Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.324
Filtrar
1.
Front Immunol ; 15: 1372904, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38742116

RESUMO

Introduction: The California purple sea urchin, Strongylocentrotus purpuratus, relies solely on an innate immune system to combat the many pathogens in the marine environment. One aspect of their molecular defenses is the SpTransformer (SpTrf) gene family that is upregulated in response to immune challenge. The gene sequences are highly variable both within and among animals and likely encode thousands of SpTrf isoforms within the sea urchin population. The native SpTrf proteins bind foreign targets and augment phagocytosis of a marine Vibrio. A recombinant (r)SpTrf-E1-Ec protein produced by E. coli also binds Vibrio but does not augment phagocytosis. Methods: To address the question of whether other rSpTrf isoforms function as opsonins and augment phagocytosis, six rSpTrf proteins were expressed in insect cells. Results: The rSpTrf proteins are larger than expected, are glycosylated, and one dimerized irreversibly. Each rSpTrf protein cross-linked to inert magnetic beads (rSpTrf::beads) results in different levels of surface binding and phagocytosis by phagocytes. Initial analysis shows that significantly more rSpTrf::beads associate with cells compared to control BSA::beads. Binding specificity was verified by pre-incubating the rSpTrf::beads with antibodies, which reduces the association with phagocytes. The different rSpTrf::beads show significant differences for cell surface binding and phagocytosis by phagocytes. Furthermore, there are differences among the three distinct types of phagocytes that show specific vs. constitutive binding and phagocytosis. Conclusion: These findings illustrate the complexity and effectiveness of the sea urchin innate immune system driven by the natSpTrf proteins and the phagocyte cell populations that act to neutralize a wide range of foreign pathogens.


Assuntos
Fagócitos , Fagocitose , Proteínas Recombinantes , Animais , Fagocitose/imunologia , Fagócitos/imunologia , Fagócitos/metabolismo , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Ligação Proteica , Strongylocentrotus purpuratus/imunologia , Strongylocentrotus purpuratus/genética , Imunidade Inata , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Ouriços-do-Mar/imunologia , Vibrio/imunologia , Proteínas Opsonizantes/metabolismo , Proteínas Opsonizantes/imunologia
2.
FEBS Lett ; 598(8): 902-914, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38529702

RESUMO

Apolipoprotein E (apoE) is a regulator of lipid metabolism, cholesterol transport, and the clearance and aggregation of amyloid ß in the brain. The three human apoE isoforms apoE2, apoE3, and apoE4 only differ in one or two residues. Nevertheless, the functions highly depend on the isoform types and lipidated states. Here, we generated novel anti-apoE monoclonal antibodies (mAbs) and obtained an apoE4-selective mAb whose epitope is within residues 110-117. ELISA and bio-layer interferometry measurements demonstrated that the dissociation constants of mAbs are within the nanomolar range. Using the generated antibodies, we successfully constructed sandwich ELISA systems, which can detect all apoE isoforms or selectively detect apoE4. These results suggest the usability of the generated anti-apoE mAbs for selective detection of apoE isoforms.


Assuntos
Anticorpos Monoclonais , Apolipoproteínas E , Isoformas de Proteínas , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/química , Humanos , Isoformas de Proteínas/imunologia , Apolipoproteínas E/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/química , Apolipoproteínas E/imunologia , Animais , Epitopos/imunologia , Epitopos/química , Ensaio de Imunoadsorção Enzimática/métodos , Camundongos , Apolipoproteína E4/genética , Apolipoproteína E4/imunologia , Apolipoproteína E4/metabolismo , Camundongos Endogâmicos BALB C , Apolipoproteína E3/imunologia , Apolipoproteína E3/genética , Apolipoproteína E3/química , Apolipoproteína E3/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35131898

RESUMO

Type I interferons (IFN-I) exert pleiotropic biological effects during viral infections, balancing virus control versus immune-mediated pathologies, and have been successfully employed for the treatment of viral diseases. Humans express 12 IFN-alpha (α) subtypes, which activate downstream signaling cascades and result in distinct patterns of immune responses and differential antiviral responses. Inborn errors in IFN-I immunity and the presence of anti-IFN autoantibodies account for very severe courses of COVID-19; therefore, early administration of IFN-I may be protective against life-threatening disease. Here we comprehensively analyzed the antiviral activity of all IFNα subtypes against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to identify the underlying immune signatures and explore their therapeutic potential. Prophylaxis of primary human airway epithelial cells (hAEC) with different IFNα subtypes during SARS-CoV-2 infection uncovered distinct functional classes with high, intermediate, and low antiviral IFNs. In particular, IFNα5 showed superior antiviral activity against SARS-CoV-2 infection in vitro and in SARS-CoV-2-infected mice in vivo. Dose dependency studies further displayed additive effects upon coadministration with the broad antiviral drug remdesivir in cell culture. Transcriptomic analysis of IFN-treated hAEC revealed different transcriptional signatures, uncovering distinct, intersecting, and prototypical genes of individual IFNα subtypes. Global proteomic analyses systematically assessed the abundance of specific antiviral key effector molecules which are involved in IFN-I signaling pathways, negative regulation of viral processes, and immune effector processes for the potent antiviral IFNα5. Taken together, our data provide a systemic, multimodular definition of antiviral host responses mediated by defined IFN-I. This knowledge will support the development of novel therapeutic approaches against SARS-CoV-2.


Assuntos
Tratamento Farmacológico da COVID-19 , Interferon-alfa/farmacologia , SARS-CoV-2/efeitos dos fármacos , Transcriptoma , Replicação Viral/efeitos dos fármacos , Animais , COVID-19/imunologia , COVID-19/virologia , Chlorocebus aethiops , Clonagem Molecular , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Interferon-alfa/genética , Interferon-alfa/imunologia , Camundongos , Isoformas de Proteínas/classificação , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/farmacologia , Proteínas Recombinantes/classificação , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Transdução de Sinais , Células Vero
4.
Am J Hum Genet ; 109(2): 299-310, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35090584

RESUMO

Spontaneous clearance of acute hepatitis C virus (HCV) infection is associated with single nucleotide polymorphisms (SNPs) on the MHC class II. We fine-mapped the MHC region in European (n = 1,600; 594 HCV clearance/1,006 HCV persistence) and African (n = 1,869; 340 HCV clearance/1,529 HCV persistence) ancestry individuals and evaluated HCV peptide binding affinity of classical alleles. In both populations, HLA-DQß1Leu26 (p valueMeta = 1.24 × 10-14) located in pocket 4 was negatively associated with HCV spontaneous clearance and HLA-DQß1Pro55 (p valueMeta = 8.23 × 10-11) located in the peptide binding region was positively associated, independently of HLA-DQß1Leu26. These two amino acids are not in linkage disequilibrium (r2 < 0.1) and explain the SNPs and classical allele associations represented by rs2647011, rs9274711, HLA-DQB1∗03:01, and HLA-DRB1∗01:01. Additionally, HCV persistence classical alleles tagged by HLA-DQß1Leu26 had fewer HCV binding epitopes and lower predicted binding affinities compared to clearance alleles (geometric mean of combined IC50 nM of persistence versus clearance; 2,321 nM versus 761.7 nM, p value = 1.35 × 10-38). In summary, MHC class II fine-mapping revealed key amino acids in HLA-DQß1 explaining allelic and SNP associations with HCV outcomes. This mechanistic advance in understanding of natural recovery and immunogenetics of HCV might set the stage for much needed enhancement and design of vaccine to promote spontaneous clearance of HCV infection.


Assuntos
Cadeias beta de HLA-DQ/genética , Hepacivirus/patogenicidade , Hepatite C/genética , Interações Hospedeiro-Patógeno/genética , Polimorfismo de Nucleotídeo Único , Doença Aguda , Alelos , Substituição de Aminoácidos , População Negra , Feminino , Expressão Gênica , Estudo de Associação Genômica Ampla , Genótipo , Cadeias beta de HLA-DQ/imunologia , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/imunologia , Hepatite C/etnologia , Hepatite C/imunologia , Hepatite C/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Leucina/imunologia , Leucina/metabolismo , Masculino , Prolina/imunologia , Prolina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Remissão Espontânea , População Branca
5.
Nat Commun ; 13(1): 105, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013224

RESUMO

Zika virus (ZIKV) infection can be associated with neurological pathologies, such as microcephaly in newborns and Guillain-Barre syndrome in adults. Effective therapeutics are currently not available. As such, a comprehensive understanding of virus-host interactions may guide the development of medications for ZIKV. Here we report a human genome-wide overexpression screen to identify host factors that regulate ZIKV infection and find TMEM120A as a ZIKV restriction factor. TMEM120A overexpression significantly inhibits ZIKV replication, while TMEM120A knockdown increases ZIKV infection in cell lines. Moreover, Tmem120a knockout in mice facilitates ZIKV infection in primary mouse embryonic fibroblasts (MEF) cells. Mechanistically, the antiviral activity of TMEM120A is dependent on STING, as TMEM120A interacts with STING, promotes the translocation of STING from the endoplasmic reticulum (ER) to ER-Golgi intermediate compartment (ERGIC) and enhances the phosphorylation of downstream TBK1 and IRF3, resulting in the expression of multiple antiviral cytokines and interferon-stimulated genes. In summary, our gain-of-function screening identifies TMEM120A as a key activator of the antiviral signaling of STING.


Assuntos
Interações Hospedeiro-Patógeno/genética , Canais Iônicos/genética , Proteínas de Membrana/genética , Infecção por Zika virus/genética , Zika virus/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/virologia , Feminino , Regulação da Expressão Gênica , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Hepatócitos/imunologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Interferon beta/genética , Interferon beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Canais Iônicos/deficiência , Canais Iônicos/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Transdução de Sinais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Zika virus/crescimento & desenvolvimento , Zika virus/patogenicidade , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
6.
Front Immunol ; 12: 752394, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34737751

RESUMO

FOXP3 is the master transcription factor in both murine and human FOXP3+ regulatory T cells (Tregs), a T-cell subset with a central role in controlling immune responses. Loss of the functional Foxp3 protein in scurfy mice leads to acute early-onset lethal lymphoproliferation. Similarly, pathogenic FOXP3 mutations in humans lead to immunodysregulation, polyendocrinopathy, enteropathy, and X-linked (IPEX) syndrome, which are characterized by systemic autoimmunity that typically begins in the first year of life. However, although pathogenic FOXP3 mutations lead to overlapping phenotypic consequences in both systems, FOXP3 in human Tregs, but not mouse, is expressed as two predominant isoforms, the full length (FOXP3FL) and the alternatively spliced isoform, delta 2 (FOXP3Δ2). Here, using CRISPR/Cas9 to generate FOXP3 knockout CD4+ T cells (FOXP3KOGFP CD4+ T cells), we restore the expression of each isoform by lentiviral gene transfer to delineate their functional roles in human Tregs. When compared to FOXP3FL or FOXP3Δ2 alone, or double transduction of the same isoform, co-expression of FOXP3FL and FOXP3Δ2 induced the highest overall FOXP3 protein expression in FOXP3KOGFP CD4+ T cells. This condition, in turn, led to optimal acquisition of Treg-like cell phenotypes including downregulation of cytokines, such as IL-17, and increased suppressive function. Our data confirm that co-expression of FOXP3FL and FOXP3Δ2 leads to optimal Treg-like cell function and supports the need to maintain the expression of both when engineering therapeutics designed to restore FOXP3 function in otherwise deficient cells.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Linfócitos T Reguladores/imunologia , Técnicas de Inativação de Genes , Humanos , Fenótipo , Isoformas de Proteínas/imunologia
7.
J Immunol ; 207(11): 2699-2709, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34750204

RESUMO

IFN-γ-inducible protein 16 (IFI16) recognizes viral DNAs from both nucleus-replicating viruses and cytoplasm-replicating viruses. Isoform 2 of IFI16 (IFI16-iso2) with nuclear localization sequence (NLS) has been studied extensively as a well-known DNA sensor. However, the characteristics and functions of other IFI16 isoforms are almost unknown. Here, we find that IFI16-iso1, with exactly the same length as IFI16-iso2, lacks the NLS and locates in the cytoplasm. To distinguish the functions of IFI16-iso1 and IFI16-iso2, we have developed novel nuclear viral DNA mimics that can be recognized by the nuclear DNA sensors, including IFI16-iso2 and hnRNPA2B1. The hexanucleotide motif 5'-AGTGTT-3' DNA form of the nuclear localization sequence (DNLS) effectively drives cytoplasmic viral DNA nuclear translocation. These nuclear viral DNA mimics potently induce IFN-ß and antiviral IFN-stimulated genes in human A549 cells, HEK293T cells, and mouse macrophages. The subcellular location difference of IFI16 isoforms determines their differential functions in recognizing viral DNA and activating type I IFN-dependent antiviral immunity. IFI16-iso1 preferentially colocalizes with cytoplasmic HSV60mer and cytoplasm-replicating vaccinia virus (VACV), whereas IFI16-iso2 mainly colocalizes with nuclear HSV60-DNLS and nucleus-replicating HSV-1. Compared with IFI16-iso2, IFI16-iso1 induces more transcription of IFN-ß and IFN-stimulated genes, as well as stronger antiviral immunity upon HSV60mer transfection or VACV infection. IFI16-iso2, with the ability of nuclear-cytoplasmic shuttling, clears both invaded HSV type 1 and VACV significantly. However, IFI16-iso2 induces more type I IFN-dependent antiviral immunity than IFI16-iso1 upon HSV60-DNLS transfection or HSV type 1 infection. Our study has developed potent agonists for nuclear DNA sensors and also has demonstrated that IFI16 isoforms with cytoplasmic and nuclear locations play differential roles in innate immunity against DNA viruses.


Assuntos
Núcleo Celular/imunologia , Vírus de DNA/imunologia , Proteínas Nucleares/imunologia , Fosfoproteínas/imunologia , Células Cultivadas , Humanos , Isoformas de Proteínas/imunologia
8.
Front Immunol ; 12: 754710, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34712242

RESUMO

Rhesus macaques are a common non-human primate model used in the evaluation of human monoclonal antibodies, molecules whose effector functions depend on a conserved N-linked glycan in the Fc region. This carbohydrate is a target of glycoengineering efforts aimed at altering antibody effector function by modulating the affinity of Fcγ receptors. For example, a reduction in the overall core fucose content is one such strategy that can increase antibody-mediated cellular cytotoxicity by increasing Fc-FcγRIIIa affinity. While the position of the Fc glycan is conserved in macaques, differences in the frequency of glycoforms and the use of an alternate monosaccharide in sialylated glycan species add a degree of uncertainty to the testing of glycoengineered human antibodies in rhesus macaques. Using a panel of 16 human IgG1 glycovariants, we measured the affinities of macaque FcγRs for differing glycoforms via surface plasmon resonance. Our results suggest that macaques are a tractable species in which to test the effects of antibody glycoengineering.


Assuntos
Afinidade de Anticorpos/imunologia , Imunoglobulina G/imunologia , Macaca mulatta/imunologia , Modelos Animais , Receptores de IgG/imunologia , Animais , Glicosilação , Humanos , Macaca mulatta/metabolismo , Engenharia de Proteínas , Isoformas de Proteínas/imunologia , Receptores de IgG/metabolismo
9.
Front Immunol ; 12: 767505, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34712246

RESUMO

Interferon λ (IFN-λ) is critical for host viral defense at mucosal surfaces and stimulates immunomodulatory signals, acting on epithelial cells and few other cell types due to restricted IFN-λ receptor expression. Epithelial cells of the intestine play a critical role in the pathogenesis of Inflammatory Bowel Disease (IBD), and the related type II interferons (IFN-γ) have been extensively studied in the context of IBD. However, a role for IFN-λ in IBD onset and progression remains unclear. Recent investigations of IFN-λ in IBD are beginning to uncover complex and sometimes opposing actions, including pro-healing roles in colonic epithelial tissues and potentiation of epithelial cell death in the small intestine. Additionally, IFN-λ has been shown to act through non-epithelial cell types, such as neutrophils, to protect against excessive inflammation. In most cases IFN-λ demonstrates an ability to coordinate the host antiviral response without inducing collateral hyperinflammation, suggesting that IFN-λ signaling pathways could be a therapeutic target in IBD. This mini review discusses existing data on the role of IFN-λ in the pathogenesis of inflammatory bowel disease, current gaps in the research, and therapeutic potential of modulating the IFN-λ-stimulated response.


Assuntos
Células Epiteliais/imunologia , Imunidade Inata/imunologia , Doenças Inflamatórias Intestinais/imunologia , Interferons/imunologia , Mucosa Intestinal/imunologia , Transdução de Sinais/imunologia , Animais , Apoptose/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Células Epiteliais/metabolismo , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Interferons/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Modelos Imunológicos , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Fatores de Transcrição STAT/imunologia , Fatores de Transcrição STAT/metabolismo , Junções Íntimas/imunologia , Junções Íntimas/metabolismo , Interferon lambda
10.
Nat Commun ; 12(1): 5947, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34642338

RESUMO

Systemic sclerosis (SSc) is a chronic multisystem disorder characterized by fibrosis and autoimmunity. Interleukin (IL)-31 has been implicated in fibrosis and T helper (Th) 2 immune responses, both of which are characteristics of SSc. The exact role of IL-31 in SSc pathogenesis is unclear. Here we show the overexpression of IL-31 and IL-31 receptor A (IL-31RA) in dermal fibroblasts (DFs) from SSc patients. We elucidate the dual role of IL-31 in SSc, where IL-31 directly promotes collagen production in DFs and indirectly enhances Th2 immune responses by increasing pro-Th2 cytokine expression in DFs. Furthermore, blockade of IL-31 with anti-IL-31RA antibody significantly ameliorates fibrosis and Th2 polarization in a mouse model of SSc. Therefore, in addition to defining IL-31 as a mediator of fibrosis and Th2 immune responses in SSc, our study provides a rationale for targeting the IL-31/IL-31RA axis in the treatment of SSc.


Assuntos
Fibroblastos/imunologia , Interleucinas/genética , Receptores de Interleucina/genética , Escleroderma Sistêmico/imunologia , Células Th2/imunologia , Adulto , Idoso , Animais , Anticorpos Monoclonais/farmacologia , Colágeno Tipo I/genética , Colágeno Tipo I/imunologia , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica , Humanos , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-4/genética , Interleucina-4/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucinas/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Receptores de Interleucina/antagonistas & inibidores , Receptores de Interleucina/imunologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/patologia , Pele/efeitos dos fármacos , Pele/imunologia , Pele/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Células Th2/efeitos dos fármacos , Células Th2/patologia
11.
PLoS Comput Biol ; 17(9): e1009411, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34529669

RESUMO

Immunotherapies provide effective treatments for previously untreatable tumors and identifying tumor-specific epitopes can help elucidate the molecular determinants of therapy response. Here, we describe a pipeline, ISOTOPE (ISOform-guided prediction of epiTOPEs In Cancer), for the comprehensive identification of tumor-specific splicing-derived epitopes. Using RNA sequencing and mass spectrometry for MHC-I associated proteins, ISOTOPE identified neoepitopes from tumor-specific splicing events that are potentially presented by MHC-I complexes. Analysis of multiple samples indicates that splicing alterations may affect the production of self-epitopes and generate more candidate neoepitopes than somatic mutations. Although there was no difference in the number of splicing-derived neoepitopes between responders and non-responders to immune therapy, higher MHC-I binding affinity was associated with a positive response. Our analyses highlight the diversity of the immunogenic impacts of tumor-specific splicing alterations and the importance of studying splicing alterations to fully characterize tumors in the context of immunotherapies. ISOTOPE is available at https://github.com/comprna/ISOTOPE.


Assuntos
Epitopos/genética , Epitopos/imunologia , Neoplasias/genética , Neoplasias/imunologia , Processamento Alternativo/genética , Processamento Alternativo/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Carcinoma de Células Pequenas/genética , Carcinoma de Células Pequenas/imunologia , Linhagem Celular Tumoral , Biologia Computacional , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Masculino , Melanoma/genética , Melanoma/imunologia , Modelos Genéticos , Modelos Imunológicos , Mutação , Neoplasias/terapia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Splicing de RNA/genética , Splicing de RNA/imunologia , RNA-Seq
12.
Biomolecules ; 11(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34439806

RESUMO

Histones play a key role in chromatin remodeling and gene transcription. Further, free histones in the blood act as damage-associated molecules. Administration of histones to animals results in systemic inflammatory and toxic effects. Myelin basic protein is the principal constituent element of the myelin-proteolipid sheath of axons. Abzymes (antibodies with catalytic activities) are the original features of some autoimmune diseases. In this study, electrophoretically homogeneous IgGs against H1, H2A, H2B, H3, and H4 histones and myelin basic protein (MBP) were isolated from the blood sera of multiple sclerosis (MS) patients by several affinity chromatographies. Using MALDI mass spectrometry, the sites of H1 histone cleavage by IgGs against H1, H2A, H2B, H3, H4, and MBP were determined. It was shown that IgGs against H1 split H1 at 12 sites, while the number of cleavage sites by abzymes against other histones was lower: H2A (9), H2B (7), H3 (3), and H4 (3). The minimum rate of H1 hydrolysis was observed for antibodies against H3 and H4. A high rate of hydrolysis and the maximum number of H1 hydrolysis sites (17) were found for antibodies against MBP. Only a few sites of H1 hydrolysis by anti-H1 antibodies coincided with those for IgGs against H2A, H2B, H3, H4, and MBP. Thus, the polyreactivity of complexation and the enzymatic cross-activity of antibodies against H1, four other histones, and MBP have first been shown. Since histones act as damage molecules, abzymes against histones and MBP can play a negative role in the pathogenesis of MS and probably other different diseases as well.


Assuntos
Anticorpos Catalíticos/química , Autoanticorpos/química , Histonas/química , Imunoglobulina G/química , Esclerose Múltipla/sangue , Proteína Básica da Mielina/química , Sequência de Aminoácidos , Anticorpos Catalíticos/sangue , Anticorpos Catalíticos/isolamento & purificação , Autoanticorpos/sangue , Autoanticorpos/isolamento & purificação , Sítios de Ligação , Cromatografia de Afinidade , Histonas/sangue , Histonas/imunologia , Humanos , Hidrólise , Imunoglobulina G/sangue , Imunoglobulina G/isolamento & purificação , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Proteína Básica da Mielina/sangue , Proteína Básica da Mielina/imunologia , Ligação Proteica , Isoformas de Proteínas/sangue , Isoformas de Proteínas/química , Isoformas de Proteínas/imunologia , Proteólise , Especificidade por Substrato
13.
Immunobiology ; 226(5): 152124, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34333403

RESUMO

Asthma is a chronic respiratory disease characterized by airway inflammation and remodeling as well as hyper-responsiveness. Thymic stromal lymphopoietin (TSLP), which is a crucial inflammatory cytokine in immune homeostasis, consists of two isoforms, the long isoform lfTSLP and short isoform sfTSLP. The lfTSLP promotes inflammation and plays a pivotal role in asthma pathogenesis, while sfTSLP had been reported to have anti-asthma effects. Experiments have shown that lfTSLP could induce autophagy in hepatocytes. It is unknown whether lfTSLP or sfTSLP could influence autophagy and affect the progression of asthma. Using house dust mite (HDM)-stimulated airway smooth muscle cells as an in vitro model and HDM-induced asthma mice as in vivo model, we found that lfTSLP could induce autophagy and remodeling, while sfTSLP has the reverse effect. Strikingly, sfTSLP treatment in vivo reversed HDM-mediated activation of inflammation and airway remodeling, partly determined by autophagy change. These findings may help us understand the function of TSLP isoforms in the pathogenesis of asthma, and they support the use of drugs targeting sfTSLP and TSLP for asthma treatment.


Assuntos
Remodelação das Vias Aéreas/imunologia , Asma/imunologia , Citocinas/imunologia , Alérgenos/imunologia , Animais , Asma/sangue , Asma/patologia , Autofagia , Líquido da Lavagem Broncoalveolar/citologia , Linhagem Celular , Citocinas/sangue , Feminino , Humanos , Inflamação/sangue , Inflamação/imunologia , Inflamação/patologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/imunologia , Isoformas de Proteínas/imunologia , Pyroglyphidae/imunologia
14.
Cell Rep Med ; 2(6): 100320, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34195684

RESUMO

Necrotizing enterocolitis (NEC) is a deadly intestinal inflammatory disorder that primarily affects premature infants and lacks adequate therapeutics. Interleukin (IL)-22 plays a critical role in gut barrier maintenance, promoting epithelial regeneration, and controlling intestinal inflammation in adult animal models. However, the importance of IL-22 signaling in neonates during NEC remains unknown. We investigated the role of IL-22 in the neonatal intestine under homeostatic and inflammatory conditions by using a mouse model of NEC. Our data reveal that Il22 expression in neonatal murine intestine is negligible until weaning, and both human and murine neonates lack IL-22 production during NEC. Mice deficient in IL-22 or lacking the IL-22 receptor in the intestine display a similar susceptibility to NEC, consistent with the lack of endogenous IL-22 during development. Strikingly, treatment with recombinant IL-22 during NEC substantially reduces inflammation and enhances epithelial regeneration. These findings may provide a new therapeutic strategy to attenuate NEC.


Assuntos
Enterocolite Necrosante/imunologia , Interleucinas/genética , Mucosa Intestinal/imunologia , Proteínas Recombinantes/farmacologia , Regeneração/imunologia , Animais , Animais Recém-Nascidos , Quimiocina CXCL1/genética , Quimiocina CXCL1/imunologia , Quimiocina CXCL2/genética , Quimiocina CXCL2/imunologia , Modelos Animais de Doenças , Enterocolite Necrosante/tratamento farmacológico , Enterocolite Necrosante/microbiologia , Enterocolite Necrosante/patologia , Microbioma Gastrointestinal/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Recém-Nascido , Doenças do Recém-Nascido/imunologia , Doenças do Recém-Nascido/microbiologia , Doenças do Recém-Nascido/patologia , Recém-Nascido Prematuro , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucinas/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Knockout , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Receptores de Interleucina/genética , Receptores de Interleucina/imunologia , Regeneração/genética , Transdução de Sinais , Desmame , Interleucina 22
15.
Cancer Sci ; 112(10): 4187-4197, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34327762

RESUMO

Breast cancer is the most common cancer among women. Glycoprotein non-metastatic melanoma protein B (GPNMB), a type I transmembrane protein that is highly expressed in many cancers, including breast cancer, has been shown to be a prognostic factor. We previously reported that GPNMB overexpression confers tumorigenic potential, as evidenced by invasive tumor growth in vivo, sphere formation, and cellular migration and invasion to non-tumorigenic mammary epithelial cells. In this study, we focused on the serine (S) residue in the intracellular domain of GPNMB (S530 in human isoform b and S546 in mouse), which is predicted to be a phosphorylation site. To investigate the roles of this serine residue, we made an antibody specific for S530-phosphorylated human GPNMB and a point mutant in which S530 is replaced by an alanine (A) residue, GPNMB(SA). Established GPNMB(SA) overexpressing cells showed a significant reduction in sphere formation in vitro and tumor growth in vivo as a result of decreased stemness-related gene expression compared to that in GPNMB(WT)-expressing cells. In addition, GPNMB(SA) impaired GPNMB-mediated cellular migration. Furthermore, we found that tyrosine kinase receptor signaling triggered by epidermal growth factor or fibroblast growth factor 2 induces the serine phosphorylation of GPNMB through activation of downstream oncoproteins RAS and RAF.


Assuntos
Glicoproteínas de Membrana/fisiologia , Serina/metabolismo , Animais , Especificidade de Anticorpos , Linhagem Celular Tumoral , Movimento Celular/genética , Fator de Crescimento Epidérmico/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Células MCF-7 , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação , Mutação Puntual , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Quinases raf/metabolismo , Proteínas ras/metabolismo
16.
Biochemistry ; 60(32): 2463-2470, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34319067

RESUMO

The role of glutamate in excitatory neurotransmission depends on its transport into synaptic vesicles by the vesicular glutamate transporters (VGLUTs). The three VGLUT isoforms exhibit a complementary distribution in the nervous system, and the knockout of each produces severe, pleiotropic neurological effects. However, the available pharmacology lacks sensitivity and specificity, limiting the analysis of both transport mechanism and physiological role. To develop new molecular probes for the VGLUTs, we raised six mouse monoclonal antibodies to VGLUT2. All six bind to a structured region of VGLUT2, five to the luminal face, and one to the cytosolic. Two are specific to VGLUT2, whereas the other four bind to both VGLUT1 and 2; none detect VGLUT3. Antibody 8E11 recognizes an epitope spanning the three extracellular loops in the C-domain that explains the recognition of both VGLUT1 and 2 but not VGLUT3. 8E11 also inhibits both glutamate transport and the VGLUT-associated chloride conductance. Since the antibody binds outside the substrate recognition site, it acts allosterically to inhibit function, presumably by restricting conformational changes. The isoform specificity also shows that allosteric inhibition provides a mechanism to distinguish between closely related transporters.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas Vesiculares de Transporte de Glutamato/imunologia , Regulação Alostérica/imunologia , Animais , Cloretos/metabolismo , Epitopos/química , Epitopos/imunologia , Ácido Glutâmico/metabolismo , Células HEK293 , Humanos , Isoformas de Proteínas/imunologia , Proteína Vesicular 1 de Transporte de Glutamato/química , Proteína Vesicular 1 de Transporte de Glutamato/imunologia , Proteína Vesicular 2 de Transporte de Glutamato/química , Proteína Vesicular 2 de Transporte de Glutamato/imunologia , Proteínas Vesiculares de Transporte de Glutamato/química , Xenopus laevis
17.
J Interferon Cytokine Res ; 41(7): 235-243, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34280028

RESUMO

Atopic diseases, such as atopic dermatitis (AD), allergic asthma (AA), and allergic rhinitis (AR), are increasingly becoming a worldwide issue. This atopic triad originates at an early age and on a multifactorial basis, causing significant discomfort to susceptible individuals. The global case number is now reaching new highs, so exploring immune system regulation and its components is becoming critical. One cytokine, interleukin-32 (IL-32), is involved in inflammation and regulation of the immune system. It has nine isoforms that show varying degrees of expression, both intracellularly and extracellularly. IL-32 is secreted by immune cells, such as monocytes, macrophages, natural killer cells, and T cells, and by nonimmune cells, including fibroblasts, keratinocytes, and endothelial cells. Its production is regulated and augmented by microorganisms, mitogens, and other cytokines. Early studies demonstrated that IL-32 was an immune regulator that functioned to protect against inflammatory diseases, including AD, AA, and AR, and proposed a proinflammatory role for IL-32 in immune regulation and symptom exacerbation. However, several later reports suggested that IL-32 is downregulated in inflammatory diseases and exerts an anti-inflammatory effect. This review article focuses on recent findings regarding the detrimental and protective roles of IL-32 in development and management of inflammatory diseases. The exact role of IL-32 in AD, AA, and AR still remains to be elucidated. Future research should explore new avenues of IL-32 functionality in human inflammatory diseases.


Assuntos
Asma/imunologia , Citocinas/imunologia , Dermatite Atópica/imunologia , Interleucinas/imunologia , Rinite Alérgica/imunologia , Idade de Início , Asma/genética , Citocinas/classificação , Citocinas/genética , Dermatite Atópica/genética , Células Endoteliais/imunologia , Células Endoteliais/patologia , Fibroblastos/imunologia , Fibroblastos/patologia , Regulação da Expressão Gênica , Humanos , Interleucinas/genética , Queratinócitos/imunologia , Queratinócitos/patologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Macrófagos/imunologia , Macrófagos/patologia , Monócitos/imunologia , Monócitos/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Rinite Alérgica/genética , Rinite Alérgica/patologia , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/patologia
18.
Front Immunol ; 12: 705260, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34168659

RESUMO

The glycoprotein CD58, also known as lymphocyte-function antigen 3 (LFA-3), is a costimulatory receptor distributed on a broad range of human tissue cells. Its natural ligand CD2 is primarily expressed on the surface of T/NK cells. The CD2-CD58 interaction is an important component of the immunological synapse (IS) that induces activation and proliferation of T/NK cells and triggers a series of intracellular signaling in T/NK cells and target cells, respectively, in addition to promoting cell adhesion and recognition. Furthermore, a soluble form of CD58 (sCD58) is also present in cellular supernatant in vitro and in local tissues in vivo. The sCD58 is involved in T/NK cell-mediated immune responses as an immunosuppressive factor by affecting CD2-CD58 interaction. Altered accumulation of sCD58 may lead to immunosuppression of T/NK cells in the tumor microenvironment, allowing sCD58 as a novel immunotherapeutic target. Recently, the crucial roles of costimulatory molecule CD58 in immunomodulation seem to be reattracting the interests of investigators. In particular, the CD2-CD58 interaction is involved in the regulation of antiviral responses, inflammatory responses in autoimmune diseases, immune rejection of transplantation, and immune evasion of tumor cells. In this review, we provide a comprehensive summary of CD58 immunobiology.


Assuntos
Antígenos CD58/imunologia , Antígenos de Neoplasias/imunologia , Doenças Autoimunes/imunologia , Antígenos CD2/imunologia , Antígenos CD58/genética , Adesão Celular , Citocinas/fisiologia , Infecções por Citomegalovirus/imunologia , Células Endoteliais/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Rejeição de Enxerto/imunologia , Humanos , Sinapses Imunológicas/imunologia , Imunomodulação/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Células Matadoras Naturais/imunologia , Leucemia/imunologia , Linfoma/imunologia , Neoplasias/imunologia , Neoplasias/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia , Evasão Tumoral/imunologia , Microambiente Tumoral/imunologia
19.
Genome Biol ; 22(1): 190, 2021 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-34183059

RESUMO

Resistance to CD19-directed immunotherapies in lymphoblastic leukemia has been attributed, among other factors, to several aberrant CD19 pre-mRNA splicing events, including recently reported excision of a cryptic intron embedded within CD19 exon 2. While "exitrons" are known to exist in hundreds of human transcripts, we discovered, using reporter assays and direct long-read RNA sequencing (dRNA-seq), that the CD19 exitron is an artifact of reverse transcription. Extending our analysis to publicly available datasets, we identified dozens of questionable exitrons, dubbed "falsitrons," that appear only in cDNA-seq, but never in dRNA-seq. Our results highlight the importance of dRNA-seq for transcript isoform validation.


Assuntos
Processamento Alternativo , Artefatos , RNA Mensageiro/genética , Receptores de Antígenos de Linfócitos T/genética , Transcrição Reversa , Anticorpos Biespecíficos/farmacologia , Antineoplásicos Imunológicos/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/patologia , Pareamento de Bases , Sequência de Bases , Linhagem Celular Tumoral , Conjuntos de Dados como Assunto , Éxons , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunoterapia/métodos , Íntrons , Modelos Biológicos , Conformação de Ácido Nucleico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , RNA Mensageiro/química , RNA Mensageiro/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
20.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34001619

RESUMO

SERINC5 is a potent lentiviral restriction factor that gets incorporated into nascent virions and inhibits viral fusion and infectivity. The envelope glycoprotein (Env) is a key determinant for SERINC restriction, but many aspects of this relationship remain incompletely understood, and the mechanism of SERINC5 restriction remains unresolved. Here, we have used mutants of HIV-1 and HIV-2 to show that truncation of the Env cytoplasmic tail (ΔCT) confers complete resistance of both viruses to SERINC5 and SERINC3 restriction. Critically, fusion of HIV-1 ΔCT virus was not inhibited by SERINC5 incorporation into virions, providing a mechanism to explain how EnvCT truncation allows escape from restriction. Neutralization and inhibitor assays showed ΔCT viruses have an altered Env conformation and fusion kinetics, suggesting that EnvCT truncation dysregulates the processivity of entry, in turn allowing Env to escape targeting by SERINC5. Furthermore, HIV-1 and HIV-2 ΔCT viruses were also resistant to IFITMs, another entry-targeting family of restriction factors. Notably, while the EnvCT is essential for Env incorporation into HIV-1 virions and spreading infection in T cells, HIV-2 does not require the EnvCT. Here, we reveal a mechanism by which human lentiviruses can evade two potent Env-targeting restriction factors but show key differences in the capacity of HIV-1 and HIV-2 to exploit this. Taken together, this study provides insights into the interplay between HIV and entry-targeting restriction factors, revealing viral plasticity toward mechanisms of escape and a key role for the long lentiviral EnvCT in regulating these processes.


Assuntos
HIV-1/genética , HIV-2/genética , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Vírion/genética , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Linhagem Celular , Regulação da Expressão Gênica , Células HEK293 , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/patogenicidade , HIV-2/imunologia , HIV-2/patogenicidade , Células HeLa , Humanos , Evasão da Resposta Imune , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/imunologia , Domínios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Transdução de Sinais , Células-Tronco , Vírion/imunologia , Vírion/patogenicidade , Internalização do Vírus , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...