Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cells ; 10(5)2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33919154

RESUMO

Interleukin-18 receptor accessory protein (IL18RAP) is an indispensable subunit for the IL-18 receptor (IL-18R) complex's ability to mediate high-affinity IL-18 binding and signalling transduction. Interest in IL-18 in systemic lupus erythematosus (SLE) has been mostly focused on its role as a type 1 T helper cell-driving cytokine. The functional significance of IL18RAP in mediating the IL-18-driven response in myeloid cells in SLE remains largely unexplored. This study aimed to investigate the expression and function significance of IL18RAP in neutrophils of SLE patients. By qRT-PCR and Western blot analyses, elevated expressions of IL18RAP mRNA and protein were observed in neutrophils from SLE patients-particularly those with a history of nephritis. IL18RAP expression correlated negatively with complement 3 level and positively with disease activity, with higher expression in patients exhibiting renal and immunological manifestations. The increased IL18RAP expression in SLE neutrophils could be attributed to elevated type I interferon level in sera. Functionally, neutrophils from SLE patients showed higher IL-18-mediated enhancement in reactive oxygen species (ROS) generation, which showed positive correlation with IL18RAP expression and could be neutralized by anti-IL18RAP blocking antibodies. Taken together, our findings suggest that IL-18 could contribute to SLE pathogenesis through mediation of neutrophil dysfunction via the upregulation of IL18RAP expression.


Assuntos
Subunidade beta de Receptor de Interleucina-18/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Neutrófilos/imunologia , Espécies Reativas de Oxigênio/imunologia , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Interferon Tipo I/imunologia , Interleucina-18/imunologia , Masculino , Pessoa de Meia-Idade , Neutrófilos/citologia
2.
Adv Exp Med Biol ; 1253: 309-374, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32445101

RESUMO

Multiple sclerosis (MS) is an aggravating autoimmune disease that cripples young patients slowly with physical, sensory and cognitive deficits. The break of self-tolerance to neuronal antigens is the key to the pathogenesis of MS, with autoreactive T cells causing demyelination that subsequently leads to inflammation-mediated neurodegenerative events in the central nervous system. The exact etiology of MS remains elusive; however, the interplay of genetic and environmental factors contributes to disease development and progression. Given that genetic variation only accounts for a fraction of risk for MS, extrinsic risk factors including smoking, infection and lack of vitamin D or sunshine, which cause changes in gene expression, contribute to disease development through epigenetic regulation. To date, there is a growing body of scientific evidence to support the important roles of epigenetic processes in MS. In this chapter, the three main layers of epigenetic regulatory mechanisms, namely DNA methylation, histone modification and microRNA-mediated gene regulation, will be discussed, with a particular focus on the role of epigenetics on dysregulated immune responses and neurodegenerative events in MS. Also, the potential for epigenetic modifiers as biomarkers and therapeutics for MS will be reviewed.


Assuntos
Epigênese Genética , Esclerose Múltipla/genética , Metilação de DNA , Humanos , Fatores de Risco
3.
Int J Mol Sci ; 20(18)2019 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-31546763

RESUMO

Systemic lupus erythematosus (SLE) is a chronic, multifactorial autoimmune disease that predominantly affects young females. Dysregulation of different immune cell populations leads to self-tolerance breakdown and subsequent multiple organ damage as the disease develops. Plasmacytoid dendritic cells (pDCs) are potent producers of type I interferon (IFN), while myeloid dendritic cells (mDCs) are more specialized in antigen presentations. We have previously reported that bone-marrow (BM)-derived pDCs from the murine lupus model New Zealand black/white F1 (BWF1) possess abnormalities. Therefore, this study continues to investigate what aberrant properties peripheral pDCs and mDCs possess in BWF1 and how they mediate SLE progression, by comparing their properties in pre-symptomatic and symptomatic mice. Results showed that CD11chiCD11b+ myeloid DCs expanded during the disease state with down-regulation of co-stimulatory molecules and major histocompatibility complex class II molecules (MHC II), but their capacity to stimulate T cells was not hampered. During the disease state, this subset of mDCs displayed heightened toll-like receptors 7 and 9 (TLR 7/9) responses with increased interleukin 10 (IL-10) and C-X-C motif chemokine ligand 13 (CXCL13) expressions. Moreover, the expressions of myeloid differentiation primary response 88 (Myd88) and nuclear factor kappa B subunit 1 (Nfkb1) were higher in CD11chiCD11b+ DCs at the disease stage, leading to higher nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 phosphorylation activity. In summary, we reported aberrant phenotypic properties with enhanced TLR7/9 responses of CD11chiCD11b+ DCs in SLE mediated by aberrant NF-κB signaling pathway. Our findings add additional and novel information to our current understanding of the role of DCs in lupus immunopathogenesis. Lastly, molecular candidates in the NF-κB pathway should be exploited for developing therapeutic targets for SLE.


Assuntos
Antígenos CD11/imunologia , Antígeno CD11b/imunologia , Quimiocina CXCL13/imunologia , Células Dendríticas/imunologia , Interleucina-10/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Glicoproteínas de Membrana/imunologia , Subunidade p50 de NF-kappa B/imunologia , Receptor 7 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Animais , Antígenos CD11/genética , Antígeno CD11b/genética , Quimiocina CXCL13/genética , Células Dendríticas/patologia , Modelos Animais de Doenças , Feminino , Interleucina-10/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Células Mieloides/imunologia , Células Mieloides/patologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Subunidade p50 de NF-kappa B/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 7 Toll-Like/genética , Receptor Toll-Like 9/genética
4.
Cancer Lett ; 431: 201-212, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29859876

RESUMO

MicroRNAs (miRNAs) are endogenous small, non-coding RNAs that regulate genome expression at the post-transcriptional level. They are involved in a wide range of physiological processes including the maintenance of immune homeostasis and normal function. Accumulating evidence from animal studies show that alterations in pan or specific miRNA expression would break immunological tolerance, leading to autoimmunity. Differential miRNA expressions have also been documented in patients of many autoimmune disorders. In this review, we highlight the evidence that signifies the critical role of miRNAs in autoimmunity, specifically on their regulatory roles in the pathogenesis of several rheumatic diseases including systemic lupus erythematosus, rheumatoid arthritis and spondyloarthritis. The potential of miRNAs as biomarkers and therapeutic targets is also discussed. Manipulation of dysregulated miRNAs in vivo through miRNA delivery or inhibition offers promise for new therapeutic strategies in treating rheumatic diseases.


Assuntos
MicroRNAs/metabolismo , Doenças Reumáticas/imunologia , Doenças Reumáticas/metabolismo , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Autoimunidade , Biomarcadores/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica , Homeostase , Humanos , Sistema Imunitário , Tolerância Imunológica , Leucócitos/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Espondilite Anquilosante/imunologia , Espondilite Anquilosante/metabolismo , Linfócitos T Reguladores/metabolismo
5.
Mol Microbiol ; 106(5): 690-703, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28868744

RESUMO

The flagellar motor is an important virulence factor in infection by many bacterial pathogens. Motor function can be modulated by chemotactic proteins and recently appreciated proteins that are not part of the flagellar or chemotaxis systems. How these latter proteins affect flagellar activity is not fully understood. Here, we identified spermidine synthase SpeE as an interacting partner of switch protein FliM in Helicobacter pylori using pull-down assay and mass spectrometry. To understand how SpeE contributes to flagellar motility, a speE-null mutant was generated and its motility behavior was evaluated. We found that deletion of SpeE did not affect flagellar formation, but induced clockwise rotation bias. We further determined the crystal structure of the FliM-SpeE complex at 2.7 Å resolution. SpeE dimer binds to FliM with micromolar binding affinity, and their interaction is mediated through the ß1' and ß2' region of FliM middle domain. The FliM-SpeE binding interface partially overlaps with the FliM surface that interacts with FliG and is essential for proper flagellar rotational switching. By a combination of protein sequence conservation analysis and pull-down assays using FliM and SpeE orthologues in E. coli, our data suggest that FliM-SpeE association is unique to Helicobacter species.


Assuntos
Proteínas de Bactérias/metabolismo , Espermidina Sintase/metabolismo , Sítios de Ligação , Movimento Celular , Escherichia coli/metabolismo , Flagelos/metabolismo , Helicobacter pylori/metabolismo , Espectrometria de Massas , Proteínas de Membrana/metabolismo , Modelos Moleculares , Ligação Proteica , Elementos Estruturais de Proteínas
6.
Exp Cell Res ; 352(2): 304-312, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28223137

RESUMO

CLEC16A is genetically linked with multiple autoimmune disorders but its functional relevance in autoimmunity remains obscure. Recent evidence has signposted the emerging role of autophagy in autoimmune disease development. Here, by ectopic expression and siRNA silencing, we show that CLEC16A has an inhibitory role in starvation-induced autophagy in human cells. Combining quantitative proteomics and immunoblotting analyses, we found that CLEC16A likely regulates autophagy by activating mTOR pathway. Overexpression of CLEC16A was found to sensitize cells towards the availability of nutrients, resulting in a heightened mTOR activity, which in turn diminished LC3 autophagic activity following nutrient deprivation. CLEC16A deficiency, on the other hand, delayed mTOR activity in response to nutrient sensing, thereby resulted in an augmented autophagic response. CLEC16A was found residing in cytosolic vesicles and the Golgi, and nutrient removal promoted a stronger clustering within the Golgi, where it was possibly in a vantage position to activate mTOR upon nutrient replenishment. These findings suggest that Golgi-associated CLEC16A negatively regulates autophagy via modulation of mTOR activity, and may provide support for a functional link between CLEC16A and autoimmunity.


Assuntos
Autofagia , Lectinas Tipo C/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Vesículas Citoplasmáticas/metabolismo , Complexo de Golgi/metabolismo , Células HEK293 , Células HeLa , Humanos , Lectinas Tipo C/genética , Proteínas de Transporte de Monossacarídeos/genética
7.
Int J Mol Sci ; 17(8)2016 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-27509492

RESUMO

Systemic lupus erythematosus (SLE) is a chronic multi-organ autoimmune disease characterized by hyperactivated immune responses to self-antigens and persistent systemic inflammation. Previously, we reported abnormalities in circulating and bone marrow (BM)-derived plasmacytoid dendritic cells (pDCs) from SLE patients. Here, we aim to seek for potential regulators that mediate functional aberrations of pDCs in SLE. BM-derived pDCs from NZB/W F1 mice before and after the disease onset were compared for toll-like receptor (TLR) induced responses and microRNA profile changes. While pDCs derived from symptomatic mice were phenotypically comparable to pre-symptomatic ones, functionally they exhibited hypersensitivity to TLR7 but not TLR9 stimulation, as represented by the elevated upregulation of CD40, CD86 and MHC class II molecules upon R837 stimulation. Upregulated induction of miR-155 in symptomatic pDCs following TLR7 stimulation was observed. Transfection of miR-155 mimics in pre-symptomatic pDCs induced an augmented expression of Cd40, which is consistent with the increased CD40 expression in symptomatic pDCs. Overall, our results provide evidence for miR-155-mediated regulation in pDC functional abnormalities in SLE. Findings from this study contribute to a better understanding of SLE pathogenesis and ignite future interests in evaluating the molecular regulation in autoimmunity.


Assuntos
Antígenos CD40/genética , Células Dendríticas/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , MicroRNAs/fisiologia , Animais , Apresentação de Antígeno , Células da Medula Óssea/metabolismo , Antígenos CD40/metabolismo , Células Cultivadas , Feminino , Expressão Gênica , Glicoproteínas de Membrana/metabolismo , Camundongos Transgênicos , Interferência de RNA , Receptor 7 Toll-Like/metabolismo
8.
Restor Neurol Neurosci ; 34(3): 443-53, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27163251

RESUMO

PURPOSE: Systemic Lupus Erythematosus (SLE) is an autoimmune disease which is characterised by elevated levels of autoantibodies and cytokines in the body. Via alteration of the regulation of inflammation, damage to different organ systems, including the central nervous system (CNS), was found in SLE patients. Patients diagnosed with SLE were reported to suffer from different kinds of psychiatric signs and symptoms. As neurogenesis has been suggested to be a potential key player of psychiatric symptoms and emotional behavior disturbances, this study aims to investigate whether neurogenesis is altered in an animal model of SLE. Also, neuroinflammation was studied. METHODS: Female NZB/W F1 mice were used as an animal model of SLE. Animals were divided into two groups: 1. pre-diseased mice (lupus-prone NZB/W F1 female mice, age 10-15 weeks, negative for proteinuria and with basal levels of serum anti-dsDNA autoantibodies) and 2. diseased mice (NZB/W F1 female mice, > 25 weeks of age, with elevated serum levels of anti-dsDNA autoantibodies and with persistent proteinuria of > 3 mg/ml for more than 2 weeks). Comparisons of the levels of neurogenesis and neuroinflammtion between two groups of mice were studied by the immunohistochemistry. RESULTS: After the onset of SLE symptoms, a reduction of neurogenesis in the hippocampus was found, while there was a dramatic increase of doublecortin (DCX+) neuronal precursor cells in the corpus callosum (CC) and in the subventricular zone (SVZ). Meanwhile, exacerbated inflammation was present in the corpus callosum of the diseased mice, which was suggested by the increased number of GFAP+ cells and IBA-1+ cells. CONCLUSIONS: To the best of our knowledge, this is the first study showing an increase of neuronal precursor cells in the corpus callosum of the female NZB/W F1 mice. The present study suggests a coincidence but not a causal relationship between neurogenesis and neuroinflammation. The present results have also provided new insight showing that the altered neurogenesis and neuroinflammation may be a potential neurological mechanism for the cognitive and mood disturbance found in the SLE patients.


Assuntos
Corpo Caloso/patologia , Encefalite/etiologia , Encefalite/patologia , Lúpus Eritematoso Sistêmico/complicações , Células-Tronco Neurais/patologia , Animais , Autoanticorpos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Contagem de Células , Citocinas/metabolismo , DNA/imunologia , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo
9.
Liver Int ; 36(5): 737-45, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26351778

RESUMO

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is often associated with metastasis and recurrence leading to a poor prognosis. Therefore, development of novel treatment regimens is urgently needed to improve the survival of HCC patients. In this study, we aimed to investigate the in vitro and in vivo effects of anti-CD47 antibody alone and in combination with chemotherapy in HCC. METHODS: In this study, we examined the functional effects of anti-CD47 antibody (B6H12) on cell proliferation, sphere formation, migration and invasion, chemosensitivity, macrophage-mediated phagocytosis and tumourigenicity both in vitro and in vivo. The therapeutic efficacy of anti-CD47 antibody alone or in combination with doxorubicin was examined in patient-derived HCC xenograft. RESULTS: Blocking CD47 with anti-CD47 monoclonal antibody (B6H12) at 10 µg/ml could suppress self-renewal, tumourigenicity and migration and invasion abilities of MHCC-97L and Huh-7 cells. Interestingly, anti-CD47 antibody synergized the effect of HCC cells to chemotherapeutic drugs including doxorubicin and cisplatin. Blockade of CD47 by anti-CD47 antibody induced macrophage-mediated phagocytosis. Using a patient-derived HCC xenograft mouse model, we found that anti-CD47 antibody (400 µg/mouse) in combination with doxorubicin (2 mg/kg) exerted maximal effects on tumour suppression, as compared with doxorubicin and anti-CD47 antibody alone. CONCLUSIONS: Anti-CD47 antibody treatment could complement chemotherapy which may be a promising therapeutic strategy for the treatment of HCC patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígeno CD47/imunologia , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Ann Rheum Dis ; 74(6): 1302-10, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24573745

RESUMO

OBJECTIVE: Although Th17 cells have been increasingly recognised as an important effector in various autoimmune diseases, their function in the pathogenesis of Sjögren's syndrome (SS) remains largely uncharacterised. This study aims to determine the role of Th17 cells in the development of experimental SS (ESS). METHODS: The ESS was induced in wildtype and IL-17A knockout (IL-17 KO) C57BL/6 mice immunised with salivary glands (SG) proteins. Phenotypic analysis of immune cells in the draining cervical lymph nodes (CLN) and SG was performed by flow cytometry and immunofluorescence microscopy. To determine the role of Th17 cells in ESS, immunised IL-17 KO mice were adoptively transferred with in vitro-generated Th17 cells and monitored for SS development. The salivary flow rate was measured, whereas inflammatory infiltration and tissue destruction in SG were assessed by histopathology. RESULTS: SG protein-immunised mice developed overt SS symptoms with increased Th17 cells detected in CLN and within lymphocytic foci in inflamed SG. Notably, immunised IL-17 KO mice were completely resistant for SS induction, showing no evidence of disease symptoms and histopathological changes in SG. Adoptive transfer of Th17 cells rapidly induced the onset of ESS in immunised IL-17 KO mice with markedly reduced saliva secretion, elevated autoantibody production and pronounced inflammation and tissue damage in SG. CONCLUSIONS: Our findings have defined a critical role of Th17 cells in the pathogenesis of ESS. Further studies may validate Th17 cell as a potential target for treating SS.


Assuntos
Linfonodos/citologia , Proteínas e Peptídeos Salivares/imunologia , Síndrome de Sjogren/imunologia , Células Th17/imunologia , Animais , Autoanticorpos/imunologia , Modelos Animais de Doenças , Imunização , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pescoço
11.
Immune Netw ; 14(3): 138-48, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24999310

RESUMO

MicroRNAs (miRNAs) are endogenous small RNA molecules best known for their function in post-transcriptional gene regulation. Immunologically, miRNA regulates the differentiation and function of immune cells and its malfunction contributes to the development of various autoimmune diseases including systemic lupus erythematosus (SLE). Over the last decade, accumulating researches provide evidence for the connection between dysregulated miRNA network and autoimmunity. Interruption of miRNA biogenesis machinery contributes to the abnormal T and B cell development and particularly a reduced suppressive function of regulatory T cells, leading to systemic autoimmune diseases. Additionally, multiple factors under autoimmune conditions interfere with miRNA generation via key miRNA processing enzymes, thus further skewing the miRNA expression profile. Indeed, several independent miRNA profiling studies reported significant differences between SLE patients and healthy controls. Despite the lack of a consistent expression pattern on individual dysregulated miRNAs in SLE among these studies, the aberrant expression of distinct groups of miRNAs causes overlapping functional outcomes including perturbed type I interferon signalling cascade, DNA hypomethylation and hyperactivation of T and B cells. The impact of specific miRNA-mediated regulation on function of major immune cells in lupus is also discussed. Although research on the clinical application of miRNAs is still immature, through an integrated approach with advances in next generation sequencing, novel tools in bioinformatics database analysis and new in vitro and in vivo models for functional evaluation, the diagnostic and therapeutic potentials of miRNAs may bring to fruition in the future.

12.
Cell Mol Immunol ; 10(2): 133-42, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23455017

RESUMO

Autoreactive B cells are one of the key immune cells that have been implicated in the pathogenesis of systemic lupus erythematosus (SLE). In addition to the production of harmful auto-antibodies (auto-Abs), B cells prime autoreactive T cells as antigen-presenting cells and secrete a wide range of pro-inflammatory cytokines that have both autocrine and paracrine effects. Agents that modulate B cells may therefore be of potential therapeutic value. Current strategies include targeting B-cell surface antigens, cytokines that promote B-cell growth and functions, and B- and T-cell interactions. In this article, we review the role of B cells in SLE in animal and human studies, and we examine previous reports that support B-cell modulation as a promising strategy for the treatment of this condition. In addition, we present an update on the clinical trials that have evaluated the therapeutic efficacy and safety of agents that antagonize CD20, CD22 and B-lymphocyte stimulator (BLyS) in human SLE. While the results of many of these studies remain inconclusive, belimumab, a human monoclonal antibody against BLyS, has shown promise and has recently been approved by the US Food and Drug Administration as an indicated therapy for patients with mild to moderate SLE. Undoubtedly, advances in B-cell immunology will continue to lead us to a better understanding of SLE pathogenesis and the development of novel specific therapies that target B cells.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/terapia , Terapia de Alvo Molecular/métodos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/patologia , Subpopulações de Linfócitos B/patologia , Morte Celular/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Inibidores do Crescimento/metabolismo , Humanos , Lúpus Eritematoso Sistêmico/patologia
13.
Autoimmun Rev ; 11(12): 890-7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22503660

RESUMO

Dendritic cells (DCs) constitute a heterogeneous population of professional antigen presenting cells which are the initiators and key regulators for both immunity and tolerance induction. The significance and impact of DC biology in contemporary immunology and medical research is heightened by the award of the 2011 Nobel Prize for Medicine and Physiology to Ralf Steinman for his discovery and subsequent work on the role of DC in adaptive immunity. As a central regulator of immune responses, DCs also play a pivotal role in the pathogenesis of chronic inflammatory autoimmune conditions such as systemic lupus erythematosus (SLE). In this review, we will focus on the respective role of the two major subsets of blood DC, namely myeloid (m)-DC and plasmacytoid (p)-DC, in SLE immunopathogenesis. Accumulating evidence has highlighted pDCs as the culprit for SLE pathogenesis, mainly through type-I interferon production. Latest findings in the field also decipher the mechanisms by which pDCs interact with neutrophils and platelets and contribute to SLE development. The recent surge of interest in studying microRNA regulation in SLE pathogenesis will also be discussed.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Células Mieloides/imunologia , Animais , Apresentação de Antígeno , Plaquetas/imunologia , Comunicação Celular , Humanos , Lúpus Eritematoso Sistêmico/genética , MicroRNAs/genética , Neutrófilos/imunologia
14.
Hum Immunol ; 71(7): 702-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20359516

RESUMO

CD209 (DC-SIGN) is an important C-type lectin which acts a receptor of many pathogens. The single nucleotide polymorphism (SNP) -336A>G in the CD209 promoter has been demonstrated to regulate promoter activity and to be associated with several important infectious diseases, such as human immunodeficiency virus-1 (HIV-1), Mycobacterium tuberculosis, and Dengue fever. CD209 facilitates severe acute respiratory syndrome (SARS)-coronavirus spike protein-bearing pseudotype driven infection of permissive cells in vitro. In keeping with previously published findings, our in vitro studies confirmed that this SNP modulates gene promoter activity. Genetic association analysis of this SNP with clinico-pathologic outcomes in 824 serologic confirmed SARS patients showed that the -336AG/GG genotype SARS patients was associated with lower standardized lactate-dehydrogenase (LDH) levels compared with the -336AA patients (p = 0.014, odds ratio = 0.40). High LDH levels are known to be an independent predictor for poor clinical outcome, probably related to tissue destruction from immune hyperactivity. Hence, SARS patients with the CD209 -336 AA genotype carry a 60% chance of having a poorer prognosis. This association is in keeping with the role of CD209 in modulating immune response to viral infection. The relevance of these findings for other infectious diseases and inflammatory conditions would be worth investigating.


Assuntos
Moléculas de Adesão Celular/genética , Lectinas Tipo C/genética , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Receptores de Superfície Celular/genética , Síndrome Respiratória Aguda Grave/genética , Adulto , Antígenos CD/genética , Povo Asiático/genética , Moléculas de Adesão Celular/metabolismo , DNA/metabolismo , Sondas de DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Frequência do Gene/genética , Genótipo , Células HeLa , Heterozigoto , Homozigoto , Hong Kong , Humanos , L-Lactato Desidrogenase/sangue , Lectinas Tipo C/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/metabolismo , Ligação Proteica/genética , Receptores de Superfície Celular/metabolismo , Síndrome Respiratória Aguda Grave/sangue , Fator de Transcrição Sp1/genética , Fator de Transcrição AP-2/genética , Transfecção
15.
Int J Cancer ; 124(8): 1971-9, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19107930

RESUMO

Nasopharyngeal carcinoma (NPC) is a common neoplasm in Southeastern Asia, and cisplatin-containing regimens for combinational chemotherapy are widely used for treating locally recurrent or metastatic diseases. However, resistance to cisplatin is not infrequently seen and its associated side effects may be life-threatening. In this report, another metallo-pharmaceutical agent gold(III) porphyrin complex [Au(TPP)]Cl was investigated in comparison to cisplatin for its in vitro and in vivo anticancer effects. Through induction of the intrinsic apoptosis pathway, [Au(TPP)]Cl exhibited 100-fold higher potency than cisplatin in killing NPC cells, including cisplatin-sensitive and cisplatin-resistant variants, and also an variant harboring the Epstein-Barr virus. In addition, a safety concentration window was demonstrated, allowing [Au(TPP)]Cl to kill tumors with minimal cytotoxicity to noncancerous cells. More importantly, weekly intraperitoneal injection of 3 mg/kg [Au(TPP)]Cl was more effective than the same dose of cisplatin in inducing tumor apoptosis in vivo and remarkably inhibited tumor growth in animals without any noticeable side effect. [Au(TPP)]Cl therefore is a promising chemotherapeutic agent that deserves further development as a novel drug for the treatment of advanced NPC, in particular, for cases with cisplatin-resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma/tratamento farmacológico , Cisplatino/uso terapêutico , Ouro/administração & dosagem , Neoplasias Nasofaríngeas/tratamento farmacológico , Porfirinas/administração & dosagem , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Feminino , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Modelos Químicos , Resultado do Tratamento
16.
Bioconjug Chem ; 20(1): 24-31, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19102689

RESUMO

Nasopharyngeal carcinoma is a neoplasm with a high incidence in Southeast Asia, and it is strongly associated with Epstein-Barr virus (EBV) activation involving the expression of a weakly immunogenic protein, namely, latent membrane protein (LMP)-2. Previous immunological studies already identified the human leukocyte antigen (HLA)-A11 restricted peptide epitope (SSCSSCPLSK) in the LMP-2 antigen. In this work, we prepared gold nanoparticle (AuNP)-peptide conjugate 1 by treating the nanoparticles with the N-cysteinated LMP-2 epitope. The AuNP-peptide conjugates have been characterized by TEM (15-24 nm in diameter) and UV-vis spectroscopy (surface plasmon resonance absorption band at lambda(max) = 520 nm). In the presence of a CALNN capping peptide, the AuNP-peptide conjugates are stable in solution without aggregation at room temperature for at least 48 h. By ELIspot studies, AuNP-peptide conjugate 1 was found to elicit a significantly stronger INF-gamma response [number of spot forming cells (SPC) = 727 +/- 198] from peripheral blood mononuclear cells of healthy HLA-A11 donors when compared to that induced by the unconjugated LMP-2 peptides (SFC = 73 +/- 28). Further studies showed that dendritic cells treated with conjugate 1 can effect CD8+ T-cell activation leading to epitope-specific cytotoxic T lymphocyte killing responses in vitro.


Assuntos
Antígenos Virais/imunologia , Herpesvirus Humano 4/imunologia , Nanopartículas Metálicas/química , Linfócitos T Citotóxicos/imunologia , Proteínas da Matriz Viral/imunologia , Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Ouro , Humanos , Ativação Linfocitária/imunologia , Proteínas da Matriz Viral/química
17.
J Exp Med ; 204(11): 2529-36, 2007 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-17923501

RESUMO

Identification of the nature of severe acute respiratory syndrome (SARS)-infected cells is crucial toward understanding the pathogenesis. Using multicolor colocalization techniques, we previously reported that SARS(+) cells in the lung of fatally infected patients expressed the only known functional receptor, angiotensin-converting enzyme 2, and also a binding receptor, liver/lymph node-specific ICAM-3-grabbing non-integrin (CD209L). In this study, we show that SARS-infected cells also express the stem/progenitor cell markers CD34 and Oct-4, and do not express cytokeratin or surfactant. These putative lung stem/progenitor cells can also be identified in some non-SARS individuals and can be infected by SARS-coronavirus ex vivo. Infection of these cells may contribute to the loss of lung repair capacity that leads to respiratory failure as clinically observed.


Assuntos
Antígenos CD34/análise , Pulmão/fisiologia , Pulmão/virologia , Fator 3 de Transcrição de Octâmero/análise , Síndrome Respiratória Aguda Grave/patologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Células-Tronco/fisiologia , Idoso , Idoso de 80 Anos ou mais , Antígenos Virais/análise , Autopsia , Feminino , Humanos , Queratinas/análise , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Alvéolos Pulmonares/patologia , Surfactantes Pulmonares/análise , Síndrome Respiratória Aguda Grave/mortalidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA