Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34108245

RESUMEN

Patients with severe COVID-19 infection exhibit a low level of oxygen in affected tissue and blood. To understand the pathophysiology of COVID-19 infection, it is therefore necessary to understand cell function during hypoxia. We investigated aspects of human monocyte activation under hypoxic conditions. HMGB1 is an alarmin released by stressed cells. Under normoxic conditions, HMGB1 activates interferon regulatory factor (IRF)5 and nuclear factor-κB in monocytes, leading to expression of type I interferon (IFN) and inflammatory cytokines including tumor necrosis factor α, and interleukin 1ß, respectively. When hypoxic monocytes are activated by HMGB1, they produce proinflammatory cytokines but fail to produce type I IFN. Hypoxia-inducible factor-1α, induced by hypoxia, functions as a direct transcriptional repressor of IRF5 and IRF3. As hypoxia is a stressor that induces secretion of HMGB1 by epithelial cells, hypoxia establishes a microenvironment that favors monocyte production of inflammatory cytokines but not IFN. These findings have implications for the pathogenesis of COVID-19.


Asunto(s)
Hipoxia de la Célula/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Monocitos/inmunología , COVID-19/inmunología , Células Cultivadas , Citocinas/inmunología , Humanos , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interleucina-1beta/metabolismo , Monocitos/metabolismo , FN-kappa B/inmunología , FN-kappa B/metabolismo , Oxígeno/metabolismo , SARS-CoV-2/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
2.
Clin Immunol ; 250: 109317, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37015317

RESUMEN

The receptor for advanced glycation end-products (RAGE) is a pattern recognition receptor that regulates inflammation, cell migration, and cell fate. Systemic lupus erythematosus (SLE) is a chronic multiorgan autoimmune disease. To understand the function of RAGE in SLE, we generated RAGE-deficient (Ager-/-) lupus-prone mice by backcrossing MRL/MpJ-Faslpr/J (MRL-lpr) mice with Ager-/- C57BL/6 mice. In 18-week-old Ager-/- MRL-lpr, the weights of the spleen and lymph nodes, as well as the frequency of CD3+CD4-CD8- cells, were significantly decreased. Ager-/- MRL-lpr mice had significantly reduced urine albumin/creatinine ratios and markedly improved renal pathological scores. Moreover, neutrophil infiltration and neutrophil extracellular trap formation in the glomerulus were significantly reduced in Ager-/- MRL-lpr. Our study is the first to reveal that RAGE can have a pathologic role in immune cells, particularly neutrophils and T cells, in inflammatory tissues and suggests that the inhibition of RAGE may be a potential therapeutic strategy for SLE.


Asunto(s)
Trampas Extracelulares , Lupus Eritematoso Sistémico , Nefritis Lúpica , Ratones , Animales , Receptor para Productos Finales de Glicación Avanzada/genética , Reacción de Maillard , Ratones Endogámicos MRL lpr , Ratones Endogámicos C57BL
3.
Proc Natl Acad Sci U S A ; 116(46): 23254-23263, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31570601

RESUMEN

Macrophage polarization is critical to inflammation and resolution of inflammation. We previously showed that high-mobility group box 1 (HMGB1) can engage receptor for advanced glycation end product (RAGE) to direct monocytes to a proinflammatory phenotype characterized by production of type 1 IFN and proinflammatory cytokines. In contrast, HMGB1 plus C1q form a tetramolecular complex cross-linking RAGE and LAIR-1 and directing monocytes to an antiinflammatory phenotype. Lipid mediators, as well as cytokines, help establish a milieu favoring either inflammation or resolution of inflammation. This study focuses on the induction of lipid mediators by HMGB1 and HMGB1 plus C1q and their regulation of IRF5, a transcription factor critical for the induction and maintenance of proinflammatory macrophages. Here, we show that HMGB1 induces leukotriene production through a RAGE-dependent pathway, while HMGB1 plus C1q induces specialized proresolving lipid mediators lipoxin A4, resolvin D1, and resolvin D2 through a RAGE- and LAIR-1-dependent pathway. Leukotriene exposure contributes to induction of IRF5 in a positive-feedback loop. In contrast, resolvins (at 20 nM) block IRF5 induction and prevent the differentiation of inflammatory macrophages. Finally, we have generated a molecular mimic of HMGB1 plus C1q, which cross-links RAGE and LAIR-1 and polarizes monocytes to an antiinflammatory phenotype. These findings may provide a mechanism to control nonresolving inflammation in many pathologic conditions.


Asunto(s)
Complemento C1q/metabolismo , Proteína HMGB1/metabolismo , Macrófagos/fisiología , Animales , Araquidonato 5-Lipooxigenasa/metabolismo , Factores Reguladores del Interferón/metabolismo , Leucotrieno B4/biosíntesis , Ratones Endogámicos C57BL , Monocitos/metabolismo , Peritonitis/inducido químicamente , Peritonitis/inmunología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptores Inmunológicos/metabolismo
4.
Mol Med ; 27(1): 125, 2021 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-34602056

RESUMEN

BACKGROUND: C1q has been reported to reveal complement-independent roles in immune and non-immune cells. C1q binds to its specific receptors to regulate distinct functions that rely on the environment and cell types. Discoidin domain receptor 2 (DDR2) is activated by collagen and functions in wound healing by controlling matrix metalloproteinase (MMP) expression. Since C1q exhibits a collagen-like structure, we hypothesized that C1q might engage DDR2 to regulate wound healing and extracellular matrix (ECM) remodeling. METHODS: Cell-based assay, proximity ligation assay, ELISA, and surface plasmon analysis were utilized to investigate DDR2 and C1q binding. We also investigate the C1q-mediated in vitro wound healing ability using the human fibrosarcoma cell line, HT1080. RESULTS: C1q induced the phosphorylation of DDR2, p38 kinase, and ERK1/2. C1q and DDR2 binding improved cell migration and induced MMP2 and MMP9 expression. DDR2-specific shRNA reduced C1q-mediated cell migration for wound healing. CONCLUSIONS: C1q is a new DDR2 ligand that promotes wound healing. These findings have therapeutic implications in wound healing-related diseases.


Asunto(s)
Movimiento Celular/fisiología , Colágeno/metabolismo , Complemento C1q/metabolismo , Receptor con Dominio Discoidina 2/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Colágeno/química , Complemento C1q/química , Receptor con Dominio Discoidina 1/genética , Receptor con Dominio Discoidina 1/metabolismo , Receptor con Dominio Discoidina 2/genética , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Microscopía Confocal , Péptidos/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal , Cicatrización de Heridas/fisiología
5.
Immunol Rev ; 269(1): 100-17, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26683148

RESUMEN

Numerous risk alleles for systemic lupus erythematosus (SLE) have now been identified. Analysis of the expression of genes with risk alleles in cells of hematopoietic origin demonstrates them to be most abundantly expressed in B cells and dendritic cells (DCs), suggesting that these cell types may be the drivers of the inflammatory changes seen in SLE. DCs are of particular interest as they act to connect the innate and the adaptive immune response. Thus, DCs can transform inflammation into autoimmunity, and autoantibodies are the hallmark of SLE. In this review, we focus on mechanisms of tolerance that maintain DCs in a non-activated, non-immunogenic state. We demonstrate, using examples from our own studies, how alterations in DC function stemming from either DC-intrinsic abnormalities or DC-extrinsic regulators of function can predispose to autoimmunity.


Asunto(s)
Linfocitos B/fisiología , Células Dendríticas/fisiología , Lupus Eritematoso Sistémico/inmunología , Inmunidad Adaptativa , Alelos , Autoanticuerpos/metabolismo , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Tolerancia Inmunológica , Inmunidad Innata , Lupus Eritematoso Sistémico/genética , Polimorfismo Genético , Factores de Riesgo
6.
Blood ; 128(18): 2218-2228, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27683415

RESUMEN

A healthy immune system results from a balance of stimulatory and inhibitory pathways that allow effective responses to acute insults, without descending into chronic inflammation. Failed homeostasis is characteristic of autoimmune diseases such as systemic lupus erythematosus. Although HMGB1 induces proinflammatory M1-like macrophage differentiation, we describe a mechanism by which C1q modulates this activity and collaborates with HMGB1 to induce the differentiation of monocytes to anti-inflammatory M2-like macrophages. These anti-inflammatory macrophages are unresponsive to dendritic cell induction factors, effectively removing them from participation in an adaptive immune response. This pathway is mediated through a complex with RAGE and LAIR-1 and depends on relative levels of C1q and HMGB1. Importantly, these data provide insight into a homeostatic mechanism in which C1q and HMGB1 can cooperate to terminate inflammation, and which may be impaired in C1q-deficient patients with autoimmune disease.


Asunto(s)
Diferenciación Celular/inmunología , Complemento C1q/metabolismo , Proteína HMGB1/metabolismo , Macrófagos/citología , Transducción de Señal/inmunología , Polaridad Celular , Complemento C1q/inmunología , Proteína HMGB1/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo
7.
Mol Med ; 20: 559-68, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25247291

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by abnormal function of both the innate and the adaptive immune system, leading to a loss of tolerance to self-antigens. Monocytes are a key component of the innate immune system and are efficient producers of multiple cytokines. In SLE, inappropriate activation of monocytes is thought to contribute to the loss of self-tolerance. In this study, we demonstrate that type 1 interferon (IFN) production by CpG-challenged monocytes can be suppressed by C1q through activating leukocyte-associated Ig-like receptor-1 (LAIR-1), which contains immunoreceptor tyrosine-based inhibition motifs (ITIMs). The phosphorylation of LAIR-1 and the interaction of LAIR-1 with SH2 domain-containing protein tyrosine phosphatase-1 (SHP-1) were enhanced after LAIR-1 engagement by C1q. Moreover, engagement of LAIR-1 by C1q inhibited nuclear translocation of interferon regulatory factor (IRF)-3 and IRF5 in CpG-stimulated monocytes. These data suggest a model in which LAIR-1 engagement by C1q helps maintain monocyte tolerance, specifically with respect to Toll-like receptor-9-mediated monocyte activation.


Asunto(s)
Complemento C1q/metabolismo , Monocitos/metabolismo , Receptores Inmunológicos/metabolismo , Células Cultivadas , Islas de CpG , Regulación de la Expresión Génica , Humanos , Interferón gamma/metabolismo , Interleucina-6 , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , ARN Interferente Pequeño/genética , Receptores Inmunológicos/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
Proc Natl Acad Sci U S A ; 109(46): E3160-7, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23093673

RESUMEN

C1q, the first component of complement, and leukocyte-associated Ig-like receptor 1 (LAIR-1; CD305), an inhibitory receptor expressed on hematopoietic cells, have both been associated with arrest of monocyte-derived dendritic cell (DC) differentiation and inhibition of Toll-like receptor activity in plasmacytoid DCs. Defects in both molecules have been implicated in susceptibility to, and progression of, systemic lupus erythematosus. Inhibitory signaling partners for C1q on monocytes and DCs remain undefined. Because C1q contains collagen-like motifs and LAIR-1 is a universal collagen receptor, we hypothesized that C1q is a functional ligand for LAIR-1. Binding analyses in cell-free systems and on the cell membrane demonstrate that C1q and its collagen tail associate with LAIR-1 and LAIR-2 (CD306), a soluble inhibitor of LAIR-1. Both C1q and its collagen tail trigger phosphorylation of LAIR-1 immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in monocytes. Functional analyses show that C1q-mediated inhibition of monocyte-DC differentiation and C1q-mediated inhibition of IFN-α production by plasmacytoid DCs were both reversed by LAIR-2. Moreover, C1q-mediated inhibition of DC differentiation was reversed by LAIR-1 siRNA. Thus, C1q is a functional ligand for LAIR-1 restricting immune cell differentiation and activation. The discovery of C1q interactions with LAIR-1 and LAIR-2 lends much needed insight into molecular mechanisms operating to prevent the loss of tolerance, particularly in systemic lupus erythematosus.


Asunto(s)
Diferenciación Celular/fisiología , Complemento C1q/inmunología , Células Dendríticas/inmunología , Monocitos/inmunología , Receptores Inmunológicos/inmunología , Transducción de Señal/fisiología , Secuencias de Aminoácidos , Complemento C1q/genética , Células Dendríticas/citología , Células HEK293 , Humanos , Tolerancia Inmunológica/fisiología , Interferón-alfa/inmunología , Ligandos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Monocitos/citología , Unión Proteica , Receptores Inmunológicos/genética
9.
J Autoimmun ; 44: 34-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23834843

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disorder that involves multiple organ systems and typically presents as a chronic inflammatory disease. Antibodies to double-stranded (ds) DNA are present in approximately 70% of patients and form nucleic acid containing immune complexes which activate dendritic cells through engagement of toll-like receptors, leading to a pro-inflammatory, pro-immunogenic milieu. In addition, anti-dsDNA antibodies deposit in kidneys to initiate glomerulonephritis. Antibodies to C1q have also been implicated in lupus nephritis and are found in 30-50% of patients. C1q is a known suppressor of immune activation and C1q deficiency is the strongest risk factor for SLE. We previously identified a subset of anti-DNA antibodies that binds the N-methyl-D-aspartate receptor. We now show that both mouse and human anti-DNA antibodies with this specificity bind C1q. These antibodies bind to Clq in glomeruli and exhibit decreased glomerular deposition in the absence of C1q. We propose that this subset of anti-DNA antibodies participates in lupus pathogenesis through direct targeting of C1q on glomeruli and also through removal of soluble C1q thereby limiting the ability of C1q to mediate immune homeostasis.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Complemento C1q/inmunología , Animales , Complejo Antígeno-Anticuerpo/inmunología , Enfermedades Autoinmunes/inmunología , Reacciones Cruzadas/inmunología , ADN/inmunología , Femenino , Humanos , Glomérulos Renales/inmunología , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos C57BL
10.
Exp Mol Med ; 54(5): 567-572, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35562585

RESUMEN

The importance of the complement component C1q has been highlighted by its involvement in autoimmunity, infection, inflammatory diseases, and tumors. The unique tulip-like structure of C1q has both a collagen-like stalk (C1q tail) and heterotrimeric globular head (gC1q), each with different binding specificities, and the binding of these components to their respective receptors leads to functional complexities in the body and bridges innate and adaptive immunity. This review describes the fundamental roles of C1q in various microenvironments and focuses on the importance of the interactions of C1q and its receptors with the inhibitory receptor LAIR-1 in maintaining homeostasis. Current therapeutic opportunities modulating LAIR-1 are also discussed.


Asunto(s)
Inmunidad Adaptativa , Complemento C1q , Proteínas Portadoras , Complemento C1q/química , Complemento C1q/metabolismo , Homeostasis
11.
Nat Commun ; 13(1): 7127, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-36443291

RESUMEN

Peptides, polymers of amino acids, comprise a vital and expanding therapeutic approach. Their rapid degradation by proteases, however, represents a major limitation to their therapeutic utility and chemical modifications to native peptides have been employed to mitigate this weakness. Herein, we describe functionalized thiocarbazate scaffolds as precursors of aza-amino acids, that, upon activation, can be integrated in a peptide sequence to generate azapeptides using conventional peptide synthetic methods. This methodology facilitates peptide editing-replacing targeted amino acid(s) with aza-amino acid(s) within a peptide-to form azapeptides with preferred therapeutic characteristics (extending half-life/bioavailability, while at the same time typically preserving structural features and biological activities). We demonstrate the convenience of this azapeptide synthesis platform in two well-studied peptides with short half-lives: FSSE/P5779, a tetrapeptide inhibitor of HMGB1/MD-2/TLR4 complex formation, and bradykinin, a nine-residue vasoactive peptide. This bench-stable thiocarbazate platform offers a robust and universal approach to optimize peptide-based therapeutics.


Asunto(s)
Aminoácidos , Bradiquinina , Semivida , Péptido Hidrolasas , Endopeptidasas
12.
J Biol Chem ; 285(10): 7271-80, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20007709

RESUMEN

The T cell-specific tyrosine kinase, p56(lck), plays crucial roles in T cell receptor (TCR)-mediated T cell activation. Here, we report that SOCS-6 (suppressor of cytokine signaling-6) is a negative regulator of p56(lck). SOCS-6 was identified as a protein binding to the kinase domain of p56(lck) through yeast two-hybrid screening. SOCS-6 bound specifically to p56(lck) (F505), which mimics the active form of p56(lck), but not to wild type p56(lck). In Jurkat T cells, SOCS-6 binding to p56(lck) was detected 1-2 h after TCR stimulation. Confocal microscopy showed that upon APC-T cell conjugation, SOCS-6 was recruited to the immunological synapse and colocalized with the active form of p56(lck). SOCS-6 promoted p56(lck) ubiquitination and its subsequent targeting to the proteasome. Moreover, SOCS-6 overexpression led to repression of TCR-dependent interleukin-2 promoter activity. These results establish that SOCS-6 acts as a negative regulator of T cell activation by promoting ubiquitin-dependent proteolysis.


Asunto(s)
Activación de Linfocitos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Linfocitos T/fisiología , Animales , Sitios de Unión , Humanos , Sinapsis Inmunológicas/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Células Jurkat , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Linfocitos T/citología , Técnicas del Sistema de Dos Híbridos
13.
Cells ; 10(3)2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807604

RESUMEN

The disruption of the immune tolerance induces autoimmunity such as systemic lupus erythematosus and vasculitis. A chromatin-binding non-histone protein, high mobility group box 1 (HMGB1), is released from the nucleus to the extracellular milieu in particular environments such as autoimmunity, sepsis and hypoxia. Extracellular HMGB1 engages pattern recognition receptors, including Toll-like receptors (TLRs) and the receptor for advanced glycation endproducts (RAGE). While the HMGB1-RAGE axis drives inflammation in various diseases, recent studies also focus on the anti-inflammatory effects of HMGB1 and RAGE. This review discusses current perspectives on HMGB1 and RAGE's roles in controlling inflammation and immune tolerance. We also suggest how RAGE heterodimers responding microenvironments functions in immune responses.


Asunto(s)
Proteína HMGB1/metabolismo , Tolerancia Inmunológica/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Humanos , Ratones
14.
J Exp Med ; 198(10): 1463-73, 2003 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-14610044

RESUMEN

In this study, we identify and characterize a novel transmembrane adaptor protein, designated Lck-interacting membrane protein (LIME), as a binding partner of the Lck Src homology (SH)2 domain. LIME possesses a short extracellular domain, a transmembrane domain, and a cytoplasmic tail containing five tyrosine-based motifs. The protein is primarily expressed in hematopoietic cells and lung. Interestingly, LIME expression is up-regulated by TCR stimulation and sustained up to 24 h, suggesting that LIME acts throughout the early to late stages of T cell activation. LIME is localized to membrane rafts and distributed within the T cell-APC contact site. Upon TCR stimulation of Jurkat T cells, LIME associates with Lck as a tyrosine-phosphorylated protein. Experiments using Jurkat T cells expressing CD8-LIME chimera reveal that the protein associates with phosphatidylinositol 3-kinase, Grb2, Gads, and SHP2, and activates ERK1/2 and JNK but not p38. Moreover, overexpression of LIME in Jurkat T cells induces transcriptional activation of the IL-2 promoter. Our data collectively show that LIME is a raft-associated transmembrane adaptor protein linking TCR stimuli to downstream signaling pathways via associations with Lck.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , ADN Complementario , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/inmunología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Especificidad de Órganos
15.
Front Immunol ; 11: 1057, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32536928

RESUMEN

The high-mobility group box 1 (HMGB1) has been shown to exert proinflammatory effects on many cells of the innate immune system. Originally identified as a nuclear protein, HMGB1 has been found to play an important role in mediating inflammation when released from apoptotic or necrotic cells as a damage-associated molecular pattern (DAMP). Systemic lupus erythematosus (SLE) is a disease of non-resolving inflammation, characterized by the presence of autoantibodies and systemic inflammation involving multiple organ systems. SLE patients have impaired clearance of apoptotic debris, which releases HMGB1 and other DAMPs extracellularly. HMGB1 activity is implicated in multiple disease phenotypes in SLE, including lupus nephritis and neuropsychiatric lupus. Elucidating the various properties of HMGB1 in SLE provides a better understanding of the disease and opens up new opportunities for designing potential therapeutics.


Asunto(s)
Proteína HMGB1/inmunología , Lupus Eritematoso Sistémico/inmunología , Inmunidad Adaptativa , Proteína HMGB1/antagonistas & inhibidores , Humanos , Inmunidad Innata , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/etiología , Nefritis Lúpica/etiología , Nefritis Lúpica/inmunología , Vasculitis por Lupus del Sistema Nervioso Central/etiología , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Modelos Inmunológicos
16.
Mol Cells ; 43(11): 921-934, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33243936

RESUMEN

Lck-interacting transmembrane adaptor 1 (LIME) has been previously identified as a raft-associated transmembrane protein expressed predominantly in T and B lymphocytes. Although LIME is shown to transduce the immunoreceptor signaling and immunological synapse formation via its tyrosine phosphorylation by Lck, a Src-family kinase, the in vivo function of LIME has remained elusive in the previous studies. Here we report that LIME is preferentially expressed in effector T cells and mediates chemokine-mediated T cell migration. Interestingly, in LIME-/- mice, while T cell receptor stimulation-dependent proliferation, differentiation to effector T cells, cytotoxic T lymphocyte (CTL) function and regulatory T lymphocyte (Treg) function were normal, only T cell-mediated inflammatory response was significantly defective. The reduced inflammation was accompanied by the impaired infiltration of leukocytes and T cells to the inflammatory sites of LIME-/- mice. More specifically, the absence of LIME in effector T cells resulted in the reduced migration and defective morphological polarization in response to inflammatory chemokines such as CCL5 and CXCL10. Consistently, LIME-/- effector T cells were found to be defective in chemokine-mediated activation of Rac1 and Rap1, and dysregulated phosphorylation of Pyk2 and Cas. Taken together, the present findings show that LIME is a critical regulator of inflammatory chemokine-mediated signaling and the subsequent migration of effector T cells to inflammatory sites.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Quimiocinas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Movimiento Celular , Humanos , Transducción de Señal
17.
Front Immunol ; 11: 1189, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32587593

RESUMEN

Most extracellular proteins are secreted via the classical endoplasmic reticulum (ER)/Golgi-dependent secretion pathway; however, some proteins, including a few danger-associated molecular patterns (DAMPs), are secreted via non-classical ER/Golgi-independent secretion pathways. The evolutionarily conserved high mobility group box1 (HMGB1) is a ubiquitous nuclear protein that can be released by almost all cell types. HMGB1 lacks signal peptide and utilizes diverse non-canonical secretion mechanisms for its extracellular export. Although the post-translational modifications of HMGB1 were demonstrated, the oxidation of HMGB1 and secretion mechanisms are not highlighted yet. We currently investigated that peroxiredoxins I and II (PrxI/II) induce the intramolecular disulfide bond formation of HMGB1 in the nucleus. Disulfide HMGB1 is preferentially transported out of the nucleus by binding to the nuclear exportin chromosome-region maintenance 1 (CRM1). We determined the kinetics of HMGB1 oxidation in bone marrow-derived macrophage as early as a few minutes after lipopolysaccharide treatment, peaking at 4 h while disulfide HMGB1 accumulation was observed within the cells, starting to secrete in the late time point. We have shown that HMGB1 oxidation status, which is known to determine the biological activity in extracellular HMGB1, is crucial for the secretion of HMGB1 from the nucleus. This review summarizes selected aspects of HMGB1 redox biology relevant to the induction and propagation of inflammatory diseases. We implicate the immunological significance and the need for novel HMGB1 inhibitors through mechanism-based studies.


Asunto(s)
Proteína HMGB1/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Humanos , Oxidación-Reducción , Transporte de Proteínas/fisiología
18.
Front Immunol ; 9: 2032, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30237803

RESUMEN

Patients surviving a septic episode exhibit persistent immune impairment and increased mortality due to enhanced vulnerability to infections. In the present study, using the cecal ligation and puncture (CLP) model of polymicrobial sepsis, we addressed the hypothesis that altered vagus nerve activity contributes to immune impairment in sepsis survivors. CLP-surviving mice exhibited less TNFα in serum following administration of LPS, a surrogate for an infectious challenge, than control-operated (control) mice. To evaluate the role of the vagus nerve in the diminished response to LPS, mice were subjected to bilateral subdiaphragmatic vagotomy at 2 weeks post-CLP. CLP-surviving vagotomized mice exhibited increased serum and tissue TNFα levels in response to LPS-challenge compared to CLP-surviving, non-vagotomized mice. Moreover, vagus nerve stimulation in control mice diminished the LPS-induced TNFα responses while having no effect in CLP mice, suggesting constitutive activation of vagus nerve signaling in CLP-survivors. The percentage of splenic CD4+ ChAT-EGFP+ T cells that relay vagus signals to macrophages was increased in CLP-survivors compared to control mice, and vagotomy in CLP-survivors resulted in a reduced percentage of ChAT-EGFP+ cells. Moreover, CD4 knockout CLP-surviving mice exhibited an enhanced LPS-induced TNFα response compared to wild-type mice, supporting a functional role for CD4+ ChAT+ T cells in mediating inhibition of LPS-induced TNFα responses in CLP-survivors. Blockade of the cholinergic anti-inflammatory pathway with methyllcaconitine, an α7 nicotinic acetylcholine receptor antagonist, restored LPS-induced TNFα responses in CLP-survivors. Our study demonstrates that the vagus nerve is constitutively active in CLP-survivors and contributes to the immune impairment.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Bacterias Gramnegativas/fisiología , Infecciones por Bacterias Grampositivas/inmunología , Sepsis/inmunología , Nervio Vago/fisiología , Animales , Ciego/cirugía , Modelos Animales de Enfermedad , Infecciones por Bacterias Grampositivas/metabolismo , Humanos , Tolerancia Inmunológica , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Sepsis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Nervio Vago/cirugía , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
19.
Front Immunol ; 9: 2824, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619247

RESUMEN

A primary mechanism for activation of innate immunity is recognition of damage or pathogen associated molecular patterns by pattern recognition receptors (PRRs). Nucleic acid is a damage associated molecular pattern molecule that when internalized into a monocyte and recognized by intracellular nucleic acid sensing toll like receptors will cause production of type 1 interferon. The process by which DNA or RNA is delivered into the cytosol of monocytes in systemic lupus erythematosus remains incompletely understood, and therapeutic approaches to prevent DNA-mediated monocyte activation are needed. We identified two mechanisms for internalization of DNA by monocytes. IgG-bound DNA was internalized by interacting with Fc gamma receptor IIa, while high-mobility group box-1 protein-bound DNA was internalized by interacting with the receptor for advanced glycation end products. Both pathways contribute to an inflammatory phenotype in monocytes exposed to serum from patients with SLE. Moreover, both of these pathways can be inhibited by a pentapeptide, DWEYS, which is a DNA mimetope. In one instance DWEYS directly competes with DNA for antibody binding and in the other DWEYS binds high-mobility group box-1 and blocks its interaction with RAGE. Our data highlight distinct pathways involved in nucleic acid enters monocytes in SLE, and identify a potential therapeutic to prevent nucleic acid internalization in SLE.


Asunto(s)
Anticuerpos Antinucleares , Ácidos Nucleicos Libres de Células , Inmunoglobulina G , Interferones , Lupus Eritematoso Sistémico , Monocitos , Animales , Anticuerpos Antinucleares/sangre , Anticuerpos Antinucleares/inmunología , Ácidos Nucleicos Libres de Células/sangre , Ácidos Nucleicos Libres de Células/inmunología , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interferones/sangre , Interferones/inmunología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Masculino , Ratones Mutantes , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , omegacloroacetofenona
20.
Front Immunol ; 9: 705, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29696019

RESUMEN

High-mobility group box 1 (HMGB1), a well-known danger-associated molecular pattern molecule, acts as a pro-inflammatory molecule when secreted by activated immune cells or released after necrotic cell damage. HMGB1 binds to immunogenic bacterial components and augments septic inflammation. In this study, we show how HMGB1 mediates complement activation, promoting sterile inflammation. We show that HMGB1 activates the classical pathway of complement system in an antibody-independent manner after binding to C1q. The C3a complement activation product in human plasma and C5b-9 membrane attack complexes on cell membrane surface are detected after the addition of HMGB1. In an acetaminophen (APAP)-induced hepatotoxicity model, APAP injection reduced HMGB1 levels and elevated C3 levels in C1q-deficient mouse serum samples, compared to that in wild-type (WT) mice. APAP-induced C3 consumption was inhibited by sRAGE treatment in WT mice. Moreover, in a mouse model of brain ischemia-reperfusion injury based on middle cerebral arterial occlusion, C5b-9 complexes were deposited on vessels where HMGB1 was accumulated, an effect that was suppressed upon HMGB1 neutralization. We propose that the HMGB1 released after cell necrosis and in ischemic condition can trigger the classical pathway of complement activation to exacerbate sterile inflammation.


Asunto(s)
Activación de Complemento/genética , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Proteína HMGB1/genética , Inflamación/genética , Inflamación/inmunología , Acetaminofén/efectos adversos , Animales , Anticuerpos Neutralizantes/inmunología , Biomarcadores , Línea Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Vía Clásica del Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Proteína HMGB1/metabolismo , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Unión Proteica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA