Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 151(7): 1557-68, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260142

RESUMEN

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.


Asunto(s)
Antígenos HLA-D/química , Antígenos HLA-D/metabolismo , Antígeno HLA-DR1/química , Antígeno HLA-DR1/metabolismo , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia
2.
Nat Immunol ; 14(7): 699-705, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23666294

RESUMEN

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.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Muromegalovirus/inmunología , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Secuencias de Aminoácidos/inmunología , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Transducción de Señal/inmunología , Organismos Libres de Patógenos Específicos , Resonancia por Plasmón de Superficie
3.
Mol Cell ; 68(4): 659-672.e9, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29149594

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2 , Mitocondrias , Péptidos , Proteína Destructora del Antagonista Homólogo bcl-2 , Animales , Proteína 11 Similar a Bcl2/química , Proteína 11 Similar a Bcl2/farmacología , Línea Celular Transformada , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Péptidos/química , Péptidos/farmacología , Unión Proteica , Relación Estructura-Actividad , Proteína Destructora del Antagonista Homólogo bcl-2/química , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(48): e2208058119, 2022 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-36409917

RESUMEN

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.


Asunto(s)
Receptores de Antígenos de Linfocitos B , Linfocitos T , Humanos , Ratones , Animales , Receptores de Antígenos de Linfocitos B/metabolismo , Linfocitos T/metabolismo , Microscopía por Crioelectrón , Receptores de Antígenos de Linfocitos T/metabolismo , Membrana Celular/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
5.
Genome Res ; 30(12): 1781-1788, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33093069

RESUMEN

Effective public response to a pandemic relies upon accurate measurement of the extent and dynamics of an outbreak. Viral genome sequencing has emerged as a powerful approach to link seemingly unrelated cases, and large-scale sequencing surveillance can inform on critical epidemiological parameters. Here, we report the analysis of 864 SARS-CoV-2 sequences from cases in the New York City metropolitan area during the COVID-19 outbreak in spring 2020. The majority of cases had no recent travel history or known exposure, and genetically linked cases were spread throughout the region. Comparison to global viral sequences showed that early transmission was most linked to cases from Europe. Our data are consistent with numerous seeds from multiple sources and a prolonged period of unrecognized community spreading. This work highlights the complementary role of genomic surveillance in addition to traditional epidemiological indicators.


Asunto(s)
COVID-19 , Genoma Viral , Pandemias , Filogenia , SARS-CoV-2/genética , Secuenciación Completa del Genoma , COVID-19/epidemiología , COVID-19/genética , COVID-19/transmisión , Femenino , Humanos , Masculino , Ciudad de Nueva York
6.
Immunol Cell Biol ; 101(10): 923-935, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37721869

RESUMEN

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.


Asunto(s)
Inteligencia Artificial , Investigación Biomédica , Tecnología
7.
Nat Immunol ; 12(1): 54-61, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21131964

RESUMEN

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.


Asunto(s)
Antígenos HLA-D/metabolismo , Antígeno HLA-DR2/metabolismo , Proteínas Mutantes/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Presentación de Antígeno , Células CHO , Catálisis , Cricetinae , Cricetulus , Antígenos HLA-D/química , Antígenos HLA-D/genética , Antígeno HLA-DR2/química , Antígeno HLA-DR2/genética , Humanos , Modelos Químicos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Unión Proteica , Resonancia por Plasmón de Superficie , Transgenes/genética
8.
J Biol Chem ; 297(1): 100900, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34157285

RESUMEN

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.


Asunto(s)
Antígeno B7-2/química , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Membrana Celular/metabolismo , Regulación hacia Abajo , Células HEK293 , Células HeLa , Humanos , Mutación , Dominios Proteicos , Transporte de Proteínas
9.
Blood ; 135(4): 287-292, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31697803

RESUMEN

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.


Asunto(s)
Sustitución de Aminoácidos , Trastornos Mieloproliferativos/genética , Receptores de Trombopoyetina/genética , Animales , Línea Celular , Exones , Humanos , Ratones , Modelos Moleculares , Mutación , Dominios Proteicos , Receptores de Trombopoyetina/química
10.
Am J Pathol ; 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32650002

RESUMEN

The Publisher regrets that this article is an accidental duplication of an article that has already been published, https://doi.org/10.1016/j.ajpath.2020.07.001. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.

11.
Am J Pathol ; 190(9): 1881-1887, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32628931

RESUMEN

The dynamics of viral load (VL) of the severe acute respiratory syndrome coronavirus 2 and its association with different clinical parameters remain poorly characterized in the US patient population. Herein, we investigate associations between VL and parameters, such as severity of symptoms, disposition (admission versus direct discharge), length of hospitalization, admission to the intensive care unit, length of oxygen support, and overall survival in 205 patients from a tertiary care center in New York City. VL was determined using quantitative PCR and log10 transformed for normalization. Associations were tested with univariate and multivariate regression models. Diagnostic VL was significantly lower in hospitalized patients than in patients not hospitalized (log10 VL = 3.3 versus 4.0; P = 0.018) after adjusting for age, sex, race, body mass index, and comorbidities. Higher VL was associated with shorter duration of the symptoms in all patients and hospitalized patients only and shorter hospital stay (coefficient = -2.02, -2.61, and -2.18; P < 0.001, P = 0.002, and P = 0.013, respectively). No significant association was noted between VL, admission to intensive care unit, length of oxygen support, and overall survival. Our findings suggest a higher shedding risk in less symptomatic patients, an important consideration for containment strategies. Furthermore, we identify a novel association between VL and history of cancer. Larger studies are warranted to validate our findings.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/terapia , Neumonía Viral/epidemiología , Neumonía Viral/terapia , Carga Viral , Adulto , COVID-19 , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Masculino , Persona de Mediana Edad , Ciudad de Nueva York/epidemiología , Pandemias , Factores de Riesgo , SARS-CoV-2
12.
J Biol Chem ; 294(7): 2470-2485, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30554144

RESUMEN

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.


Asunto(s)
Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Proteínas de la Membrana/biosíntesis , Ubiquitina-Proteína Ligasas/biosíntesis , Antígenos CD4/química , Antígenos CD4/genética , Antígenos CD4/metabolismo , Células HEK293 , Antígeno HLA-A2/química , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Dominios Proteicos , Estructura Secundaria de Proteína , Serina/química , Serina/genética , Serina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
13.
Proc Natl Acad Sci U S A ; 114(28): E5645-E5654, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28652325

RESUMEN

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.


Asunto(s)
Complejo CD3/metabolismo , Membrana Celular/metabolismo , Receptores de IgG/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Proteínas Ligadas a GPI/metabolismo , Glicosilación , Células HEK293 , Humanos , Enlace de Hidrógeno , Espectroscopía de Resonancia Magnética , Ratones , Mutagénesis Sitio-Dirigida , Dominios Proteicos , Multimerización de Proteína , Receptores de IgE/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal
14.
Int J Mol Sci ; 21(19)2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33050044

RESUMEN

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.


Asunto(s)
Ingeniería Celular/métodos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Comunicación Celular/inmunología , Humanos , Neoplasias/terapia , Dominios Proteicos , Receptores Quiméricos de Antígenos/química
15.
Langmuir ; 35(25): 8344-8356, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31122018

RESUMEN

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.


Asunto(s)
Cristalización/métodos , Membrana Dobles de Lípidos/química , Glicéridos/química , Difracción de Rayos X
16.
Proc Natl Acad Sci U S A ; 113(43): E6649-E6658, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27791034

RESUMEN

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.


Asunto(s)
Complejo CD3/química , Membrana Celular/ultraestructura , Subunidades de Proteína/química , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T gamma-delta/química , Secuencia de Aminoácidos , Sitios de Unión , Complejo CD3/genética , Complejo CD3/inmunología , Membrana Celular/química , Membrana Celular/inmunología , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Linfocitos T/química , Linfocitos T/inmunología
17.
Biophys J ; 114(5): 1030-1035, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29395047

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/metabolismo , Secuencia de Aminoácidos , Humanos , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Multimerización de Proteína , Estructura Cuaternaria de Proteína
18.
J Biol Chem ; 290(42): 25307-21, 2015 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-26324715

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Eritrocitos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Receptores de Complemento/metabolismo , Animales , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/inmunología
19.
Immunol Cell Biol ; 99(7): 674-676, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34180552
20.
J Biol Chem ; 289(12): 8240-51, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24488493

RESUMEN

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.


Asunto(s)
Proteínas Aviares/química , Complejo CD3/química , Pollos/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Secuencia de Aminoácidos , Animales , Complejo CD3/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Multimerización de Proteína , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA