Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(37): 13469-74, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25187559

RESUMO

Animals exploit different germ-line-encoded proteins with various domain structures to detect the signature molecules of pathogenic microbes. These molecules are known as pathogen-associated molecular patterns (PAMPs), and the host proteins that react with PAMPs are called pattern recognition proteins (PRPs). Here, we present a novel type of protein domain structure capable of binding to bacterial peptidoglycan (PGN) and the minimal PGN motif muramyl dipeptide (MDP). This domain is designated as apextrin C-terminal domain (ApeC), and its presence was confirmed in several invertebrate phyla and subphyla. Two apextrin-like proteins (ALP1 and ALP2) were identified in a basal chordate, the Japanese amphioxus Branchiostoma japonicum (bj). bjALP1 is a mucosal effector secreted into the gut lumen to agglutinate the Gram-positive bacterium Staphylococcus aureus via PGN binding. Neutralization of secreted bjALP1 by anti-bjALP1 monoclonal antibodies caused serious damage to the gut epithelium and rapid death of the animals after bacterial infection. bjALP2 is an intracellular PGN sensor that binds to TNF receptor-associated factor 6 (TRAF6) and prevents TRAF6 from self-ubiquitination and hence from NF-κB activation. MDP was found to compete with TRAF6 for bjALP2, which released TRAF6 to activate the NF-κB pathway. BjALP1 and bjALP2 therefore play distinct and complementary functions in amphioxus gut mucosal immunity. In conclusion, discovery of the ApeC domain and the functional analyses of amphioxus ALP1 and ALP2 allowed us to define a previously undocumented type of PRP that is represented across different animal phyla.


Assuntos
Bactérias/imunologia , Espaço Extracelular/microbiologia , Espaço Intracelular/microbiologia , Anfioxos/imunologia , Anfioxos/microbiologia , Proteínas/imunologia , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Aglutinação/efeitos dos fármacos , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Anfioxos/efeitos dos fármacos , Modelos Biológicos , Dados de Sequência Molecular , NF-kappa B/metabolismo , Peptidoglicano/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas/química , Proteínas/genética , Proteínas/ultraestrutura , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator 6 Associado a Receptor de TNF/metabolismo , Ubiquitinação/efeitos dos fármacos
2.
J Immunol ; 190(4): 1491-500, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23335747

RESUMO

Recently, amphioxus has served as a model for studying the origin and evolution of vertebrate immunity. However, little is known about how microRNAs (miRNAs) are involved in the immune defense in amphioxus. In this article, we present a systematic study of amphioxus miRNAs in the acute-phase response to bacterial infection; miR-92d was found to regulate the complement pathway in this basal chordate. We identified all 155 possible miRNAs present in the amphioxus Branchiostoma belcheri genome by bioinformatics analyses, including 57 newly identified miRNAs (called bbe-miRNAs), and characterized the miRNA expression pattern. Four miRNAs (bbe-miR-7, bbe-miR-4868a, bbe-miR-2065, and bbe-miR-34b) were upregulated and bbe-miR-92d was downregulated under the challenge of both Vibrio anguillarum and Staphylococcus aureus bacteria. We further predicted miRNA targets and identified mRNA targets of immune-related miRNA using the hybrid PCR method. We propose that miR-92d regulates the complement pathway through targeting C3 for controlling the acute immune response to bacterial infections. This study provides evidence for the complex immune regulation of miRNAs in the acute-phase response in basal chordates.


Assuntos
Cordados não Vertebrados/genética , Cordados não Vertebrados/imunologia , Complemento C3/metabolismo , Estudo de Associação Genômica Ampla/métodos , MicroRNAs/metabolismo , Imunidade Adaptativa/genética , Animais , Cordados não Vertebrados/microbiologia , Complemento C3/genética , Modelos Animais de Doenças , Evolução Molecular , Marcação de Genes/métodos , Células HEK293 , Humanos , MicroRNAs/biossíntese , MicroRNAs/genética , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/imunologia , Vibrioses/genética , Vibrioses/imunologia
3.
Int J Rheum Dis ; 27(2): e15077, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38402418

RESUMO

BACKGROUND: SAPHO syndrome is recognized as a rare entity with damage to skin and bones due to inflammation. Currently, the treatment for SAPHO syndrome is still a challenge in clinical practice. In this study, an integrated transcriptomics and network pharmacology approach was applied to explore the therapeutic effect and mechanism of Wang-Bi tablet (WBT) on SAPHO syndrome. METHODS: The main components of WBT and their targets, as well as the targets of SAPHO syndrome, were collected from databases. Network visualization was performed using Cytoscape software. The GO and KEGG enrichment analysis was executed by David dataset. Then, the molecular mechanism of WBT improving SAPHO syndrome was validated by transcriptomics of peripheral blood neutrophils in SAPHO syndrome. Finally, the above results were validated by molecular docking. RESULTS: The Network Pharmacology results showed there are 152 core targets for WBT treatment on SAPHO syndrome. RNA-seq data showed 442 differentially expressed genes (DEGs) in peripheral blood neutrophils of SAPHO patients. Intriguingly, NIK/NF-kappaB-, MyD88-dependent toll-like receptor-, and MAPK pathway were included in the enrichment results of network pharmacology and RNA-seq. Moreover, we verified that the core components of WBT have good affinity with the core targets of NIK/NF-kappaB-, MyD88-dependent toll-like receptor-, and MAPK pathway by molecular docking. CONCLUSIONS: This study illustrated that the possible mechanisms of WBT against SAPHO syndrome may be related to NIK/NF-kappaB-, MyD88-dependent toll-like receptor-, and MAPK pathway, and further experiments are needed to prove these predictions.


Assuntos
Síndrome de Hiperostose Adquirida , Medicamentos de Ervas Chinesas , Humanos , Síndrome de Hiperostose Adquirida/diagnóstico , Síndrome de Hiperostose Adquirida/tratamento farmacológico , Síndrome de Hiperostose Adquirida/genética , NF-kappa B , Simulação de Acoplamento Molecular , Fator 88 de Diferenciação Mieloide , Farmacologia em Rede , Perfilação da Expressão Gênica , Proteínas Adaptadoras de Transdução de Sinal , Receptores Toll-Like
4.
J Immunol ; 187(8): 3962-71, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21911601

RESUMO

Recently, receptor interacting protein (RIP)-1 has been recognized as an intracellular sensor at the crossroads of apoptosis, necroptosis, and cell survival. To reveal when this crucial molecule originated and how its function in integrating stress signals evolved, in this study we report on two RIP1 homologs in Chinese amphioxus (Branchiostoma belcheri tsingtauense), designated B. belcheri tsingtauense RIP1a and B. belcheri tsingtauense RIP1b. Phylogenetic analysis indicates that they are generated by domain recombination and lineage-specific duplication. Similar to human RIP1, both B. belcheri tsingtauense RIP1a and B. belcheri tsingtauense RIP1b activate NF-κB in a kinase activity-independent manner and induce apoptosis through the Fas-associated death domain protein-caspase cascade. Moreover, we found that the natural point mutation of Q to I in the RIP homotypic interaction motif of B. belcheri tsingtauense RIP1a provides negative feedback for amphioxus RIP1-mediated signaling. Thus, our study not only suggests that RIP1 has emerged as a molecular switch in triggering cell death or survival in a basal chordate, but also adds new insights into the regulation mechanisms of RIP1-related signaling, providing a novel perspective on human diseases mediated by RIP1.


Assuntos
Apoptose/genética , Linhagem da Célula , Sobrevivência Celular/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais/genética , Sequência de Aminoácidos , Animais , Apoptose/imunologia , Sequência de Bases , Cordados não Vertebrados , Imunofluorescência , Células HEK293 , Humanos , Imunoprecipitação , Hibridização In Situ , Dados de Sequência Molecular , Filogenia , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
5.
J Biol Chem ; 286(42): 36739-48, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21832079

RESUMO

The ficolin-mediated complement pathway plays an important role in vertebrate immunity, but it is not clear whether this pathway exists in invertebrates. Here we identified homologs of ficolin pathway components from the cephalochordate amphioxus and investigated whether they had been co-opted into a functional ficolin pathway. Four of these homologs, ficolin FCN1, serine protease MASP1 and MASP3, and complement component C3, were highly expressed in mucosal tissues and gonads, and were significantly up-regulated following bacterial infection. Recombinant FCN1 could induce hemagglutination, discriminate among sugar components, and specifically recognize and aggregate several bacteria (especially gram-positive strains) without showing bactericidal activity. This suggested that FCN1 is a dedicated pattern-recognition receptor. Recombinant serine protease MASP1/3 formed complexes with recombinant FCN1 and facilitated the activation of native C3 protein in amphioxus humoral fluid, in which C3 acted as an immune effector. We conclude that amphioxus have developed a functional ficolin-complement pathway. Because ficolin pathway components have not been reported in non-chordate species, our findings supported the idea that this pathway may represent a chordate-specific innovation in the evolution of the complement system.


Assuntos
Cordados não Vertebrados/imunologia , Complemento C3/imunologia , Evolução Molecular , Regulação da Expressão Gênica/fisiologia , Bactérias Gram-Positivas/imunologia , Lectinas/imunologia , Animais , Cordados não Vertebrados/genética , Cordados não Vertebrados/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Lectinas/genética , Lectinas/metabolismo , Especificidade de Órgãos/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Ficolinas
6.
J Immunol ; 181(10): 7024-32, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18981122

RESUMO

C1q is the target recognition protein of the classical complement pathway and a major connecting link between innate and adaptive immunities. Its globular signature domain is also found in a variety of noncomplement protein that can be grouped together as a C1q family. In this study, we have cloned and identified a novel C1q family member in cephalochordate amphioxus and named it as AmphiC1q1. The high transcriptional levels of this gene were detected during all stages of embryonic development, and the section in situ hybridization demonstrated that AmphiC1q1 was mainly expressed in the ovary, intestine, and nerve system of mature individuals. The transcript of AmphiC1q1 was up-regulated by LPS and gram-negative bacteria, but hardly by lipoteichoic acid and Staphylococcus aureus. The recombinant AmphiC1q1 protein could not bind with N-acetyl-glucosamine and did not possess hemagglutinating activity, indicating that AmphiC1q1 could not act as its lamprey homologue. But both the full-length protein and its truncated globular domain of C1q protein could interact with LPS. Moreover, recombinant AmphiC1q1 protein could inhibit collagen-induced platelet aggregation, but the truncated globular C1q domain protein would not, indicating that the blocking activity of AmphiC1q1 protein was via the collagen region of the protein. Our study on the primitive form of C1q family in protochordate will shed a light on understanding the gradual functional evolution of C1q family and eventual formation of mammalian homologues.


Assuntos
Cordados não Vertebrados/genética , Cordados não Vertebrados/metabolismo , Complemento C1q/genética , Complemento C1q/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Evolução Biológica , Southern Blotting , Expressão Gênica , Perfilação da Expressão Gênica , Hibridização In Situ , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência
7.
Mol Immunol ; 44(5): 756-62, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16762414

RESUMO

Two novel tumor necrosis factor receptors, Bbt-TNFR1 and Bbt-TNFR2, were isolated from Chinese amphioxus, the closest relative to vertebrate. The mRNA of Bbt-TNFR1 encoded a type I membrane protein of 452 amino acids, including four cysteine-rich domains in the extracellular region and a putative TRAF6-binding site at its 154aa long cytoplasmic tail. Bbt-TNFR2 was a 304aa long type I membrane protein, featuring three cysteine-rich domains and a short cytoplasmic tail of just 13 amino acids. Southern blot revealed that Bbt-TNFR1 was a single copy gene, while Bbt-TNFR2 was presented in multiple copies. Sequence comparison indicated that both Bbt-TNFR1 and Bbt-TNFR2 were weakly similar to LT-bR, HVEM, TNFR2, CD40, OX40 and DcR3. Real-time PCR showed that Bbt-TNFR1 and Bbt-TNFR2 were regulated during development and finally had high expression in mucosa-rich tissues in adult stage. Furthermore, up-regulated expression of both genes was also observed in guts after Gram-positive bacteria challenge. However, not like Bbt-TNFR2's slowly and gradually augmentation in the following 48 h, expression of Bbt-TNFR1 dramatically surged up within 4 h and then subsided rapidly. Taking together, Bbt-TNFR1 and Bbt-TNFR2 may involve in the host defense of Chinese amphioxus via distinct fashions.


Assuntos
Cordados não Vertebrados/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Cordados não Vertebrados/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus
8.
Genome Res ; 18(7): 1112-26, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18562681

RESUMO

It has been speculated that before vertebrates evolved somatic diversity-based adaptive immunity, the germline-encoded diversity of innate immunity may have been more developed. Amphioxus occupies the basal position of the chordate phylum and hence is an important reference to the evolution of vertebrate immunity. Here we report the first comprehensive genomic survey of the immune gene repertoire of the amphioxus Branchiostoma floridae. It has been reported that the purple sea urchin has a vastly expanded innate receptor repertoire not previously seen in other species, which includes 222 toll-like receptors (TLRs), 203 NOD/NALP-like receptors (NLRs), and 218 scavenger receptors (SRs). We discovered that the amphioxus genome contains comparable expansion with 71 TLR gene models, 118 NLR models, and 270 SR models. Amphioxus also expands other receptor-like families, including 1215 C-type lectin models, 240 LRR and IGcam-containing models, 1363 other LRR-containing models, 75 C1q-like models, 98 ficolin-like models, and hundreds of models containing complement-related domains. The expansion is not restricted to receptors but is likely to extend to intermediate signal transducers because there are 58 TIR adapter-like models, 36 TRAF models, 44 initiator caspase models, and 541 death-fold domain-containing models in the genome. Amphioxus also has a sophisticated TNF system and a complicated complement system not previously seen in other invertebrates. Besides the increase of gene number, domain combinations of immune proteins are also increased. Altogether, this survey suggests that the amphioxus, a species without vertebrate-type adaptive immunity, holds extraordinary innate complexity and diversity.


Assuntos
Cordados não Vertebrados/genética , Cordados não Vertebrados/imunologia , Variação Genética/imunologia , Genoma/imunologia , Imunidade Inata/genética , Fatores Imunológicos/genética , Receptores Imunológicos/genética , Animais , Cordados não Vertebrados/embriologia , Drosophila melanogaster , Humanos , Lampreias , Camundongos , Dados de Sequência Molecular , Ouriços-do-Mar , Análise de Sequência de DNA , Especificidade da Espécie , Peixe-Zebra
9.
Glycobiology ; 17(7): 774-83, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17442707

RESUMO

A novel F4-carbohydrate recognition domain (CRD)-linker-F3-CRD-type bi-CRD Branchiostoma belcheri tsingtauense galectin (BbtGal)-L together with its alternatively spliced mono-CRD isoform BbtGal-S from amphioxus intestine was encoded by a 9488-bp unique gene with eight exons and seven introns. The recombinant proteins of BbtGal were found to have beta-galactoside-binding activity, indicating that BbtGal was a member of the galectin family. Phylogenetic analysis of this gene along with its splicing form and genome structure suggested that the BbtGal gene was the primitive form of the chordate galectin family. Real-time polymerase chain reaction analyses (PCR) indicated that BbtGal mRNA was expressed during all stages of embryonic development. In terms of tissue distribution, BbtGal-L mRNA was mainly expressed in the immunity-related organs, such as hepatic diverticulum, intestine, and gill, but BbtGal-S was ubiquitously expressed in all tissues. The expression of BbtGal-L mRNA was elevated after acute challenge with various microorganisms, but BbtGal-L only bound to specific bacteria. The immune function of BbtGal was consistent with its localization both outside and inside the cell. Our study on amphioxus galectin may help further understanding of the evolution of chordate galectin in terms of host-pathogen interaction in the immune system.


Assuntos
Cordados/microbiologia , Escherichia coli/metabolismo , Galectinas/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Carboidratos/química , Cordados/metabolismo , Evolução Molecular , Galectinas/metabolismo , Lactose/química , Modelos Biológicos , Modelos Genéticos , Dados de Sequência Molecular , Filogenia , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos
10.
J Immunol ; 179(12): 8425-34, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18056389

RESUMO

To investigate the evolution and immune function of C-type lectin in amphioxus, the primitive representative of the chordate phylum, we identified three C-type lectins consisting solely of a carbohydrate recognition domain and N-terminal signal peptide and found that they had distinct express patterns in special tissues and immune response to stimulations analyzed by quantitative real-time PCR. We characterized the biochemical and biological properties of AmphiCTL1, which was dramatically up-regulated in amphioxus challenged with Staphylococcus aureus, Saccharomyces cerevisiae, and zymosan. Immunohistochemistry demonstrated that the localization of AmphiCTL1 protein was exclusively detected in the inner folding tissues of the hepatic diverticulum. Recombinant AmphiCTL1 was characterized as a typical Ca2+-dependent carbohydrate-binding protein possessing hemagglutinating activity, preferentially bound to all examined four Gram-positive bacteria and two yeast strains, but had little binding activity toward four Gram-negative bacteria we tested. It aggregated S. aureus and S. cerevisiae in a Ca2+-dependent manner and specifically bound to insoluble peptidoglycan and glucan, but not to LPS, lipoteichoic acid, and mannan. Calcium increased the intensity of the interaction between AmphiCTL1 and those components, but was not essential. This lectin directly killed S. aureus and S. cerevisiae in a Ca2+-independent fashion, and its binding to microorganism cell wall polysaccharides such as peptidoglycan and glucan preceded microbial killing activity. These findings suggested that AmphiCTL1 acted as a direct microbial killing C-type lectin through binding microbial targets via interaction with peptidoglycan and glucan. Thus, AmphiCTL1 may be an evolutionarily primitive form of antimicrobial protein involved in lectin-mediated innate immunity.


Assuntos
Cordados/imunologia , Cordados/microbiologia , Glucanos/metabolismo , Lectinas Tipo C/metabolismo , Peptidoglicano/metabolismo , Sequência de Aminoácidos , Animais , Cordados/genética , Clonagem Molecular , Lectinas Tipo C/análise , Lectinas Tipo C/genética , Dados de Sequência Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/imunologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/imunologia , Zimosan/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA