. Understanding the chemical and physical determinants recognized by these mAbs provides direct insight into the mechanisms of pathway blockade, the organization of the antigen-antibody complexes at the cell surface, and opportunities to further engineer affinity and selectivity. Here, we report the 3.0 Å resolution X-ray crystal structure of the complex formed by ipilimumab with its human CTLA-4 target. This structure reveals that ipilimumab contacts the front ß-sheet of CTLA-4 and intersects with the CTLA-4:Β7 recognition surface, indicating that direct steric overlap between ipilimumab and the B7 ligands is a major mechanistic contributor to ipilimumab function. The crystallographically observed binding interface was confirmed by a comprehensive cell-based binding assay against a library of CTLA-4 mutants and by direct biochemical approaches. This structure also highlights determinants responsible for the selectivity exhibited by ipilimumab toward CTLA-4 relative to the homologous and functionally related CD28.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Antineoplásicos Imunológicos/farmacologia , Sítios de Ligação de Anticorpos/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Ipilimumab/farmacologia , Melanoma/tratamento farmacológico , Fatores Biológicos/farmacologia , Antígeno CTLA-4/imunologia , Linhagem Celular , Cristalografia por Raios X , Células HEK293 , Humanos , Imunoterapia/métodos , Ligação Proteica , Estrutura Terciária de Proteína
3.
Biochemistry ; 57(8): 1293-1305, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29261291

RESUMO

Disc large 1 (Dlg1) proteins, members of the MAGUK protein family, are linked to cell polarity via their participation in multiprotein assemblies. At their N-termini, Dlg1 proteins contain a L27 domain. Typically, the L27 domains participate in the formation of obligate hetero-oligomers with the L27 domains from their cognate partners. Among the MAGUKs, Dlg1 proteins exist as homo-oligomers, and the oligomerization is solely dependent on the L27 domain. Here we provide biochemical and structural evidence of homodimerization via the L27 domain of Dlg1 from Drosophila melanogaster. The structure reveals that the core of the dimer is formed by a distinctive six-helix assembly, involving all three conserved helices from each subunit (monomer). The homodimer interface is extended by the C-terminal tail of the L27 domain of Dlg1, which forms a two-stranded antiparallel ß-sheet. The structure reconciles and provides a structural context for a large body of available mutational data. From our analyses, we conclude that the observed L27 homodimerization is most likely a feature unique to the Dlg1 orthologs within the MAGUK family.


Assuntos
Proteínas de Drosophila/química , Drosophila melanogaster/química , Proteínas Supressoras de Tumor/química , Sequência de Aminoácidos , Animais , Polaridade Celular , Drosophila melanogaster/citologia , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios Proteicos , Multimerização Proteica
4.
Biochemistry ; 57(26): 3676-3689, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29767960

RESUMO

Studying the evolution of catalytically promiscuous enzymes like those from the N-succinylamino acid racemase/ o-succinylbenzoate synthase (NSAR/OSBS) subfamily can reveal mechanisms by which new functions evolve. Some enzymes in this subfamily have only OSBS activity, while others catalyze OSBS and NSAR reactions. We characterized several NSAR/OSBS subfamily enzymes as a step toward determining the structural basis for evolving NSAR activity. Three enzymes were promiscuous, like most other characterized NSAR/OSBS subfamily enzymes. However, Alicyclobacillus acidocaldarius OSBS (AaOSBS) efficiently catalyzes OSBS activity but lacks detectable NSAR activity. Competitive inhibition and molecular modeling show that AaOSBS binds N-succinylphenylglycine with moderate affinity in a site that overlaps its normal substrate. On the basis of possible steric conflicts identified by molecular modeling and sequence conservation within the NSAR/OSBS subfamily, we identified one mutation, Y299I, that increased NSAR activity from undetectable to 1.2 × 102 M-1 s-1 without affecting OSBS activity. This mutation does not appear to affect binding affinity but instead affects kcat, by reorienting the substrate or modifying conformational changes to allow both catalytic lysines to access the proton that is moved during the reaction. This is the first site known to affect reaction specificity in the NSAR/OSBS subfamily. However, this gain of activity was obliterated by a second mutation, M18F. Epistatic interference by M18F was unexpected because a phenylalanine at this position is important in another NSAR/OSBS enzyme. Together, modest NSAR activity of Y299I AaOSBS and epistasis between sites 18 and 299 indicate that additional sites influenced the evolution of NSAR reaction specificity in the NSAR/OSBS subfamily.


Assuntos
Alicyclobacillus/enzimologia , Isomerases de Aminoácido/metabolismo , Carbono-Carbono Liases/metabolismo , Alicyclobacillus/química , Alicyclobacillus/genética , Alicyclobacillus/metabolismo , Isomerases de Aminoácido/química , Isomerases de Aminoácido/genética , Carbono-Carbono Liases/química , Carbono-Carbono Liases/genética , Domínio Catalítico , Cristalografia por Raios X , Evolução Molecular , Modelos Moleculares , Filogenia , Conformação Proteica , Especificidade por Substrato
5.
PLoS Pathog ; 12(9): e1005849, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27583447

RESUMO

The pathogenic yeast Cryptococcus neoformans causes cryptococcosis, a life-threatening fungal disease. C. neoformans has multiple virulence mechanisms that are non-host specific, induce damage and interfere with immune clearance. Microarray analysis of C. neoformans strains serially passaged in mice associated a small gene (CNAG_02591) with virulence. This gene, hereafter identified as HVA1 (hypervirulence-associated protein 1), encodes a protein that has homologs of unknown function in plant and animal fungi, consistent with a conserved mechanism. Expression of HVA1 was negatively correlated with virulence and was reduced in vitro and in vivo in both mouse- and Galleria-passaged strains of C. neoformans. Phenotypic analysis in hva1Δ and hva1Δ+HVA1 strains revealed no significant differences in established virulence factors. Mice infected intravenously with the hva1Δ strain had higher fungal burden in the spleen and brain, but lower fungal burden in the lungs, and died faster than mice infected with H99W or the hva1Δ+HVA1 strain. Metabolomics analysis demonstrated a general increase in all amino acids measured in the disrupted strain and a block in the TCA cycle at isocitrate dehydrogenase, possibly due to alterations in the nicotinamide cofactor pool. Macrophage fungal burden experiments recapitulated the mouse hypervirulent phenotype of the hva1Δ strain only in the presence of exogenous NADPH. The crystal structure of the Hva1 protein was solved, and a comparison of structurally similar proteins correlated with the metabolomics data and potential interactions with NADPH. We report a new gene that modulates virulence through a mechanism associated with changes in fungal metabolism.


Assuntos
Criptococose/microbiologia , Cryptococcus neoformans/genética , Cryptococcus neoformans/patogenicidade , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Animais , Encéfalo/patologia , Cryptococcus neoformans/metabolismo , Modelos Animais de Doenças , Metabolismo Energético , Feminino , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Humanos , Pulmão/microbiologia , Macrófagos/microbiologia , Metabolômica , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Análise de Sequência com Séries de Oligonucleotídeos , Deleção de Sequência , Virulência , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
6.
Proc Natl Acad Sci U S A ; 109(37): 14836-40, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22927415

RESUMO

Nectins are members of the Ig superfamily that mediate cell-cell adhesion through homophilic and heterophilic interactions. We have determined the crystal structure of the nectin-2 homodimer at 1.3 Å resolution. Structural analysis and complementary mutagenesis studies reveal the basis for recognition and selectivity among the nectin family members. Notably, the close proximity of charged residues at the dimer interface is a major determinant of the binding affinities associated with homophilic and heterophilic interactions within the nectin family. Our structural and biochemical data provide a mechanistic basis to explain stronger heterophilic versus weaker homophilic interactions among these family members and also offer insights into nectin-mediated transinteractions between engaging cells.


Assuntos
Moléculas de Adesão Celular/química , Adesão Celular/fisiologia , Modelos Moleculares , Sequência de Bases , Moléculas de Adesão Celular/genética , Cristalografia por Raios X , Dimerização , Humanos , Dados de Sequência Molecular , Mutagênese , Nectinas , Ligação Proteica , Mapas de Interação de Proteínas , Análise de Sequência de DNA
7.
Proc Natl Acad Sci U S A ; 108(33): 13682-7, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21825122

RESUMO

Peptide-MHC (pMHC) multimers, in addition to being tools for tracking and quantifying antigen-specific T cells, can mediate downstream signaling after T-cell receptor engagement. In the absence of costimulation, this can lead to anergy or apoptosis of cognate T cells, a property that could be exploited in the setting of autoimmune disease. Most studies with class I pMHC multimers used noncovalently linked peptides, which can allow unwanted CD8(+) T-cell activation as a result of peptide transfer to cellular MHC molecules. To circumvent this problem, and given the role of self-reactive CD8(+) T cells in the development of type 1 diabetes, we designed a single-chain pMHC complex (scK(d).IGRP) by using the class I MHC molecule H-2K(d) and a covalently linked peptide derived from islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP(206-214)), a well established autoantigen in NOD mice. X-ray diffraction studies revealed that the peptide is presented in the groove of the MHC molecule in canonical fashion, and it was also demonstrated that scK(d).IGRP tetramers bound specifically to cognate CD8(+) T cells. Tetramer binding induced death of naive T cells and in vitro- and in vivo-differentiated cytotoxic T lymphocytes, and tetramer-treated cytotoxic T lymphocytes showed a diminished IFN-γ response to antigen stimulation. Tetramer accessibility to disease-relevant T cells in vivo was also demonstrated. Our study suggests the potential of single-chain pMHC tetramers as possible therapeutic agents in autoimmune disease. Their ability to affect the fate of naive and activated CD8(+) T cells makes them a potential intervention strategy in early and late stages of disease.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Linfócitos T CD8-Positivos/efeitos dos fármacos , Antígenos de Histocompatibilidade/farmacologia , Fragmentos de Peptídeos/farmacologia , Animais , Autoantígenos , Linfócitos T CD8-Positivos/imunologia , Morte Celular/efeitos dos fármacos , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/imunologia , Glucose-6-Fosfatase/imunologia , Antígenos de Histocompatibilidade/química , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Multimerização Proteica
8.
Mol Inform ; : e202300160, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38973776

RESUMO

The insulin superfamily proteins (ISPs), in particular, insulin, IGFs and relaxin proteins are key modulators of animal physiology. They are known to have evolved from the same ancestral gene and have diverged into proteins with varied sequences and distinct functions, but maintain a similar structural architecture stabilized by highly conserved disulphide bridges. The recent surge of sequence data and the structures of these proteins prompted a need for a comprehensive analysis, which connects the evolution of these sequences (427 sequences) in the light of available functional and structural information including representative complex structures of ISPs with their cognate receptors. This study reveals (a) unusually high sequence conservation of IGFs (>90 % conservation in 184 sequences) and provides a possible structure-based rationale for such high sequence conservation; (b) provides an updated definition of the receptor-binding signature motif of the functionally diverse relaxin family members (c) provides a probable non-canonical C-peptide cleavage site in a few insulin sequences. The high conservation of IGFs appears to represent a classic case of resistance to sequence diversity exerted by physiologically important interactions with multiple partners. We also propose a probable mechanism for C-peptide cleavage in a few distinct insulin sequences and redefine the receptor-binding signature motif of the relaxin family. Lastly, we provide a basis for minimally modified insulin mutants with potential therapeutic application, inspired by concomitant changes observed in other insulin superfamily protein members supported by molecular dynamics simulation.

9.
BMC Struct Biol ; 13: 31, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24252706

RESUMO

BACKGROUND: S100A4, a member of the S100 family of Ca2+-binding proteins, modulates the motility of both non-transformed and cancer cells by regulating the localization and stability of cellular protrusions. Biochemical studies have demonstrated that S100A4 binds to the C-terminal end of the myosin-IIA heavy chain coiled-coil and disassembles myosin-IIA filaments; however, the mechanism by which S100A4 mediates myosin-IIA depolymerization is not well understood. RESULTS: We determined the X-ray crystal structure of the S100A4Δ8C/MIIA(1908-1923) peptide complex, which showed an asymmetric binding mode for the myosin-IIA peptide across the S100A4 dimer interface. This asymmetric binding mode was confirmed in NMR studies using a spin-labeled myosin-IIA peptide. In addition, our NMR data indicate that S100A4Δ8C binds the MIIA(1908-1923) peptide in an orientation very similar to that observed for wild-type S100A4. Studies of complex formation using a longer, dimeric myosin-IIA construct demonstrated that S100A4 binding dissociates the two myosin-IIA polypeptide chains to form a complex composed of one S100A4 dimer and a single myosin-IIA polypeptide chain. This interaction is mediated, in part, by the instability of the region of the myosin-IIA coiled-coil encompassing the S100A4 binding site. CONCLUSION: The structure of the S100A4/MIIA(1908-1923) peptide complex has revealed the overall architecture of this assembly and the detailed atomic interactions that mediate S100A4 binding to the myosin-IIA heavy chain. These structural studies support the idea that residues 1908-1923 of the myosin-IIA chain heavy represent a core sequence for the S100A4/myosin-IIA complex. In addition, biophysical studies suggest that structural fluctuations within the myosin-IIA coiled-coil may facilitate S100A4 docking onto a single myosin-IIA polypeptide chain.


Assuntos
Miosina não Muscular Tipo IIA/química , Miosina não Muscular Tipo IIA/metabolismo , Proteínas S100/química , Proteínas S100/metabolismo , Sítios de Ligação , Dicroísmo Circular , Cristalografia por Raios X , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Mutação , Miosinas/metabolismo , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Proteína A4 de Ligação a Cálcio da Família S100
10.
Proc Natl Acad Sci U S A ; 107(19): 8605-10, 2010 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-20421509

RESUMO

S100A4, a member of the S100 family of Ca(2+)-binding proteins, regulates carcinoma cell motility via interactions with myosin-IIA. Numerous studies indicate that S100A4 is not simply a marker for metastatic disease, but rather has a direct role in metastatic progression. These observations suggest that S100A4 is an excellent target for therapeutic intervention. Using a unique biosensor-based assay, trifluoperazine (TFP) was identified as an inhibitor that disrupts the S100A4/myosin-IIA interaction. To examine the interaction of S100A4 with TFP, we determined the 2.3 A crystal structure of human Ca(2+)-S100A4 bound to TFP. Two TFP molecules bind within the hydrophobic target binding pocket of Ca(2+)-S100A4 with no significant conformational changes observed in the protein upon complex formation. NMR chemical shift perturbations are consistent with the crystal structure and demonstrate that TFP binds to the target binding cleft of S100A4 in solution. Remarkably, TFP binding results in the assembly of five Ca(2+)-S100A4/TFP dimers into a tightly packed pentameric ring. Within each pentamer most of the contacts between S100A4 dimers occurs through the TFP moieties. The Ca(2+)-S100A4/prochlorperazine (PCP) complex exhibits a similar pentameric assembly. Equilibrium sedimentation and cross-linking studies demonstrate the cooperative formation of a similarly sized S100A4/TFP oligomer in solution. Assays examining the ability of TFP to block S100A4-mediated disassembly of myosin-IIA filaments demonstrate that significant inhibition of S100A4 function occurs only at TFP concentrations that promote S100A4 oligomerization. Together these studies support a unique mode of inhibition in which phenothiazines disrupt the S100A4/myosin-IIA interaction by sequestering S100A4 via small molecule-induced oligomerization.


Assuntos
Proclorperazina/farmacologia , Multimerização Proteica/efeitos dos fármacos , Proteínas S100/antagonistas & inibidores , Proteínas S100/química , Trifluoperazina/farmacologia , Cálcio/química , Cálcio/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Miosina não Muscular Tipo IIA/metabolismo , Proclorperazina/química , Proclorperazina/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Trifluoperazina/química , Trifluoperazina/metabolismo
11.
Immunol Rev ; 229(1): 356-86, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19426233

RESUMO

SUMMARY: Costimulatory receptors and ligands trigger the signaling pathways that are responsible for modulating the strength, course, and duration of an immune response. High-resolution structures have provided invaluable mechanistic insights by defining the chemical and physical features underlying costimulatory receptor:ligand specificity, affinity, oligomeric state, and valency. Furthermore, these structures revealed general architectural features that are important for the integration of these interactions and their associated signaling pathways into overall cellular physiology. Recent technological advances in structural biology promise unprecedented opportunities for furthering our understanding of the structural features and mechanisms that govern costimulation. In this review, we highlight unique insights that have been revealed by structures of costimulatory molecules from the immunoglobulin and tumor necrosis factor superfamilies and describe a vision for future structural and mechanistic analysis of costimulation. This vision includes simple strategies for the selection of candidate molecules for structure determination and highlights the critical role of structure in the design of mutant costimulatory molecules for the generation of in vivo structure-function correlations in a mammalian model system. This integrated 'atoms-to-animals' paradigm provides a comprehensive approach for defining atomic and molecular mechanisms.


Assuntos
Ativação Linfocitária/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Cristalografia por Raios X , Genômica , Humanos , Dados de Sequência Molecular , Conformação Proteica , Receptores de Superfície Celular/genética , Alinhamento de Sequência
12.
Proc Natl Acad Sci U S A ; 106(11): 4154-9, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19251641

RESUMO

Protein secretion is a common property of pathogenic microbes. Gram-negative bacterial pathogens use at least 6 distinct extracellular protein secretion systems to export proteins through their multilayered cell envelope and in some cases into host cells. Among the most widespread is the newly recognized Type VI secretion system (T6SS) which is composed of 15-20 proteins whose biochemical functions are not well understood. Using crystallographic, biochemical, and bioinformatic analyses, we identified 3 T6SS components, which are homologous to bacteriophage tail proteins. These include the tail tube protein; the membrane-penetrating needle, situated at the distal end of the tube; and another protein associated with the needle and tube. We propose that T6SS is a multicomponent structure whose extracellular part resembles both structurally and functionally a bacteriophage tail, an efficient machine that translocates proteins and DNA across lipid membranes into cells.


Assuntos
Proteínas de Bactérias/química , Evolução Biológica , Caudovirales/química , Bactérias Gram-Negativas/patogenicidade , Proteínas de Membrana Transportadoras/fisiologia , Proteínas Virais/química , Proteínas de Bactérias/metabolismo , Bacteriófagos/química , Bactérias Gram-Negativas/química , Proteínas de Membrana Transportadoras/metabolismo , Conformação Proteica , Homologia Estrutural de Proteína
13.
Int J Biol Macromol ; 210: 494-503, 2022 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-35504420

RESUMO

Tuberculosis, caused by Mycobacterium tuberculosis, is predominantly a disease of the lungs acquired by inhaling mycobacteria from infected individuals via airborne droplets. In order to facilitate their entry into the alveolar macrophages, mycobacteria have a collection of pathogen-associated molecular patterns (PAMPs) on their surface that are known to detect certain pattern recognition receptors present on the surface of host cells. A major group of these PAMPs includes mycobacterial lipoproteins, of which, the 19 kDa surface antigen LpqH, has been reported to play a critical role in both host-pathogen interactions as well as pleiotropic immune regulation. Despite its crucial involvement in tuberculosis, the detailed structure-function relationship of this protein remains to be explored. Here, we report the high-resolution crystal structure of the non-acylated LpqH (LpqH48-159) at a resolution of 1.26 Å, which adopts a unique fold. Flow cytometry-based experiments show that the protein can bind and induce apoptosis in PMA-activated human monocytic cell line THP-1, indicative of the preservation of functionality of the protein. Furthermore, analysis of conservation of LpqH sequences from Mycobacterium species reveals a patch of conserved residues on the surface which may play a role in its binding partner recognition and hence in host-pathogen interaction.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Lipoproteínas/metabolismo , Monócitos/metabolismo , Moléculas com Motivos Associados a Patógenos/metabolismo , Tuberculose/microbiologia
14.
Int Immunol ; 22(3): 191-203, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20093428

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease characterized by T cell-mediated destruction of insulin-producing pancreatic beta cells. In both humans and the non-obese diabetic (NOD) mouse model of T1D, class II MHC alleles are the primary determinant of disease susceptibility. However, class I MHC genes also influence risk. These findings are consistent with the requirement for both CD4(+) and CD8(+) T cells in the pathogenesis of T1D. Although a large body of work has permitted the identification of multiple mechanisms to explain the diabetes-protective effect of particular class II MHC alleles, studies examining the protective influence of class I alleles are lacking. Here, we explored this question by performing biochemical and structural analyses of the murine class I MHC molecule H-2K(wm7), which exerts a diabetes-protective effect in NOD mice. We have found that H-2K(wm7) molecules are predominantly occupied by the single self-peptide VNDIFERI, derived from the ubiquitous protein histone H2B. This unexpected finding suggests that the inability of H-2K(wm7) to support T1D development could be due, at least in part, to the failure of peptides from critical beta-cell antigens to adequately compete for binding and be presented to T cells. Predominant presentation of a single peptide would also be expected to influence T-cell selection, potentially leading to a reduced ability to select a diabetogenic CD8(+) T-cell repertoire. The report that one of the predominant peptides bound by T1D-protective HLA-A*31 is histone derived suggests the potential translation of our findings to human diabetes-protective class I MHC molecules.


Assuntos
Diabetes Mellitus Tipo 1/genética , Predisposição Genética para Doença , Antígenos H-2/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Separação Celular , Cristalografia , Feminino , Citometria de Fluxo , Antígenos H-2/química , Antígenos H-2/genética , Histonas/química , Histonas/genética , Histonas/metabolismo , Humanos , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos NOD , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Filogenia , Estrutura Quaternária de Proteína
15.
Proc Natl Acad Sci U S A ; 105(2): 635-40, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18182486

RESUMO

Glucocorticoid-induced TNF receptor ligand (GITRL), a recently identified member of the TNF superfamily, binds to its receptor, GITR, on both effector and regulatory T cells and generates positive costimulatory signals implicated in a wide range of T cell functions. In contrast to all previously characterized homotrimeric TNF family members, the mouse GITRL crystal structure reveals a previously unrecognized dimeric assembly that is stabilized via a unique "domain-swapping" interaction. Consistent with its crystal structure, mouse GITRL exists as a stable dimer in solution. Structure-guided mutagenesis studies confirmed the determinants responsible for dimerization and support a previously unrecognized receptor-recognition surface for mouse GITRL that has not been observed for any other TNF family members. Taken together, the unique structural and biochemical behavior of mouse GITRL, along with the unusual domain organization of murine GITR, support a previously unrecognized mechanism for signaling within the TNF superfamily.


Assuntos
Evolução Molecular , Fatores de Necrose Tumoral/metabolismo , Sequência de Aminoácidos , Animais , Cristalografia por Raios X/métodos , Dimerização , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Cinética , Camundongos , Conformação Molecular , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Receptores de Fator de Crescimento Neural/química , Receptores do Fator de Necrose Tumoral/química , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Linfócitos T/metabolismo , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/fisiologia
16.
Protein Sci ; 30(9): 1958-1973, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34191384

RESUMO

T-cell co-stimulation through CD28/CTLA4:B7-1/B7-2 axis is one of the extensively studied pathways that resulted in the discovery of several FDA-approved drugs for autoimmunity and cancer. However, many aspects of the signaling mechanism remain elusive, including oligomeric association and clustering of B7-2 on the cell surface. Here, we describe the structure of the IgV domain of B7-2 and its cryptic association into 1D arrays that appear to represent the pre-signaling state of B7-2 on the cell membrane. Super-resolution microscopy experiments on heterologous cells expressing B7-2 and B7-1 suggest, B7-2 form relatively elongated and larger clusters compared to B7-1. The sequence and structural comparison of other B7 family members, B7-1:CTLA4 and B7-2:CTLA-4 complex structures, support our view that the observed B7-2 1D zipper array is physiologically important. This observed 1D zipper-like array also provides an explanation for its clustering, and upright orientation on the cell surface, and avoidance of spurious signaling.


Assuntos
Antígeno B7-1/química , Antígeno B7-2/química , Antígenos CD28/química , Antígeno CTLA-4/química , Sequência de Aminoácidos , Animais , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Sítios de Ligação , Antígenos CD28/genética , Antígenos CD28/metabolismo , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Neurônios/citologia , Neurônios/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
17.
J Exp Med ; 218(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34709351

RESUMO

HVEM is a TNF (tumor necrosis factor) receptor contributing to a broad range of immune functions involving diverse cell types. It interacts with a TNF ligand, LIGHT, and immunoglobulin (Ig) superfamily members BTLA and CD160. Assessing the functional impact of HVEM binding to specific ligands in different settings has been complicated by the multiple interactions of HVEM and HVEM binding partners. To dissect the molecular basis for multiple functions, we determined crystal structures that reveal the distinct HVEM surfaces that engage LIGHT or BTLA/CD160, including the human HVEM-LIGHT-CD160 ternary complex, with HVEM interacting simultaneously with both binding partners. Based on these structures, we generated mouse HVEM mutants that selectively recognized either the TNF or Ig ligands in vitro. Knockin mice expressing these muteins maintain expression of all the proteins in the HVEM network, yet they demonstrate selective functions for LIGHT in the clearance of bacteria in the intestine and for the Ig ligands in the amelioration of liver inflammation.


Assuntos
Antígenos CD/metabolismo , Receptores Imunológicos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/química , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Antígenos CD/química , Antígenos CD/genética , Cristalografia por Raios X , Drosophila/citologia , Drosophila/genética , Feminino , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Mutação , Receptores Imunológicos/química , Receptores Imunológicos/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/química , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Yersiniose/genética , Yersiniose/patologia
18.
Proc Natl Acad Sci U S A ; 104(49): 19452-7, 2007 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-18040044

RESUMO

Glucocorticoid-induced TNF receptor ligand (GITRL), a recently identified member of the TNF family, binds to its receptor GITR on both effector and regulatory T cells and generates positive costimulatory signals implicated in a wide range of T cell functions. Structural analysis reveals that the human GITRL (hGITRL) ectodomain self-assembles into an atypical expanded homotrimer with sparse monomer-monomer interfaces. Consistent with the small intersubunit interfaces, hGITRL exhibits a relatively weak tendency to trimerize in solution and displays a monomer-trimer equilibrium not reported for other TNF family members. This unique assembly behavior has direct implications for hGITRL-GITR signaling, because enforced trimerization of soluble hGITRL ectodomain results in an approximately 100-fold increase in its receptor binding affinity and also in enhanced costimulatory activity. The apparent reduction in affinity that is the consequence of this dynamic equilibrium may represent a mechanism to realize the biologically optimal level of signaling through the hGITRL-GITR pathway, as opposed to the maximal achievable level.


Assuntos
Fatores de Necrose Tumoral/química , Sítios de Ligação , Cristalografia por Raios X , Proteína Relacionada a TNFR Induzida por Glucocorticoide , Humanos , Mutação , Conformação Proteica , Receptores de Fator de Crescimento Neural/química , Receptores do Fator de Necrose Tumoral/química , Soluções , Fatores de Necrose Tumoral/genética
19.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 5): 434-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19390148

RESUMO

Glucocorticoid-induced TNF receptor ligand (GITRL), a prominent member of the TNF superfamily, activates its receptor on both effector and regulatory T cells to generate critical costimulatory signals that have been implicated in a wide range of T-cell immune functions. The crystal structures of murine and human orthologs of GITRL recombinantly expressed in Escherichia coli have previously been determined. In contrast to all classical TNF structures, including the human GITRL structure, murine GITRL demonstrated a unique ;strand-exchanged' dimeric organization. Such a novel assembly behavior indicated a dramatic impact on receptor activation as well as on the signaling mechanism associated with the murine GITRL costimulatory system. In this present work, the 1.8 A resolution crystal structure of murine GITRL expressed in Drosophila melanogaster S2 cells is reported. The eukaryotic protein-expression system allows transport of the recombinant protein into the extracellular culture medium, thus maximizing the possibility of obtaining correctly folded material devoid of any folding/assembly artifacts that are often suspected with E. coli-expressed proteins. The S2 cell-expressed murine GITRL adopts an identical ;strand-exchanged' dimeric structure to that observed for the E. coli-expressed protein, thus conclusively demonstrating the novel quaternary structure assembly behavior of murine GITRL.


Assuntos
Fatores de Necrose Tumoral/química , Animais , Linhagem Celular , Clonagem Molecular , Cristalografia por Raios X , Dimerização , Drosophila melanogaster/citologia , Escherichia coli , Glicosilação , Camundongos , Modelos Moleculares , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase , Conformação Proteica , Dobramento de Proteína , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Especificidade da Espécie , Fatores de Necrose Tumoral/biossíntese , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/isolamento & purificação
20.
Sci Rep ; 9(1): 19191, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31844079

RESUMO

Antibodies targeting negative regulators of immune checkpoints have shown unprecedented and durable response against variety of malignancies. While the concept of blocking the negative regulators of the immune checkpoints using mAbs appears to be an outstanding approach, their limited effect and several drawbacks, calls for the rational design of next generation of therapeutics. Soluble isoforms of the negative regulators of immune checkpoint pathways are expressed naturally and regulate immune responses. This suggests, affinity-modified versions of these self-molecules could be effective lead molecules for immunotherapy. To obtain better insights on the hotspot regions for modification, we have analysed structures of 18 immune receptor:ligand complexes containing the IgV domain. Interestingly, this analysis reveals that the CC' loop of IgV domain, a loop which is distinct from CDRs of antibodies, plays a pivotal role in affinity modulation, which was previously not highlighted. It is noteworthy that a ~5-residue long CC' loop in a ~120 residue protein makes significant number of hydrophobic and polar interactions with its cognate ligand. The post-interaction movement of CC' loop to accommodate the incoming ligands, seems to provide additional affinity to the interactions. In silico replacement of the CC' loop of TIGIT with that of Nectin-2 and PVR followed by protein docking trials suggests a key role of the CC' loop in affinity modulation in the TIGIT/Nectin pathway. The CC' loop appears to be a hotspot for the affinity modification without affecting the specificity to their cognate receptors.


Assuntos
Imunoglobulinas/imunologia , Domínios Proteicos/imunologia , Receptores Imunológicos/imunologia , Anticorpos Monoclonais/imunologia , Humanos , Ligantes , Nectinas/imunologia , Neoplasias/imunologia , Isoformas de Proteínas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA