Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 822
Filtrar
1.
Mol Biol Cell ; 33(1): ar6, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34731029

RESUMO

The neonatal Fc receptor (FcRn) is responsible for the recycling of endocytosed albumin and IgG, and contributes to their long plasma half-life. We recently identified an FcRn-dependent recycling pathway from macropinosomes in macrophages; however, little is known about the dynamics of intracellular FcRn-ligand interactions to promote recycling. Here we demonstrate a multiplexed biophysical fluorescent microscopy approach to resolve the spatiotemporal dynamics of albumin-FcRn interactions in living bone marrow-derived macrophages (BMDMs). We used the phasor approach to fluorescence lifetime imaging microscopy (FLIM) of Förster resonance energy transfer (FRET) to detect the interaction of a FcRn-mCherry fusion protein with endocytosed Alexa Fluor 488-labeled human serum albumin (HSA-AF488) in BMDMs, and raster image correlation spectroscopy (RICS) analysis of single fluorescent-labeled albumin molecules to monitor the diffusion kinetics of internalized albumin. Our data identified a major fraction of immobile HSA-AF488 molecules in endosomal structures of human FcRn-positive mouse macrophages and an increase in FLIM-FRET following endocytosis, including detection of FRET in tubular-like structures. A nonbinding mutant of albumin showed minimum FLIM-FRET and high mobility. These data reveal the kinetics of FcRn-ligand binding within endosomal structures for recruitment into transport carriers for recycling. These approaches have wide applicability for analyses of intracellular ligand-receptor interactions.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Macrófagos/metabolismo , Receptores Fc/metabolismo , Albuminas/metabolismo , Animais , Endocitose/fisiologia , Endossomos/metabolismo , Feminino , Fluorescência , Transferência Ressonante de Energia de Fluorescência/métodos , Meia-Vida , Células HeLa , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Cinética , Ligantes , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Ligação Proteica , Receptores Fc/fisiologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-33583391

RESUMO

Angiotensin-converting enzyme (ACE) is a zinc-dependent dicarboxypeptidase with two catalytic components, which has an important role in regulating blood pressure by converting angiotensin I to angiotensin II. ACE breaks down other peptides besides angiotensin I and has a variety of physiological effects together with renal growth and reproduction in men. ACE also acts on innate and acquired immune systems by affecting macrophage and neutrophil function, and these outcomes are exacerbated due to the overexpression of ACE. Overexpression of ACE in macrophages imposes antitumor and antimicrobial response, and it enhances the ability of neutrophils to produced super peroxide that has a bactericidal effect. ACE is also known to contribute to the expression of Major Histocompatibility Complex (MHC) class I and MHC class II peptides through enzymatic alterations of these peptides. Apprehending the expression of ACE and its effects on myeloid cell (myelogenous cells) activity can be promising in therapeutic interventions, including treatment of infection and malignancy.


Assuntos
Imunidade/genética , Peptidil Dipeptidase A/fisiologia , Angiotensinas/metabolismo , Angiotensinas/fisiologia , Animais , Apresentação de Antígeno/genética , Apresentação de Antígeno/fisiologia , Feminino , Antígenos de Histocompatibilidade Classe I/fisiologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Humanos , Imunidade/fisiologia , Infecções/genética , Infecções/imunologia , Infecções/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Peptidil Dipeptidase A/genética
3.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34845016

RESUMO

Unlike conventional αß T cells, γδ T cells typically recognize nonpeptide ligands independently of major histocompatibility complex (MHC) restriction. Accordingly, the γδ T cell receptor (TCR) can potentially recognize a wide array of ligands; however, few ligands have been described to date. While there is a growing appreciation of the molecular bases underpinning variable (V)δ1+ and Vδ2+ γδ TCR-mediated ligand recognition, the mode of Vδ3+ TCR ligand engagement is unknown. MHC class I-related protein, MR1, presents vitamin B metabolites to αß T cells known as mucosal-associated invariant T cells, diverse MR1-restricted T cells, and a subset of human γδ T cells. Here, we identify Vδ1/2- γδ T cells in the blood and duodenal biopsy specimens of children that showed metabolite-independent binding of MR1 tetramers. Characterization of one Vδ3Vγ8 TCR clone showed MR1 reactivity was independent of the presented antigen. Determination of two Vδ3Vγ8 TCR-MR1-antigen complex structures revealed a recognition mechanism by the Vδ3 TCR chain that mediated specific contacts to the side of the MR1 antigen-binding groove, representing a previously uncharacterized MR1 docking topology. The binding of the Vδ3+ TCR to MR1 did not involve contacts with the presented antigen, providing a basis for understanding its inherent MR1 autoreactivity. We provide molecular insight into antigen-independent recognition of MR1 by a Vδ3+ γδ TCR that strengthens an emerging paradigm of antibody-like ligand engagement by γδ TCRs.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Linfócitos Intraepiteliais/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Adulto , Apresentação de Antígeno , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Linfócitos Intraepiteliais/fisiologia , Ligantes , Masculino , Antígenos de Histocompatibilidade Menor/química , Antígenos de Histocompatibilidade Menor/fisiologia , Células T Invariantes Associadas à Mucosa/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/fisiologia
4.
Science ; 370(6519): 941-950, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33122426

RESUMO

Mast cells (MCs) are central effector cells in allergic reactions that are often mediated by immunoglobulin E (IgE). Allergies commonly start at an early age, and both MCs and IgE are detectable in fetuses. However, the origin of fetal IgE and whether fetal MCs can degranulate in response to IgE-dependent activation are presently unknown. Here, we show that human and mouse fetal MCs phenotypically mature through pregnancy and can be sensitized by maternal IgE. IgE crossed the placenta, dependent on the fetal neonatal Fc receptor (FcRN), and sensitized fetal MCs for allergen-specific degranulation. Both passive and active prenatal sensitization conferred allergen sensitivity, resulting in postnatal skin and airway inflammation after the first allergen encounter. We report a role for MCs within the developing fetus and demonstrate that fetal MCs may contribute to antigen-specific vertical transmission of allergic disease.


Assuntos
Feto/imunologia , Hipersensibilidade/imunologia , Imunoglobulina E/imunologia , Mastócitos/imunologia , Troca Materno-Fetal/imunologia , Alérgenos/imunologia , Ambrosia/imunologia , Animais , Degranulação Celular/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Placenta/imunologia , Gravidez , Receptores Fc/fisiologia
5.
J Exp Med ; 217(10)2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32658257

RESUMO

IgG immune complexes (ICs) promote autoimmunity through binding fragment crystallizable (Fc) γ-receptors (FcγRs). Of these, the highly prevalent FcγRIIa (CD32a) histidine (H)-131 variant (CD32aH) is strongly linked to human autoimmune diseases through unclear mechanisms. We show that, relative to the CD32a arginine (R)-131 (CD32aR) variant, CD32aH more avidly bound human (h) IgG1 IC and formed a ternary complex with the neonatal Fc receptor (FcRn) under acidic conditions. In primary human and mouse cells, both CD32a variants required FcRn to induce innate and adaptive immune responses to hIgG1 ICs, which were augmented in the setting of CD32aH. Conversely, FcRn induced responses to IgG IC independently of classical FcγR, but optimal responses required FcRn and FcγR. Finally, FcRn blockade decreased inflammation in a rheumatoid arthritis model without reducing circulating autoantibody levels, providing support for FcRn's direct role in IgG IC-associated inflammation. Thus, CD32a and FcRn coregulate IgG IC-mediated immunity in a manner favoring the CD32aH variant, providing a novel mechanism for its disease association.


Assuntos
Autoimunidade/imunologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Imunoglobulina G/imunologia , Receptores Fc/fisiologia , Imunidade Adaptativa/imunologia , Animais , Artrite Reumatoide/imunologia , Suscetibilidade a Doenças , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunidade Inata/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Fc/imunologia , Receptores de IgG/imunologia
6.
J Hematol Oncol ; 13(1): 74, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32517713

RESUMO

BACKGROUND: Melanoma patients who have detectable serum soluble NKG2D ligands either at the baseline or post-treatment of PD1/PDL1 blockade exhibit poor overall survival. Among families of soluble human NKG2D ligands, the soluble human MHC I chain-related molecule (sMIC) was found to be elevated in melanoma patients and mostly associated with poor response to PD1/PDL1 blockade therapy. METHODS: In this study, we aim to investigate whether co-targeting tumor-released sMIC enhances the therapeutic outcome of PD1/PDL1 blockade therapy for melanoma. We implanted sMIC-expressing B16F10 melanoma tumors into syngeneic host and evaluated therapeutic efficacy of anti-sMIC antibody and anti-PDL1 antibody combination therapy in comparison with monotherapy. We analyzed associated effector mechanism. We also assessed sMIC/MIC prevalence in metastatic human melanoma tumors. RESULTS: We found that the combination therapy of the anti-PDL1 antibody with an antibody targeting sMIC significantly improved animal survival as compared to monotherapies and that the effect of combination therapy depends significantly on NK cells. We show that combination therapy significantly increased IL-2Rα (CD25) on NK cells which sensitizes NK cells to low dose IL-2 for survival. We demonstrate that sMIC negatively reprograms gene expression related to NK cell homeostatic survival and proliferation and that antibody clearing sMIC reverses the effect of sMIC and reprograms NK cell for survival. We further show that sMIC/MIC is abundantly present in metastatic human melanoma tumors. CONCLUSIONS: Our findings provide a pre-clinical proof-of-concept and a new mechanistic understanding to underscore the significance of antibody targeting sMIC to improve therapeutic efficacy of anti-PD1/PDL1 antibody for MIC/sMIC+ metastatic melanoma patients.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Antígeno B7-H1/antagonistas & inibidores , Antígenos de Histocompatibilidade Classe I/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Células Matadoras Naturais/citologia , Melanoma Experimental/tratamento farmacológico , Subfamília K de Receptores Semelhantes a Lectina de Células NK/agonistas , Proteínas de Neoplasias/antagonistas & inibidores , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/farmacologia , Linfócitos T CD8-Positivos/imunologia , Sobrevivência Celular , Sinergismo Farmacológico , Antígenos de Histocompatibilidade Classe I/fisiologia , Homeostase , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Subunidade alfa de Receptor de Interleucina-2/genética , Ligantes , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/fisiologia , Solubilidade , Organismos Livres de Patógenos Específicos , Transplante Isogênico , Microambiente Tumoral , Regulação para Cima
7.
Cancer Med ; 9(13): 4736-4742, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32368865

RESUMO

Since the neonatal IgG Fc receptor (FcRn) was discovered, it was found to be involved in immunoglobulin recycling and biodistribution, immune complexes routing, antigen presentation, humoral immune response, and cancer immunosurveillance. The latest data show that FcRn plays a part in cancer pathophysiology. In various types of cancers, such as lung and colorectal cancer, FcRn has been described as an early marker for prognosis. Dysregulation of FcRn expression by cancer cells allows them to increase their metabolism, and this process could be exploited for passive targeting of cytotoxic drugs. However, the roles of this receptor depend on whether the studied cell population is the tumor tissue or the infiltrating cells, bringing forward the need for further studies.


Assuntos
Antígenos de Histocompatibilidade Classe I/fisiologia , Monitorização Imunológica , Neoplasias/imunologia , Receptores Fc/fisiologia , Animais , Biomarcadores Tumorais , Carcinogênese/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Células Matadoras Naturais/imunologia , Camundongos , Neoplasias/metabolismo , Neoplasias/mortalidade , Prognóstico , Receptores Fc/genética , Receptores Fc/metabolismo
8.
Proc Natl Acad Sci U S A ; 117(21): 11636-11647, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32404419

RESUMO

Micropolymorphisms within human leukocyte antigen (HLA) class I molecules can change the architecture of the peptide-binding cleft, leading to differences in peptide presentation and T cell recognition. The impact of such HLA variation on natural killer (NK) cell recognition remains unclear. Given the differential association of HLA-B*57:01 and HLA-B*57:03 with the control of HIV, recognition of these HLA-B57 allomorphs by the killer cell immunoglobulin-like receptor (KIR) 3DL1 was compared. Despite differing by only two polymorphic residues, both buried within the peptide-binding cleft, HLA-B*57:01 more potently inhibited NK cell activation. Direct-binding studies showed KIR3DL1 to preferentially recognize HLA-B*57:01, particularly when presenting peptides with positively charged position (P)Ω-2 residues. In HLA-B*57:01, charged PΩ-2 residues were oriented toward the peptide-binding cleft and away from KIR3DL1. In HLA-B*57:03, the charged PΩ-2 residues protruded out from the cleft and directly impacted KIR3DL1 engagement. Accordingly, KIR3DL1 recognition of HLA class I ligands is modulated by both the peptide sequence and conformation, as determined by the HLA polymorphic framework, providing a rationale for understanding differences in clinical associations.


Assuntos
Antígenos de Histocompatibilidade Classe I/genética , Células Matadoras Naturais/fisiologia , Polimorfismo Genético/genética , Antígenos de Histocompatibilidade Classe I/química , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Ativação Linfocitária/genética , Modelos Moleculares , Polimorfismo Genético/fisiologia , Receptores KIR/genética
9.
Elife ; 92020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32238263

RESUMO

Variation in the risk and severity of many autoimmune diseases, malignancies and infections is strongly associated with polymorphisms at the HLA class I loci. These genetic associations provide a powerful opportunity for understanding the etiology of human disease. HLA class I associations are often interpreted in the light of 'protective' or 'detrimental' CD8+ T cell responses which are restricted by the host HLA class I allotype. However, given the diverse receptors which are bound by HLA class I molecules, alternative interpretations are possible. As well as binding T cell receptors on CD8+ T cells, HLA class I molecules are important ligands for inhibitory and activating killer immunoglobulin-like receptors (KIRs) which are found on natural killer cells and some T cells; for the CD94:NKG2 family of receptors also expressed mainly by NK cells and for leukocyte immunoglobulin-like receptors (LILRs) on myeloid cells. The aim of this study is to develop an immunogenetic approach for identifying and quantifying the relative contribution of different receptor-ligand interactions to a given HLA class I disease association and then to use this approach to investigate the immune interactions underlying HLA class I disease associations in three viral infections: Human T cell Leukemia Virus type 1, Human Immunodeficiency Virus type 1 and Hepatitis C Virus as well as in the inflammatory condition Crohn's disease.


When considering someone's risk of disease, every person is different but some similarities can be found when looking across populations. Some people are more likely to develop a certain disease, while others are protected in some way. Part of this variation is explained by the individual's genes, while their lifestyle and environment are other factors. Numerous studies have looked for associations between different versions of genes, known as gene variants, and the occurrence of disease to identify who is at risk. There is one cluster of genes called the HLA genes that is a well-known hotspot for disease associations. The HLA cluster is named for the group of proteins it encodes, called the human leukocyte antigen (HLA) complex. These cell-surface proteins regulate the immune system in humans. These proteins are present on the surface of cells, and they help the immune system distinguish foreign invaders such as viruses and bacteria from the body's own cells. Variants in the HLA genes are associated with more than 100 diseases, including infectious diseases like HIV, autoimmune conditions such as multiple sclerosis, and some cancers. However, while identifying which genetic variants are associated with an increased or decreased risk of disease is relatively simple, understanding why those genetic variants are associated with a particular disease is much harder. Debebe et al. have developed a new method to find out why certain gene variants in the HLA cluster are associated with disease in humans. They used this method to investigate known genetic variants associated with three viral infections: HIV, hepatitis C, and human leukemia virus ­ and one inflammatory disease: Crohn's disease. Critically, Debebe et al. looked at the interactions between different immune cells and the cell-surface proteins encoded by the HLA gene variants in different cases of these diseases. In doing so, the analysis was able to identify which cells of the immune system were responsible for the associations between gene variants and diseases. In principle, this method could be applied to study any disease in any species. It could also be used in classic gene association studies to test for false positive results and "passenger" mutations, two common problems that beset sound interpretations from these studies.


Assuntos
Infecções por HIV/imunologia , Infecções por HTLV-I/imunologia , Hepatite C/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Alelos , Doença de Crohn/imunologia , HIV-1 , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores KIR/metabolismo
10.
Proc Natl Acad Sci U S A ; 117(8): 4099-4108, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32047030

RESUMO

Mammalian cells present a fingerprint of their proteome to the adaptive immune system through the display of endogenous peptides on MHC-I complexes. MHC-I-bound peptides originate from protein degradation by the proteasome, suggesting that stably folded, long-lived proteins could evade monitoring. Here, we investigate the role in antigen presentation of the ribosome-associated quality control (RQC) pathway for the degradation of nascent polypeptides that are encoded by defective messenger RNAs and undergo stalling at the ribosome during translation. We find that degradation of model proteins by RQC results in efficient MHC-I presentation, independent of their intrinsic folding properties. Quantitative profiling of MHC-I peptides in wild-type and RQC-deficient cells by mass spectrometry showed that RQC substantially contributes to the composition of the immunopeptidome. Our results also identify endogenous substrates of the RQC pathway in human cells and provide insight into common principles causing ribosome stalling under physiological conditions.


Assuntos
Apresentação de Antígeno/fisiologia , Epitopos/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Ribossomos/fisiologia , Animais , Deleção de Genes , Regulação da Expressão Gênica , Células HeLa , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
11.
Hum Immunol ; 81(4): 127-133, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31558330

RESUMO

The human major histocompatibility complex includes a group of non-classical HLA class I genes, HLA-E, -F and -G. While nearly all focus since the discovery of these class Ib molecules have been on basic biochemistry and molecular biology of HLA-G and HLA-E, as well as their expression patterns, functions in immune modulation and during pregnancy, and also possible implications in a range of diseases, in infertility and pregnancy complications, HLA-F has nearly been ignored. However, recent discoveries show that HLA-F can be expressed as both open conformers binding to a number of KIRs on primarily NK cells, as well as peptide-bound HLA-F binding to ILT2 and ILT4. Furthermore, a number of reports indicate a possible involvement of HLA-F in viral infections, in cancer immunology, and in fertility and reproduction, which may initiate more interest in this rather unknown HLA class I molecule. In this short review, we focus on recent discoveries that indicate a functional role for HLA-F in reproduction and during pregnancy, and the role of HLA-F in relation to HLA-G.


Assuntos
Antígenos HLA-G/fisiologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Gravidez , Reprodução/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Células Matadoras Naturais/imunologia , Troca Materno-Fetal/imunologia , Complicações na Gravidez/imunologia
12.
Mol Biol Cell ; 31(3): 196-208, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31851583

RESUMO

The human herpesvirus-7 (HHV-7) U21 glycoprotein binds to class I major histocompatibility complex (MHC) molecules in the endoplasmic reticulum (ER) and reroutes them to lysosomes. How this single viral glycoprotein efficiently redirects the U21/class I MHC complex to the lysosomal compartment is poorly understood. To investigate the trafficking of HHV-7 U21, we followed synchronous release of U21 from the ER as it traffics through the secretory system. Sorting of integral membrane proteins from the trans-Golgi network (TGN) has been shown to occur through tubular carriers that emanate from the TGN or through vesicular carriers that recruit GGA (Golgi-localized, γ-ear-containing, ARF-binding protein), clathrin adaptors, and clathrin. Here, we present evidence for the existence of a third type of Golgi-derived carrier that is vesicular, yet clathrin independent. This U21-containing carrier also carries a Golgi membrane protein engineered to form inducible oligomers. We propose that U21 employs the novel mechanism of forming oligomeric complexes with class I MHC molecules that result in sorting of the oligomeric U21/class I MHC complexes to Golgi--derived quality control carriers destined for lysosomes.


Assuntos
Proteínas de Transporte/metabolismo , Herpesvirus Humano 7/metabolismo , Proteínas Virais/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Clatrina/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/virologia , Células HeLa , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Lisossomos/metabolismo , Lisossomos/fisiologia , Ligação Proteica , Transporte Proteico , Rede trans-Golgi/metabolismo
13.
J Clin Pharmacol ; 60(4): 466-476, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31729044

RESUMO

An understanding of pediatric pharmacokinetics (PK) is essential for first-in-pediatric dose selection and clinical trial design. At present, there is no reliable way to scale the PK of monoclonal antibodies and immunoglobulin G drug products from adults to young children or to premature infants-a vulnerable population with a rapidly growing drug development pipeline. In this work, pediatric physiologically based PK models are constructed in PK-Sim and Mobi to explore the PK of pagibaximab, palivizumab, MEDI8897, and intravenous immunoglobulin in preterm infants. In addition to considering ontogeny in pediatric organ volumes, organ composition, blood flow rates, and hematocrit, advanced ontogeny is applied for 3 key parameters: capillary surface area, hematopoietic cell concentration, and lymph flow rate. The role and importance of each parameter for determining pediatric clearance (CL) and volume of distribution at steady state (VSS ) are quantitatively assessed with a local sensitivity analysis. In addition, the uncertainty around parameters with limited information in pediatrics is addressed (eg, free neonatal Fc receptor concentration). The full ontogeny parameterization yields pediatric PK predictions that are within 1.5-fold prediction error >90% of the time for preterm infants, with an absolute average fold error of 1.05. This result suggests that many of the key factors related to ontogeny are appropriately addressed. Overall, this study makes a first step toward developing a platform pediatric physiologically based PK model for monoclonal antibodies and immunoglobulin G drug products by solidifying existing parameterizations, integrating new concepts, and drawing attention to unmet needs for physiologic knowledge in children.


Assuntos
Anticorpos Monoclonais/farmacocinética , Recém-Nascido Prematuro/fisiologia , Algoritmos , Capilares/fisiologia , Simulação por Computador , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Imunoglobulina G/metabolismo , Imunoglobulinas Intravenosas/farmacocinética , Recém-Nascido , Contagem de Leucócitos , Linfa/fisiologia , Modelos Biológicos , Receptores Fc/fisiologia , Software
14.
PLoS Biol ; 17(9): e3000436, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31498797

RESUMO

Bats harbor many zoonotic viruses, including highly pathogenic viruses of humans and other mammals, but they are typically asymptomatic in bats. To further understand the antiviral immunity of bats, we screened and identified a series of bat major histocompatibility complex (MHC) I Ptal-N*01:01-binding peptides derived from four different bat-borne viruses, i.e., Hendra virus (HeV), Ebola virus (EBOV), Middle East respiratory syndrome coronavirus (MERS-CoV), and H17N10 influenza-like virus. The structures of Ptal-N*01:01 display unusual peptide presentation features in that the bat-specific 3-amino acid (aa) insertion enables the tight "surface anchoring" of the P1-Asp in pocket A of bat MHC I. As the classical primary anchoring positions, the B and F pockets of Ptal-N*01:01 also show unconventional conformations, which contribute to unusual peptide motifs and distinct peptide presentation. Notably, the features of bat MHC I may be shared by MHC I from various marsupials. Our study sheds light on bat adaptive immunity and may benefit future vaccine development against bat-borne viruses of high impact on humans.


Assuntos
Apresentação de Antígeno , Quirópteros/imunologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Vírus de RNA/imunologia , Animais , Quirópteros/virologia
15.
J Cell Sci ; 133(5)2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31444284

RESUMO

The neonatal Fc receptor (FcRn) rescues albumin and IgG from degradation following endocytosis and thereby extends the half-life of these plasma proteins. However, the pathways for the uptake of these soluble FcRn ligands, and the recycling itinerary of the FcRn-ligand complexes, have not been identified in primary cells. Here, we have defined the recycling of human albumin and IgG in primary mouse macrophages selectively expressing the human FcRn. Albumin is internalised by macropinocytosis; in the absence of FcRn, internalised albumin is rapidly degraded, while in the presence of FcRn albumin colocalises to SNX5-positive membrane domains and is partitioned into tubules emanating from early macropinosomes for delivery in transport carriers to the plasma membrane. Soluble monomeric IgG was also internalised by macropinocytosis and rapidly recycled by the same pathway. In contrast, the fate of IgG bound to surface Fcγ receptors differed from monomeric IgG endocytosed by macropinocytosis. Overall, our findings identify a rapid recycling pathway for FcRn ligands from early macropinosomes to the cell surface of primary cells.


Assuntos
Albuminas/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Imunoglobulina G/metabolismo , Macrófagos/metabolismo , Pinocitose , Receptores Fc/fisiologia , Animais , Linhagem Celular , Endocitose , Endossomos/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Camundongos , Camundongos Knockout , Transporte Proteico , Receptores Fc/genética
16.
Nat Rev Immunol ; 19(10): 643-657, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31308521

RESUMO

Mucosal-associated invariant T (MAIT) cells are unique innate-like T cells that bridge innate and adaptive immunity. They are activated by conserved bacterial ligands derived from vitamin B biosynthesis and have important roles in defence against bacterial and viral infections. However, they can also have various deleterious and protective functions in autoimmune, inflammatory and metabolic diseases. MAIT cell involvement in a large spectrum of pathological conditions makes them attractive targets for potential therapeutic approaches.


Assuntos
Células T Invariantes Associadas à Mucosa/imunologia , Apoptose , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Infecções/imunologia , Inflamação/imunologia , Interleucina-17/fisiologia , Antígenos de Histocompatibilidade Menor/fisiologia , Neoplasias/imunologia
17.
Sci Rep ; 9(1): 7680, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31118488

RESUMO

The purpose of this study was to evaluate the prognostic impact of major histocompatibility complex (MHC) class I expression and programmed death-ligand 1 (PD-L1) expression in patients with head and neck squamous cell carcinoma (HNSCC). A total of 158 patients with HNSCC were evaluated retrospectively. The expression of MHC class I and PD-L1 was analyzed in tumor specimens using immunohistochemistry. The association between MHC class I/PD-L1 expression and clinical outcome was evaluated by Kaplan-Meier and Cox regression analyses. Among 158 patients, 103 (65.2%) showed positive PD-L1 expression, and 20 (12.7%) showed no detectable expression of MHC class I. The frequency of PD-L1 positive expression with concomitant MHC class I loss was 7.0%. In the PD-L1-positive group, MHC class I loss was associated with a significantly worse survival compared with MHC class I positivity (median overall survival 39.3 months vs. not reached; P = 0.005), whereas MHC class I status provided no prognostic impact in the PD-L1 negative group. Neither PD-L1 nor MHC class I alone showed a significant difference in overall survival. The loss of MHC class I expression in PD-L1-positive HNSCC was associated with a poor clinical outcome. This suggested that MHC class I expression status might be useful for the prognosis of tumor progression in HNSCC when combined with PD-L1 expression status. External validation with enough numbers of participants in such subgroup should be needed for validation.


Assuntos
Antígeno B7-H1/biossíntese , Antígenos de Histocompatibilidade Classe I/fisiologia , Proteínas de Neoplasias/biossíntese , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígeno B7-H1/genética , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes MHC Classe I , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Carcinoma de Células Escamosas de Cabeça e Pescoço/mortalidade , Adulto Jovem
18.
Cell ; 177(6): 1553-1565.e16, 2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31104841

RESUMO

Enterovirus B (EV-B), a major proportion of the genus Enterovirus in the family Picornaviridae, is the causative agent of severe human infectious diseases. Although cellular receptors for coxsackievirus B in EV-B have been identified, receptors mediating virus entry, especially the uncoating process of echovirus and other EV-B remain obscure. Here, we found that human neonatal Fc receptor (FcRn) is the uncoating receptor for major EV-B. FcRn binds to the virus particles in the "canyon" through its FCGRT subunit. By obtaining multiple cryo-electron microscopy structures at different stages of virus entry at atomic or near-atomic resolution, we deciphered the underlying mechanisms of enterovirus attachment and uncoating. These structures revealed that different from the attachment receptor CD55, binding of FcRn to the virions induces efficient release of "pocket factor" under acidic conditions and initiates the conformational changes in viral particle, providing a structural basis for understanding the mechanisms of enterovirus entry.


Assuntos
Enterovirus Humano B/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe I/ultraestrutura , Receptores Fc/metabolismo , Receptores Fc/ultraestrutura , Capsídeo/metabolismo , Microscopia Crioeletrônica , Enterovirus , Enterovirus Humano B/patogenicidade , Infecções por Enterovirus/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Modelos Moleculares , Filogenia , Receptores Fc/fisiologia , Vírion , Internalização do Vírus
19.
Cell ; 174(3): 672-687.e27, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30053426

RESUMO

TCR-signaling strength generally correlates with peptide-MHC binding affinity; however, exceptions exist. We find high-affinity, yet non-stimulatory, interactions occur with high frequency in the human T cell repertoire. Here, we studied human TCRs that are refractory to activation by pMHC ligands despite robust binding. Analysis of 3D affinity, 2D dwell time, and crystal structures of stimulatory versus non-stimulatory TCR-pMHC interactions failed to account for their different signaling outcomes. Using yeast pMHC display, we identified peptide agonists of a formerly non-responsive TCR. Single-molecule force measurements demonstrated the emergence of catch bonds in the activating TCR-pMHC interactions, correlating with exclusion of CD45 from the TCR-APC contact site. Molecular dynamics simulations of TCR-pMHC disengagement distinguished agonist from non-agonist ligands based on the acquisition of catch bonds within the TCR-pMHC interface. The isolation of catch bonds as a parameter mediating the coupling of TCR binding and signaling has important implications for TCR and antigen engineering for immunotherapy.


Assuntos
Antígenos de Histocompatibilidade Classe I/fisiologia , Ativação Linfocitária/fisiologia , Adulto , Feminino , Humanos , Cinética , Ligantes , Complexo Principal de Histocompatibilidade/fisiologia , Masculino , Pessoa de Meia-Idade , Simulação de Dinâmica Molecular , Oligopeptídeos , Peptídeos , Ligação Proteica/fisiologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais , Imagem Individual de Molécula , Linfócitos T/fisiologia
20.
Gastroenterology ; 155(5): 1366-1371.e3, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30031767

RESUMO

Killer-cell immunoglobulin-like receptors (KIRs) are transmembrane glycoproteins expressed by natural killer (NK) cells. Binding of KIR3DS1 to its recently discovered ligand, HLA-F, activates NK cells and has been associated with resolution of hepatitis C virus (HCV) infection. We investigated the mechanisms by which KIR3DS1 contributes to the antiviral immune response. Using cell culture systems, mice with humanized livers, and primary liver tissue from HCV-infected individuals, we found that the KIR3DS1 ligand HLA-F is up-regulated on HCV-infected cells, and that interactions between KIR3DS1 and HLA-F contribute to NK cell-mediated control of HCV. Strategies to promote interaction between KIR3DS1 and HLA-F might be developed for treatment of infectious diseases and cancer.


Assuntos
Hepacivirus/fisiologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Receptores KIR3DS1/fisiologia , Replicação Viral , Células Cultivadas , Hepatite C/tratamento farmacológico , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...