Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 290
Filtrar
1.
Nature ; 625(7995): 578-584, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38123677

RESUMEN

The symptoms of malaria occur during the blood stage of infection, when parasites invade and replicate within human erythrocytes. The PfPCRCR complex1, containing PfRH5 (refs. 2,3), PfCyRPA, PfRIPR, PfCSS and PfPTRAMP, is essential for erythrocyte invasion by the deadliest human malaria parasite, Plasmodium falciparum. Invasion can be prevented by antibodies3-6 or nanobodies1 against each of these conserved proteins, making them the leading blood-stage malaria vaccine candidates. However, little is known about how PfPCRCR functions during invasion. Here we present the structure of the PfRCR complex7,8, containing PfRH5, PfCyRPA and PfRIPR, determined by cryogenic-electron microscopy. We test the hypothesis that PfRH5 opens to insert into the membrane9, instead showing that a rigid, disulfide-locked PfRH5 can mediate efficient erythrocyte invasion. We show, through modelling and an erythrocyte-binding assay, that PfCyRPA-binding antibodies5 neutralize invasion through a steric mechanism. We determine the structure of PfRIPR, showing that it consists of an ordered, multidomain core flexibly linked to an elongated tail. We also show that the elongated tail of PfRIPR, which is the target of growth-neutralizing antibodies6, binds to the PfCSS-PfPTRAMP complex on the parasite membrane. A modular PfRIPR is therefore linked to the merozoite membrane through an elongated tail, and its structured core presents PfCyRPA and PfRH5 to interact with erythrocyte receptors. This provides fresh insight into the molecular mechanism of erythrocyte invasion and opens the way to new approaches in rational vaccine design.


Asunto(s)
Eritrocitos , Malaria Falciparum , Complejos Multiproteicos , Parásitos , Plasmodium falciparum , Proteínas Protozoarias , Animales , Humanos , Anticuerpos Neutralizantes/inmunología , Antígenos de Protozoos/química , Antígenos de Protozoos/inmunología , Microscopía por Crioelectrón , Disulfuros/química , Disulfuros/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Merozoítos/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Parásitos/metabolismo , Parásitos/patogenicidad , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/ultraestructura
2.
Cell Immunol ; 376: 104532, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35537322

RESUMEN

Autoimmune diseases constitute a heterogeneous group of disorders with one common feature - the loss of immune tolerance towards autoantigens. Due to the complexity of the pathogenesis of these diseases, there are still many open questions regarding their etiology. Therefore, scientists unceasingly search for new data hoping to detect dependable biomarkers and design safe and effective treatment. The research on immune checkpoints is in line with these scientific and clinical demands. Immune checkpoints may be the key to understanding the pathogenesis of many immunological disorders. BTLA-HVEM complex, the inhibitory immune checkpoint, has recently caught scientific attention as an important regulator in different immune contexts, including autoreactivity. So far, the BTLA-HVEM complex has been mainly studied in the context of cancer, but as numerous data show, it may also be a target in the treating of autoimmune diseases. In this review, we intend to focus on the mechanisms of BTLA-HVEM interactions in immune cells and summarize the available data in the context of autoimmunity.


Asunto(s)
Enfermedades Autoinmunes , Receptores Inmunológicos , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Autoinmunidad/inmunología , Humanos , Complejos Multiproteicos/inmunología , Receptores Inmunológicos/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología
3.
J Mol Biol ; 434(12): 167602, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35469831

RESUMEN

High performance affinity reagents are essential tools to enable biologists to profile the cellular location and composition of macromolecular complexes undergoing dynamic reorganization. To support further development of such tools, we have assembled a high-throughput phage display pipeline to generate Fab-based affinity reagents that target different dynamic forms of a large macromolecular complex, using the Chromosomal Passenger Complex (CPC), as an example. The CPC is critical for the maintenance of chromosomal and cytoskeleton processes during cell division. The complex contains 4 protein components: Aurora B kinase, survivin, borealin and INCENP. The CPC acts as a node to dynamically organize other partnering subcomplexes to build multiple functional structures during mitotic progression. Using phage display mutagenesis, a cohort of synthetic antibodies (sABs) were generated against different domains of survivin, borealin and INCENP. Immunofluorescence established that a set of these sABs can discriminate between the form of the CPC complex in the midbody versus the spindle. Others localize to targets, which appear to be less organized, in the nucleus or cytoplasm. This differentiation suggests that different CPC epitopes have dynamic accessibility depending upon the mitotic state of the cell. An Immunoprecipitation/Mass Spectrometry analysis was performed using sABs that bound specifically to the CPC in either the midbody or MT spindle macromolecular assemblies. Thus, sABs can be exploited as high performance reagents to profile the accessibility of different components of the CPC within macromolecular assemblies during different stages of mitosis suggesting this high throughput approach will be applicable to other complex macromolecular systems.


Asunto(s)
Anticuerpos , Aurora Quinasa B , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Fragmentos Fab de Inmunoglobulinas , Complejos Multiproteicos , Survivin , Anticuerpos/química , Anticuerpos/genética , Aurora Quinasa B/análisis , Aurora Quinasa B/inmunología , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/inmunología , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/inmunología , Citoesqueleto/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Mitosis , Complejos Multiproteicos/análisis , Complejos Multiproteicos/inmunología , Biblioteca de Péptidos , Fosforilación , Huso Acromático/metabolismo , Survivin/química , Survivin/metabolismo
4.
J Biol Chem ; 297(4): 101102, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34419446

RESUMEN

CD27 is a tumor necrosis factor (TNF) receptor, which stimulates lymphocytes and promotes their differentiation upon activation by TNF ligand CD70. Activation of the CD27 receptor provides a costimulatory signal to promote T cell, B cell, and NK cell activity to facilitate antitumor and anti-infection immunity. Aberrant increased and focused expression of CD70 on many tumor cells renders CD70 an attractive therapeutic target for direct tumor killing. However, despite their use as drug targets to treat cancers, the molecular basis and atomic details of CD27 and CD70 interaction remain elusive. Here we report the crystal structure of human CD27 in complex with human CD70. Analysis of our structure shows that CD70 adopts a classical TNF ligand homotrimeric assembly to engage CD27 receptors in a 3:3 stoichiometry. By combining structural and rational mutagenesis data with reported disease-correlated mutations, we identified the key amino acid residues of CD27 and CD70 that control this interaction. We also report increased potency for plate-bound CD70 constructs compared with solution-phase ligand in a functional activity to stimulate T-cells in vitro. These findings offer new mechanistic insight into this critical costimulatory interaction.


Asunto(s)
Ligando CD27/química , Complejos Multiproteicos/química , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/química , Ligando CD27/genética , Ligando CD27/inmunología , Cristalografía por Rayos X , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Estructura Cuaternaria de Proteína , Linfocitos T/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
5.
J Clin Invest ; 131(16)2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34185706

RESUMEN

TNFR1 and TNFR2 have received prominent attention because of their dominance in the pathogenesis of inflammation and autoimmunity. TNFR1 has been extensively studied and primarily mediates inflammation. TNFR2 remains far less studied, although emerging evidence demonstrates that TNFR2 plays an antiinflammatory and immunoregulatory role in various conditions and diseases. Herein, we report that TNFR2 regulates macrophage polarization, a highly dynamic process controlled by largely unidentified intracellular regulators. Using biochemical copurification and mass spectrometry approaches, we isolated the signaling molecule 14-3-3ε as a component of TNFR2 complexes in response to progranulin stimulation in macrophages. In addition, 14-3-3ε was essential for TNFR2 signaling-mediated regulation of macrophage polarization and switch. Both global and myeloid-specific deletion of 14-3-3ε resulted in exacerbated inflammatory arthritis and counteracted the protective effects of progranulin-mediated TNFR2 activation against inflammation and autoimmunity. TNFR2/14-3-3ε signaled through PI3K/Akt/mTOR to restrict NF-κB activation while simultaneously stimulating C/EBPß activation, thereby instructing macrophage plasticity. Collectively, this study identifies 14-3-3ε as a previously unrecognized vital component of the TNFR2 receptor complex and provides new insights into the TNFR2 signaling, particularly its role in macrophage polarization with therapeutic implications for various inflammatory and autoimmune diseases with activation of the TNFR2/14-3-3ε antiinflammatory pathway.


Asunto(s)
Proteínas 14-3-3/inmunología , Macrófagos/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Proteínas 14-3-3/química , Proteínas 14-3-3/deficiencia , Proteínas 14-3-3/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Autoinmunidad , Humanos , Inflamación/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Complejos Multiproteicos/química , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Progranulinas/inmunología , Progranulinas/metabolismo , Células RAW 264.7 , Receptores Tipo II del Factor de Necrosis Tumoral/química , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/inmunología
6.
Med Oncol ; 38(6): 69, 2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34002296

RESUMEN

Transcription factor CCAAT/enhancer-binding protein-alpha (C/EBPα) regulates myelopoiesis by coupling growth arrest with differentiation of myeloid progenitors. Mutations in one or both alleles are observed in 10-14% AML cases that render C/EBPα functionally inactive. Besides, antagonistic protein-protein interactions also impair C/EBPα expression and function. In recent independent studies, we showed that CDK2 and SKP2 downregulated C/EBPα expression in an ubiquitin-dependent proteasome degradation manner leading to differentiation block in AML. Here, we demonstrate that CDK2-instigated C/EBPα downregulation is actually mediated by SKP2. Mechanistically, we show that CDK2 stabilizes SKP2 by phosphorylating it at Ser64 and thereby potentiates C/EBPα ubiquitination and subsequent degradation in AML cells. Immunoblot experiments showed that CDK2 inhibition downregulated SKP2 levels and concomitantly enhanced C/EBPα levels in myeloid cells. We further show that while CDK2 promoted C/EBPα ubiquitination and inhibited its protein levels, negatively affected its transactivation potential and DNA binding ability, simultaneous SKP2 depletion abrogated CDK2-promoted ubiquitination and restored C/EBPα expression and function. Taken together, these findings consolidate that CDK2 potentiates SKP2-mediated C/EBPα degradation in AML and targeting CDK2-SKP2 axis can be harnessed for therapeutic benefit in AML. Hypothetical model depicts that SKP2-mediated C/EBPα proteasomal degradation is reinforced by CDK2. CDK2 phopshorylates SKP2 leading to its enhanced stabilization which in turn exaggerates C/EBPα degradation leading to differentiation arrest in AML.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/inmunología , Regulación hacia Abajo , Células HEK293 , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Complejos Multiproteicos/inmunología , Fosforilación , Estabilidad Proteica , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/inmunología , Serina/metabolismo
7.
Protein Cell ; 12(9): 680-694, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33835418

RESUMEN

Signaling pathways in innate and adaptive immunity play vital roles in pathogen recognition and the functions of immune cells. Higher-order assemblies have recently emerged as a central principle that governs immune signaling and, by extension, cellular communication in general. There are mainly two types of higher-order assemblies: 1) ordered, solid-like large supramolecular complexes formed by stable and rigid protein-protein interactions, and 2) liquid-like phase-separated condensates formed by weaker and more dynamic intermolecular interactions. This review covers key examples of both types of higher-order assemblies in major immune pathways. By placing emphasis on the molecular structures of the examples provided, we discuss how their structural organization enables elegant mechanisms of signaling regulation.


Asunto(s)
Inmunidad Innata , Inflamasomas/inmunología , Complejos Multiproteicos/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Inmunidad Adaptativa , Animales , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Proteína 58 DEAD Box/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamasomas/genética , Inflamasomas/ultraestructura , Modelos Moleculares , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Conformación Proteica , Mapeo de Interacción de Proteínas , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
8.
Front Immunol ; 12: 631271, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763075

RESUMEN

Phosphoinositide 3-kinases (PI3Ks) play a central role in adaptive immunity by transducing signals from the T cell antigen receptor (TCR) via production of PIP3. PI3Kδ is a heterodimer composed of a p110δ catalytic subunit associated with a p85α or p85ß regulatory subunit and is preferentially engaged by the TCR upon T cell activation. The molecular mechanisms leading to PI3Kδ recruitment and activation at the TCR signalosome remain unclear. In this study, we have used quantitative mass spectrometry, biochemical approaches and CRISPR-Cas9 gene editing to uncover the p110δ interactome in primary CD4+ T cells. Moreover, we have determined how the PI3Kδ interactome changes upon the differentiation of small naïve T cells into T cell blasts expanded in the presence of IL-2. Our interactomic analyses identified multiple constitutive and inducible PI3Kδ-interacting proteins, some of which were common to naïve and previously-activated T cells. Our data reveals that PI3Kδ rapidly interacts with as many as seven adaptor proteins upon TCR engagement, including the Gab-family proteins, GAB2 and GAB3, a CD5-CBL signalosome and the transmembrane proteins ICOS and TRIM. Our results also suggest that PI3Kδ pre-forms complexes with the adaptors SH3KBP1 and CRKL in resting cells that could facilitate the localization and activation of p110δ at the plasma membrane by forming ternary complexes during early TCR signalling. Furthermore, we identify interactions that were not previously known to occur in CD4+ T cells, involving BCAP, GAB3, IQGAP3 and JAML. We used CRISPR-Cas9-mediated gene knockout in primary T cells to confirm that BCAP is a positive regulator of PI3K-AKT signalling in CD4+ T cell blasts. Overall, our results provide evidence for a large protein network that regulates the recruitment and activation of PI3Kδ in T cells. Finally, this work shows how the PI3Kδ interactome is remodeled as CD4+ T cells differentiate from naïve T cells to activated T cell blasts. These activated T cells upregulate additional PI3Kδ adaptor proteins, including BCAP, GAB2, IQGAP3 and ICOS. This rewiring of TCR-PI3K signalling that occurs upon T cell differentiation may serve to reduce the threshold of activation and diversify the inputs for the PI3K pathway in effector T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/inmunología , Complejos Multiproteicos/biosíntesis , Complejos Multiproteicos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/clasificación , Linfocitos T CD4-Positivos/efectos de los fármacos , Sistemas CRISPR-Cas , Edición Génica , Técnicas de Inactivación de Genes , Interleucina-2/farmacología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal , Organismos Libres de Patógenos Específicos
9.
Cell Rep ; 34(6): 108742, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33567280

RESUMEN

Glucocorticoids (GCs) are effective anti-inflammatory drugs; yet, their mechanisms of action are poorly understood. GCs bind to the glucocorticoid receptor (GR), a ligand-gated transcription factor controlling gene expression in numerous cell types. Here, we characterize GR's protein interactome and find the SETD1A (SET domain containing 1A)/COMPASS (complex of proteins associated with Set1) histone H3 lysine 4 (H3K4) methyltransferase complex highly enriched in activated mouse macrophages. We show that SETD1A/COMPASS is recruited by GR to specific cis-regulatory elements, coinciding with H3K4 methylation dynamics at subsets of sites, upon treatment with lipopolysaccharide (LPS) and GCs. By chromatin immunoprecipitation sequencing (ChIP-seq) and RNA-seq, we identify subsets of GR target loci that display SETD1A occupancy, H3K4 mono-, di-, or tri-methylation patterns, and transcriptional changes. However, our data on methylation status and COMPASS recruitment suggest that SETD1A has additional transcriptional functions. Setd1a loss-of-function studies reveal that SETD1A/COMPASS is required for GR-controlled transcription of subsets of macrophage target genes. We demonstrate that the SETD1A/COMPASS complex cooperates with GR to mediate anti-inflammatory effects.


Asunto(s)
Elementos de Facilitación Genéticos/inmunología , Macrófagos/inmunología , Complejos Multiproteicos , RNA-Seq , Receptores de Glucocorticoides , Transcripción Genética/inmunología , Animales , Inflamación/genética , Inflamación/inmunología , Ratones , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/inmunología
10.
J Clin Invest ; 131(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33444292

RESUMEN

DREAM (Dp, Rb-like, E2F, and MuvB) is a transcriptional repressor complex that regulates cell proliferation, and its loss causes neonatal lethality in mice. To investigate DREAM function in adult mice, we used an assembly-defective p107 protein and conditional deletion of its redundant family member p130. In the absence of DREAM assembly, mice displayed shortened survival characterized by systemic amyloidosis but no evidence of excessive cellular proliferation. Amyloid deposits were found in the heart, liver, spleen, and kidneys but not the brain or bone marrow. Using laser-capture microdissection followed by mass spectrometry, we identified apolipoproteins as the most abundant components of amyloids. Intriguingly, apoA-IV was the most detected amyloidogenic protein in amyloid deposits, suggesting apoA-IV amyloidosis (AApoAIV). AApoAIV is a recently described form, whereby WT apoA-IV has been shown to predominate in amyloid plaques. We determined by ChIP that DREAM directly regulated Apoa4 and that the histone variant H2AZ was reduced from the Apoa4 gene body in DREAM's absence, leading to overexpression. Collectively, we describe a mechanism by which epigenetic misregulation causes apolipoprotein overexpression and amyloidosis, potentially explaining the origins of nongenetic amyloid subtypes.


Asunto(s)
Amiloide/metabolismo , Apolipoproteínas A/metabolismo , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas/metabolismo , Complejos Multiproteicos/inmunología , Proteína p107 Similar a la del Retinoblastoma/deficiencia , Amiloide/genética , Animales , Apolipoproteínas A/genética , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas/genética , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas/patología , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Especificidad de Órganos/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo
11.
Biomolecules ; 10(11)2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33143355

RESUMEN

Anti-DNA antibodies are usually produced against histone-DNA complexes appearing during cell apoptosis, while histones are known as damage-associated molecules. A myelin sheath of axons contains myelin basic protein (MBP) playing an important role in the pathogenesis of autoimmune diseases. Antibodies with enzymatic activities (abzymes) are distinctive features of some autoimmune and viral diseases. Abzymes against different proteins can usually only hydrolyze these specific proteins. Using sequential chromatographies of homogeneous IgG preparations from sera of HIV-infected patients on columns with immobilized MBP, H2a, and H2b histones, the anti-MBP, anti-H2a, and anti-H2b antibodies were obtained. It was first shown that IgGs against H2a and H2b effectively hydrolyze these histones and MBP, while anti-MBP split MBP, H2a, and H2b, but no other control proteins. Using the MALDI mass spectrometry, the cleavage sites of H2a, H2b, and MBP by abzymes against these three proteins were found. Among 14 sites of hydrolysis of H2a by IgGs against H2a and 10 sites by anti-MBP IgGs, only one site of hydrolysis was the same for these abzymes. Eleven cleavage sites of H2b with IgGs against H2b and 10 sites of its hydrolysis with antibodies against MBP were different. Anti-H2a, anti-H2b, and anti-MBP abzymes are unpredictable examples of IgGs possessing not only cross-complexation but also catalytic cross-reactivity, which may be a common phenomenon for such abzymes in patients with different autoimmune diseases. The existence of cross-reactivity of abzymes against H2a and H2b histones and MBP represent a great danger to humans since, in contrast with MBP, histones due to cell apoptosis constantly occur in human blood. Anti-H2a, anti-H2b, and anti-MBP can attack and hydrolyze myelin basic protein of the myelin sheath of axons and plays a negative role in the pathogenesis of several pathologies.


Asunto(s)
Infecciones por VIH/inmunología , Histonas/inmunología , Complejos Multiproteicos/inmunología , Proteína Básica de Mielina/inmunología , Anticuerpos Antinucleares/sangre , Anticuerpos Antinucleares/genética , Anticuerpos Antinucleares/inmunología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , VIH/patogenicidad , Infecciones por VIH/sangre , Infecciones por VIH/genética , Histonas/sangre , Humanos , Hidrólisis , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Complejos Multiproteicos/sangre , Proteína Básica de Mielina/sangre , Proteolisis , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
12.
Molecules ; 25(20)2020 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-33080923

RESUMEN

Electrospray mass spectrometry is applied to determine apparent binding energies and quasi equilibrium dissociation constants of immune complex dissociation reactions in the gas phase. Myoglobin, a natural protein-ligand complex, has been used to develop the procedure which starts from determining mean charge states and normalized and averaged ion intensities. The apparent dissociation constant KD m0g#= 3.60 × 10-12 for the gas phase heme dissociation process was calculated from the mass spectrometry data and by subsequent extrapolation to room temperature to mimic collision conditions for neutral and resting myoglobin. Similarly, for RNAse S dissociation at room temperature a KD m0g#= 4.03 × 10-12 was determined. The protocol was tested with two immune complexes consisting of epitope peptides and monoclonal antibodies. For the epitope peptide dissociation reaction of the FLAG peptide from the antiFLAG antibody complex an apparent gas phase dissociation constant KD m0g#= 4.04 × 10-12 was calculated. Likewise, an apparent KD m0g#= 4.58 × 10-12 was calculated for the troponin I epitope peptide-antiTroponin I antibody immune complex dissociation. Electrospray mass spectrometry is a rapid method, which requires small sample amounts for either identification of protein-bound ligands or for determination of the apparent gas phase protein-ligand complex binding strengths.


Asunto(s)
Complejo Antígeno-Anticuerpo/química , Epítopos/química , Complejos Multiproteicos/química , Mioglobina/química , Anticuerpos/química , Anticuerpos/inmunología , Complejo Antígeno-Anticuerpo/genética , Complejo Antígeno-Anticuerpo/inmunología , Epítopos/inmunología , Hemo/química , Hemo/inmunología , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Ligandos , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Mioglobina/genética , Mioglobina/inmunología , Oligopéptidos/química , Oligopéptidos/genética , Oligopéptidos/inmunología , Péptidos/química , Péptidos/inmunología , Ribonucleasas/química , Ribonucleasas/inmunología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
J Immunol ; 204(5): 1085-1090, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969387

RESUMEN

Lymphotoxin ß receptor (LTßR) signaling is crucial for lymphoid tissue organogenesis and immune homeostasis. To identify novel regulatory mechanisms for signaling, we implemented a two-step screen that uses coexpression analysis of human fibroblasts undergoing LTßR stimulation and affinity-purification mass spectrometry for the LTßR signaling protein TNFR-associated factor 3 (TRAF3). We identify Ewing sarcoma (EWS) protein as a novel LTßR signaling component that associates with TRAF3 but not with TNFR-associated factor 2 (TRAF2). The EWS:TRAF3 complex forms under unligated conditions that are disrupted following activation of the LTßR. We conclude that EWS limits expression of proinflammatory molecules, GM-CSF, and ERK-2, promoting immune homeostasis.


Asunto(s)
Receptor beta de Linfotoxina/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Complejos Multiproteicos/inmunología , Proteína EWS de Unión a ARN/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Células HEK293 , Humanos , Receptor beta de Linfotoxina/genética , Sistema de Señalización de MAP Quinasas/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Complejos Multiproteicos/genética , Proteína EWS de Unión a ARN/genética , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/inmunología , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/inmunología
14.
J Clin Invest ; 130(4): 1830-1842, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31917688

RESUMEN

Foxp3+ Tregs are key to immune homeostasis, but the contributions of various large, multiprotein complexes that regulate gene expression remain unexplored. We analyzed the role in Tregs of the evolutionarily conserved CoREST complex, consisting of a scaffolding protein, Rcor1 or Rcor2, plus Hdac1 or Hdac2 and Lsd1 enzymes. Rcor1, Rcor2, and Lsd1 were physically associated with Foxp3, and mice with conditional deletion of Rcor1 in Foxp3+ Tregs had decreased proportions of Tregs in peripheral lymphoid tissues and increased Treg expression of IL-2 and IFN-γ compared with what was found in WT cells. Mice with conditional deletion of the gene encoding Rcor1 in their Tregs had reduced suppression of homeostatic proliferation, inability to maintain long-term allograft survival despite costimulation blockade, and enhanced antitumor immunity in syngeneic models. Comparable findings were seen in WT mice treated with CoREST complex bivalent inhibitors, which also altered the phenotype of human Tregs and impaired their suppressive function. Our data point to the potential for therapeutic modulation of Treg functions by pharmacologic targeting of enzymatic components of the CoREST complex and contribute to an understanding of the biochemical and molecular mechanisms by which Foxp3 represses large gene sets and maintains the unique properties of this key immune cell.


Asunto(s)
Proteínas Co-Represoras/inmunología , Inmunidad Celular , Complejos Multiproteicos/inmunología , Neoplasias Experimentales/inmunología , Proteínas del Tejido Nervioso/inmunología , Linfocitos T Reguladores/inmunología , Animales , Línea Celular Tumoral , Proteínas Co-Represoras/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Complejos Multiproteicos/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas del Tejido Nervioso/genética , Linfocitos T Reguladores/patología
15.
J Clin Invest ; 130(3): 1301-1314, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31714898

RESUMEN

Influenza A virus (IAV) is among the most common causes of pneumonia-related death worldwide. Pulmonary epithelial cells are the primary target for viral infection and replication and respond by releasing inflammatory mediators that recruit immune cells to mount the host response. Severe lung injury and death during IAV infection result from an exuberant host inflammatory response. The linear ubiquitin assembly complex (LUBAC), composed of SHARPIN, HOIL-1L, and HOIP, is a critical regulator of NF-κB-dependent inflammation. Using mice with lung epithelial-specific deletions of HOIL-1L or HOIP in a model of IAV infection, we provided evidence that, while a reduction in the inflammatory response was beneficial, ablation of the LUBAC-dependent lung epithelial-driven response worsened lung injury and increased mortality. Moreover, we described a mechanism for the upregulation of HOIL-1L in infected and noninfected cells triggered by the activation of type I IFN receptor and mediated by IRF1, which was maladaptive and contributed to hyperinflammation. Thus, we propose that lung epithelial LUBAC acts as a molecular rheostat that could be selectively targeted to modulate the immune response in patients with severe IAV-induced pneumonia.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Complejos Multiproteicos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Viral/inmunología , Mucosa Respiratoria/inmunología , Células A549 , Animales , Perros , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Pulmón/patología , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/genética , Neumonía Viral/patología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/inmunología
16.
BMC Immunol ; 20(1): 48, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31842739

RESUMEN

BACKGROUND: Yersinia pestis, the etiological pathogen of plague, is capable of repressing the immune response of white blood cells to evade phagocytosis. The V-antigen (LcrV) was found to be involved in this process by binding to human Toll-like Receptor 2 (TLR2). The detailed mechanism behind this LcrV and TLR2 mediated immune response repression, however, is yet to be fully elucidated due to the lack of structural information. RESULTS: In this work, with protein structure modelling, we were able to construct a structure model of the heterotetramer of Y. pestis LcrV and human TLR2. Molecular dynamics simulation suggests the stability of this structure in aquatic environment. The LcrV model has a dumbbell-like structure with two globule domains (G1 at N-terminus and G2 away from membrane) connected with a coiled-coil linker (CCL) domain. The two horseshoe-shape TLR2 subunits form a V-shape structure, are not in direct contact with each other, and are held together by the LcrV homodimer. In this structure model, both the G1 and CCL domains are involved in the formation of LcrV homodimer, while all three domains are involved in LcrV-TLR2 binding. A mechanistic model was proposed based on this heterotetrameric structure model: The LcrV homodimer separates the TLR2 subunits to inhibit the dimerization of TLR2 and subsequent signal transfer for immune response; while LcrV could also inhibit the formation of heterodimers of TLR2 with other TLRs, and leads to immune response repression. CONCLUSIONS: A heterotetrameric structure of Y. pestis LcrV and human TLR2 was modelled in this work. Analysis of this modelled structure showed its stability in aquatic environments and the role of LcrV domains and residues in protein-protein interaction. A mechanistic model for the role of LcrV in Y. pestis pathogenesis is raised based on this heterotetrameric structure model. This work provides a hypothesis of LcrV function, with which further experimental validation may elucidate the role of LcrV in human immune response repression.


Asunto(s)
Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Complejos Multiproteicos/química , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/inmunología , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptor Toll-Like 2/química , Receptor Toll-Like 2/metabolismo , Dominio Catalítico , Interacciones Huésped-Patógeno/inmunología , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Conformación Molecular , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Relación Estructura-Actividad
17.
Protein Eng Des Sel ; 32(5): 219-230, 2019 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-31769480

RESUMEN

Yeast display immunoprecipitation is a combinatorial library screening platform for the discovery and engineering of antibodies against membrane proteins using detergent-solubilized membrane fractions or cell lysates as antigen sources. Here, we present the extension of this method for the screening of antibodies that bind to membrane protein complexes, enabling discovery of antibodies that target antigens involved in a functional protein-protein interaction of interest. For this proof-of-concept study, we focused on the receptor-mediated endocytosis machinery at the blood-brain barrier, and adaptin 2 (AP-2) was chosen as the functional interaction hub. The goal of this study was to identify antibodies that bound to blood-brain barrier (BBB) membrane protein complexes containing AP-2. Screening of a nonimmune yeast display antibody library was carried out using detergent-solubilized BBB plasma membranes as an antigen pool, and antibodies that could interact with protein complexes containing AP-2 were identified. Downstream characterization of isolated antibodies confirmed targeting of proteins known to play important roles in membrane trafficking. This functional yeast display immunoprecipitation screen may be applied to other systems where antibodies against other functional classes of protein complexes are sought.


Asunto(s)
Antígenos/inmunología , Barrera Hematoencefálica/inmunología , Proteínas de la Membrana/inmunología , Complejos Multiproteicos/inmunología , Saccharomyces cerevisiae , Anticuerpos de Cadena Única , Animales , Células HEK293 , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología
18.
Nat Commun ; 10(1): 4996, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676803

RESUMEN

Plants deploy various immune receptors to recognize pathogens and defend themselves. Crosstalk may happen among receptor-mediated signal transduction pathways in the same host during simultaneous infection of different pathogens. However, the related function of the receptor-like kinases (RLKs) in thwarting different pathogens remains elusive. Here, we report that NIK1, which positively regulates plant antiviral immunity, acts as an important negative regulator of antibacterial immunity. nik1 plants exhibit dwarfed morphology, enhanced disease resistance to bacteria and increased PAMP-triggered immunity (PTI) responses, which are restored by NIK1 reintroduction. Additionally, NIK1 negatively regulates the formation of the FLS2/BAK1 complex. The interaction between NIK1 and FLS2/BAK1 is enhanced upon flg22 perception, revealing a novel PTI regulatory mechanism by an RLK. Furthermore, flg22 perception induces NIK1 and RPL10A phosphorylation in vivo, activating antiviral signalling. The NIK1-mediated inverse modulation of antiviral and antibacterial immunity may allow bacteria and viruses to activate host immune responses against each other.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Inmunidad de la Planta/genética , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Arabidopsis/microbiología , Arabidopsis/virología , Proteínas de Arabidopsis/inmunología , Proteínas de Arabidopsis/metabolismo , Regulación de la Expresión Génica de las Plantas/inmunología , Interacciones Huésped-Patógeno/inmunología , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/virología , Inmunidad de la Planta/inmunología , Virus de Plantas/inmunología , Virus de Plantas/fisiología , Plantas Modificadas Genéticamente , Unión Proteica , Proteínas Quinasas/inmunología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Pseudomonas syringae/inmunología , Pseudomonas syringae/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología
19.
J Mol Biol ; 431(24): 4941-4958, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31704286

RESUMEN

The coreceptor CD8αß can greatly promote activation of T cells by strengthening T-cell receptor (TCR) binding to cognate peptide-MHC complexes (pMHC) on antigen presenting cells and by bringing p56Lck to TCR/CD3. Here, we demonstrate that CD8 can also bind to pMHC on the T cell (in cis) and that this inhibits their activation. Using molecular modeling, fluorescence resonance energy transfer experiments on living cells, biochemical and mutational analysis, we show that CD8 binding to pMHC in cis involves a different docking mode and is regulated by posttranslational modifications including a membrane-distal interchain disulfide bond and negatively charged O-linked glycans near positively charged sequences on the CD8ß stalk. These modifications distort the stalk, thus favoring CD8 binding to pMHC in cis. Differential binding of CD8 to pMHC in cis or trans is a means to regulate CD8+ T-cell responses and provides new translational opportunities.


Asunto(s)
Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Antígenos de Histocompatibilidad/química , Antígenos de Histocompatibilidad/inmunología , Complejos Multiproteicos/metabolismo , Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD8/química , Antígenos CD8/genética , Antígenos de Histocompatibilidad/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Modelos Biológicos , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/inmunología , Mutación , Péptidos/química , Péptidos/inmunología , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad
20.
Clin Exp Immunol ; 198(3): 341-350, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31397881

RESUMEN

Acquired angioedema due to C1-inhibitor (C1INH) deficiency (AAE) is caused by secondary C1INH deficiency leading to bradykinin-mediated angioedema episodes. AAE typically presents in adulthood and is associated with B cell lymphoproliferation. Anti-C1INH autoantibodies (antiC1INHAbs) are detectable in a subset of AAE cases and considered a hallmark of the disease. When free antiC1INHAbs and malignant tumors are not detectable, diagnosis relies on the finding of low C1INH levels and/or function, lack of family history and SERPING1 mutations, age at onset and low or undetectable C1q levels, none of which is specific for AAE. We tested the diagnostic value of a novel enzyme-linked immunosorbent assay (ELISA) for the detection of circulating complexes between C1INH and antiC1INHAbs (C1INH-antiC1INHAb) in the serum of 20 European AAE patients characterized on the basis of their complement levels and function. Free antiC1INHAbs were detected in nine of 20 patients [six of immunoglobulin (Ig)G class, two of IgM class and one simultaneously presenting IgG and IgM classes], whereas C1INH-antiC1INHAb complexes were found in 18 of 20 of the AAE cases, regardless of the presence or absence of detectable free anti-C1INHAbs. Of note, nine of 20 patients showed negative free antiC1INHabs, but positive C1INH-antiC1INHAb complexes in their first measurement. In the cohort presented, IgM-class C1INH-antiC1INHAb are specifically and strongly associated with low C1q serum levels. Detection of C1INH-antiC1-INHAbs provides an added value for AAE diagnosis, especially in those cases in whom no free anti-C1INH antibodies are detected. The link between IgM-class C1INH-antiC1INHAb complexes and C1q consumption could have further implications for the development of autoimmune manifestations in AAE.


Asunto(s)
Angioedema/inmunología , Angioedemas Hereditarios/inmunología , Autoanticuerpos/inmunología , Proteína Inhibidora del Complemento C1/inmunología , Complejos Multiproteicos/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Angioedema/sangre , Angioedema/diagnóstico , Angioedemas Hereditarios/sangre , Angioedemas Hereditarios/diagnóstico , Autoanticuerpos/sangre , Autoanticuerpos/metabolismo , Estudios de Cohortes , Proteína Inhibidora del Complemento C1/genética , Proteína Inhibidora del Complemento C1/metabolismo , Complemento C1q/inmunología , Complemento C1q/metabolismo , Ensayo de Inmunoadsorción Enzimática , Europa (Continente) , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complejos Multiproteicos/sangre , Complejos Multiproteicos/metabolismo , Mutación , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA