Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurovirol ; 27(4): 531-541, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34342851

RESUMO

The conjugation of small ubiquitin-like modifier (SUMO) proteins to substrates is a well-described post-translational modification that regulates protein activity, subcellular localization, and protein-protein interactions for a variety of downstream cellular activities. Several studies describe SUMOylation as an essential post-translational modification for successful viral infection across a broad range of viruses, including RNA and DNA viruses, both enveloped and un-enveloped. These viruses include but are not limited to herpes viruses, human immunodeficiency virus-1, and coronaviruses. In addition to the SUMOylation of viral proteins during infection, evidence shows that viruses manipulate the SUMO pathway for host protein SUMOylation. SUMOylation of host and viral proteins greatly impacts host innate immunity through viral manipulation of the host SUMOylation machinery to promote viral replication and pathogenesis. Other post-translational modifications like phosphorylation can also modulate SUMO function. For example, phosphorylation of COUP-TF interacting protein 2 (CTIP2) leads to its SUMOylation and subsequent proteasomal degradation. The SUMOylation of CTIP2 and subsequent degradation prevents CTIP2-mediated recruitment of a multi-enzymatic complex to the HIV-1 promoter that usually prevents the transcription of integrated viral DNA. Thus, the "SUMO switch" could have implications for CTIP2-mediated transcriptional repression of HIV-1 in latency and viral persistence. In this review, we describe the consequences of SUMO in innate immunity and then focus on the various ways that viral pathogens have evolved to hijack the conserved SUMO machinery. Increased understanding of the many roles of SUMOylation in viral infections can lead to novel insight into the regulation of viral pathogenesis with the potential to uncover new targets for antiviral therapies.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Imunidade Inata/fisiologia , Sumoilação/fisiologia , Viroses/imunologia , Viroses/metabolismo , Animais , Humanos , Processamento de Proteína Pós-Traducional , Proteína SUMO-1/imunologia , Proteína SUMO-1/metabolismo
2.
Biologicals ; 62: 22-26, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31668855

RESUMO

Salmonella is found to be a major causes of food borne diseases globally. Poultry products contaminated with this pathogen is one of the major sources of infections in humans. Outer membrane protein C (OmpC) of Salmonella Typhimurium is a promising DNA vaccine candidate to mitigate Salmonella infection in poultry. However, the large-scale production of bioactive recombinant OmpC (rOmpC) protein is hindered due to the formation of inclusion bodies in Escherichia coli. The objective of this work was to attain high level expression of rOmpC protein, purify and evaluate its functional properties. The ompC gene was optimized and fused with small ubiquitin-related modifier (SUMO) gene for high level expression as soluble protein. The fusion protein with ~58 kDa molecular weight was observed on SDS-PAGE gel. The expression levels of rOmpC fusion protein reached maximum of 38% of total soluble protein (TSP) after 8 h of 0.2% rhamnose induction. Protein purification was carried out using nickel nitrilotriacetic acid (Ni-NTA) purification column. Western blot were performed to analyse expression and immunoreactivity of rOmpC fusion protein. The results indicate that SUMO fusion system is ideal for large scale production of functional rOmpC fusion protein expression in E. coli.


Assuntos
Proteínas de Bactérias , Escherichia coli , Imunoglobulinas/imunologia , Porinas , Proteínas Recombinantes de Fusão , Proteína SUMO-1 , Salmonella typhimurium/genética , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/isolamento & purificação , Galinhas , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Porinas/biossíntese , Porinas/genética , Porinas/imunologia , Porinas/isolamento & purificação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteína SUMO-1/biossíntese , Proteína SUMO-1/genética , Proteína SUMO-1/imunologia , Proteína SUMO-1/isolamento & purificação , Salmonella typhimurium/metabolismo
3.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597768

RESUMO

Some viruses take advantage of conjugation of ubiquitin or ubiquitin-like proteins to enhance their own replication. One example is Ebola virus, which has evolved strategies to utilize these modification pathways to regulate the viral proteins VP40 and VP35 and to counteract the host defenses. Here, we show a novel mechanism by which Ebola virus exploits the ubiquitin and SUMO pathways. Our data reveal that minor matrix protein VP24 of Ebola virus is a bona fide SUMO target. Analysis of a SUMOylation-defective VP24 mutant revealed a reduced ability to block the type I interferon (IFN) pathway and to inhibit IFN-mediated STAT1 nuclear translocation, exhibiting a weaker interaction with karyopherin 5 and significantly diminished stability. Using glutathione S-transferase (GST) pulldown assay, we found that VP24 also interacts with SUMO in a noncovalent manner through a SIM domain. Mutation of the SIM domain in VP24 resulted in a complete inability of the protein to downmodulate the IFN pathway and in the monoubiquitination of the protein. We identified SUMO deubiquitinating enzyme ubiquitin-specific-processing protease 7 (USP7) as an interactor and a negative modulator of VP24 ubiquitination. Finally, we show that mutation of one ubiquitination site in VP24 potentiates the IFN modulatory activity of the viral protein and its ability to block IFN-mediated STAT1 nuclear translocation, pointing to the ubiquitination of VP24 as a negative modulator of the VP24 activity. Altogether, these results indicate that SUMO interacts with VP24 and promotes its USP7-mediated deubiquitination, playing a key role in the interference with the innate immune response mediated by the viral protein.IMPORTANCE The Ebola virus VP24 protein plays a critical role in escape of the virus from the host innate immune response. Therefore, deciphering the molecular mechanisms modulating VP24 activity may be useful to identify potential targets amenable to therapeutics. Here, we identify the cellular proteins USP7, SUMO, and ubiquitin as novel interactors and regulators of VP24. These interactions may represent novel potential targets to design new antivirals with the ability to modulate Ebola virus replication.


Assuntos
Ebolavirus/genética , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Proteína SUMO-1/química , Peptidase 7 Específica de Ubiquitina/genética , Proteínas Virais/química , Animais , Sítios de Ligação , Chlorocebus aethiops , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Transporte Proteico , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Proteína SUMO-1/genética , Proteína SUMO-1/imunologia , Transdução de Sinais , Sumoilação , Peptidase 7 Específica de Ubiquitina/imunologia , Células Vero , Proteínas Virais/genética , Proteínas Virais/imunologia , alfa Carioferinas/genética , alfa Carioferinas/imunologia
4.
J Vet Sci ; 20(5): e54, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31565897

RESUMO

Brucella is an intracellular pathogen that invades a host and settles in its immune cells; however, the mechanism of its intracellular survival is unclear. Modification of small ubiquitin-related modifier (SUMO) occurs in many cellular activities. E2 conjugating enzyme 9 (Ubc9) is the only reported ubiquitin-conjugating enzyme that links the SUMO molecule with a target protein. Brucella's intracellular survival mechanism has not been studied with respect to SUMO-related proteins and Ubc9. Therefore, to investigate the relationship between Brucella melitensis 16M and SUMO, we constructed plasmids and cells lines suitable for overexpression and knockdown of SUMO1 and Ubc9 genes. Brucella 16M activated SUMO1/Ubc9 expression in a time-dependent manner, and Brucella 16M intracellular survival was inhibited by SUMO1/Ubc9 overexpression and promoted by SUMO1/Ubc9 depletion. In macrophages, Brucella 16M-dependent apoptosis and immune factors were induced by SUMO1/Ubc9 overexpression and restricted by SUMO1/Ubc9 depletion. We noted no effect on the expressions of SUMO1 and Ubc9 in B. melitensis 16M lipopolysaccharide-prestimulated mouse RAW264.7 macrophages. Additionally, intracellular survival of the 16M△VirB2 mutant was lower than that of Brucella 16M (p < 0.05). VirB2 can affect expression levels of Ubc9, thereby increasing intracellular survival of Brucella in macrophages at the late stage of infection. Collectively, our results demonstrate that B. melitensis 16M may use the VirB IV secretion system of Brucella to interact with SUMO-related proteins during infection of host cells, which interferes with SUMO function and promotes pathogen survival in host cells.


Assuntos
Brucella melitensis/fisiologia , Proteína SUMO-1/genética , Proteína SUMO-1/imunologia , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/imunologia , Animais , Camundongos , Células RAW 264.7
5.
Fish Shellfish Immunol ; 86: 1088-1095, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30593901

RESUMO

Protein SUMOylation (SUMO is small ubiquitin-related modifier) is a dynamic process that is strictly regulated under physiological and pathological conditions. We previously cloned and characterized two SUMO homologue genes (EcSUMO1 and EcSUMO2) from orange-spotted grouper (Epinephelus coioides). In the present study, the SUMO3 homologue from E. coioides (EcSUMO3) was cloned and its possible roles in fish immunity were analyzed. The open reading frame of EcSUMO3 contains 285 base pairs encoding a 94 amino acid protein with a predicted molecular mass of 10.73 kDa. The protein sequence of EcSUMO3 revealed similar domains with mammals, including the UBQ (ubiquitin-like proteins) domain, the hydrophobic surface, the Ulp1-Smt3 interaction sites, a VKTE motif and the C-terminal Gly residues. EcSUMO3 shares 46.83% and 89.58% identity with EcSUMO1 and EcSUMO2, respectively, and it shares 94%, 98%, and 98% identity with SUMO3 from Oreochromis niloticus, Danio rerio, and Homo sapiens, respectively. Quantitative real-time polymerase chain reaction analysis indicated that EcSUMO3 was constitutively expressed in all of the analyzed tissues in healthy grouper. EcSUMO3 expression levels were remarkably (p < 0.01) up-regulated in grouper spleen (GS) cells in response to stimulation with red-spotted grouper nervous necrosis virus (RGNNV) and Singapore grouper iridovirus (SGIV). EcSUMO3 was distributed in both the cytoplasm and nucleus in GS cells. EcSUMO3 enhanced SGIV and RGNNV replication during viral infection in vitro. These results are important for better understanding of the SUMO pathway in fish and provide insights into the regulatory mechanism of viral infection in E. coioides under farmed conditions.


Assuntos
Bass/genética , Infecções por Vírus de DNA/veterinária , Doenças dos Peixes/virologia , Infecções por Vírus de RNA/veterinária , Proteína SUMO-1/genética , Sequência de Aminoácidos , Animais , Bass/imunologia , Infecções por Vírus de DNA/imunologia , Proteínas de Peixes/química , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Imunidade Inata/genética , Iridovirus/fisiologia , Nodaviridae/fisiologia , Proteína SUMO-1/imunologia , Ubiquitinas/metabolismo
6.
Nat Commun ; 9(1): 4870, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30451821

RESUMO

RORγt controls the differentiation of TH17 cells, which are mediators of autoimmune conditions such as experimental autoimmune encephalomyelitis (EAE). RORγt also regulates thymocyte development and lymph node genesis. Here we show that the function of RORγt is regulated by its sumoylation. Loss of Sumo3, but not Sumo1, dampens TH17 differentiation and delays the progression of thymic CD8+ immature single-positive cells (ISPs). RORγt is SUMO3-modified by E3 ligase PIAS4 at lysine 31 (K31), and the mutation of K31 to arginine in mice prevents RORγt sumoylation, leading to impaired TH17 differentiation, resistance to TH17-mediated EAE, accumulation of thymic ISPs, and a lack of Peyer's patches. Mechanistically, sumoylation of RORγt-K31 recruits histone acetyltransferase KAT2A, which stabilizes the binding of SRC1 to enhance RORγt transcription factor activity. This study thus demonstrates that sumoylation is a critical mechanism for regulating RORγt function, and reveals new drug targets for preventing TH17-mediated autoimmunity.


Assuntos
Encefalomielite Autoimune Experimental/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Processamento de Proteína Pós-Traducional , Células Th17/imunologia , Timócitos/microbiologia , Timo/imunologia , Ubiquitinas/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Hematopoese/genética , Hematopoese/imunologia , Linfonodos/imunologia , Linfonodos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Coativador 1 de Receptor Nuclear/genética , Coativador 1 de Receptor Nuclear/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/patologia , Proteína SUMO-1/deficiência , Proteína SUMO-1/genética , Proteína SUMO-1/imunologia , Sumoilação , Células Th17/patologia , Timócitos/imunologia , Timócitos/patologia , Timo/patologia , Ubiquitinas/deficiência , Ubiquitinas/imunologia , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/imunologia
7.
Cytokine Growth Factor Rev ; 29: 3-16, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27157810

RESUMO

Since its discovery, SUMOylation has emerged as a key post-translational modification involved in the regulation of host-virus interactions. SUMOylation has been associated with the replication of a large number of viruses, either through the direct modification of viral proteins or through the modulation of cellular proteins implicated in antiviral defense. SUMO can affect protein function via covalent or non-covalent binding. There is growing evidence that SUMO regulates several host proteins involved in intrinsic and innate immunity, thereby contributing to the process governing interferon production during viral infection; as well as the interferon-activated Jak/STAT pathway. Unlike the interferon-mediated innate immune response, intrinsic antiviral resistance is mediated by constitutively expressed antiviral proteins (defined as restriction factors), which confer direct viral resistance through a variety of mechanisms. The aim of this review is to evaluate the role of SUMO in intrinsic and innate immunity; highlighting the involvement of the TRIM family proteins, with a specific focus on the mechanism through which SUMO affects i- interferon production upon viral infection, ii-interferon Jak/STAT signaling and biological responses, iii-the relationship between restriction factors and RNA viruses.


Assuntos
Imunidade Inata , Proteína SUMO-1/imunologia , Transdução de Sinais/imunologia , Viroses/imunologia , Animais , Humanos , Janus Quinases/imunologia , Fatores de Transcrição STAT/imunologia
8.
Sci Signal ; 8(400): ra107, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26508789

RESUMO

The Drosophila Toll pathway plays important roles in innate immune responses against Gram-positive bacteria and fungi. To identify previously uncharacterized components of this pathway, we performed comparative, ex vivo, genome-wide RNA interference screening. In four screens, we overexpressed the Toll adaptor protein dMyd88, the downstream kinase Pelle, or the nuclear factor κB (NF-κB) homolog Dif, or we knocked down Cactus, the Drosophila homolog of mammalian inhibitor of NF-κB. On the basis of these screens, we identified the E3 ubiquitin ligase Sherpa as being necessary for the activation of Toll signaling. A loss-of-function sherpa mutant fly exhibited compromised production of antimicrobial peptides and enhanced susceptibility to infection by Gram-positive bacteria. In cultured cells, Sherpa mediated ubiquitylation of dMyd88 and Sherpa itself, and Sherpa and Drosophila SUMO (small ubiquitin-like modifier) were required for the proper membrane localization of an adaptor complex containing dMyd88. These findings highlight a role for Sherpa in Drosophila host defense and suggest the SUMOylation-mediated regulation of dMyd88 functions in Toll innate immune signaling.


Assuntos
Proteínas de Drosophila/imunologia , Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , RNA Interferente Pequeno/imunologia , Receptores Toll-Like/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Estudo de Associação Genômica Ampla , Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , RNA Interferente Pequeno/genética , Proteína SUMO-1/genética , Proteína SUMO-1/imunologia , Receptores Toll-Like/genética , Ubiquitina-Proteína Ligases/genética
9.
Nat Commun ; 5: 4187, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24942926

RESUMO

Small ubiquitin-related modifier (SUMO) protein conjugation onto target proteins regulates multiple cellular functions, including defence against pathogens, stemness and senescence. SUMO1 peptides are limiting in quantity and are thus mainly conjugated to high-affinity targets. Conjugation of SUMO2/3 paralogues is primarily stress inducible and may initiate target degradation. Here we demonstrate that the expression of SUMO1/2/3 is dramatically enhanced by interferons through an miRNA-based mechanism involving the Lin28/let-7 axis, a master regulator of stemness. Normal haematopoietic progenitors indeed display much higher SUMO contents than their differentiated progeny. Critically, SUMOs contribute to the antiviral effects of interferons against HSV1 or HIV. Promyelocytic leukemia (PML) nuclear bodies are interferon-induced domains, which facilitate sumoylation of a subset of targets. Our findings thus identify an integrated interferon-responsive PML/SUMO pathway that impedes viral replication by enhancing SUMO conjugation and possibly also modifying the repertoire of targets. Interferon-enhanced post-translational modifications may be essential for senescence or stem cell self-renewal, and initiate SUMO-dependent proteolysis.


Assuntos
HIV-1/fisiologia , Herpesvirus Humano 1/fisiologia , MicroRNAs/imunologia , Proteínas de Ligação a RNA/imunologia , Proteína SUMO-1/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Ubiquitinas/imunologia , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/genética , Herpes Simples/genética , Herpes Simples/imunologia , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Humanos , Interferons/imunologia , MicroRNAs/genética , Proteínas de Ligação a RNA/genética , Proteína SUMO-1/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Ubiquitinas/genética , Replicação Viral
10.
Biochem Biophys Res Commun ; 447(1): 83-8, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24695317

RESUMO

RNF4, a SUMO-targeted ubiquitin ligase (STUbL), localizes to the nucleus and functions in the DNA damage response during interphase of the cell cycle. RNF4 also exists in cells undergoing mitosis, where its regulation and function remain poorly understood. Here we showed that administration of etoposide, an anticancer DNA topoisomerase II poison, to mitotic human cervical cancer HeLa cells induced SUMO-2/3-dependent localization of RNF4 to chromosomes. The FK2 antibody signals, indicative of poly/multi-ubiquitin assembly, were detected on etoposide-exposed mitotic chromosomes, whereas the signals were negligible in cells depleted for RNF4 by RNA interference. This suggests that RNF4 functions as a STUbL in the etoposide-induced damage response during mitosis. Indeed, RNF4-depletion sensitized mitotic HeLa cells to etoposide and increased cells with micronuclei. These results indicate the importance of the RNF4-mediated STUbL pathway during mitosis for the maintenance of chromosome integrity and further implicate RNF4 as a target for topo II poison-based therapy for cancer patients.


Assuntos
Cromossomos Humanos/metabolismo , Etoposídeo/farmacologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Reparo do DNA , DNA Topoisomerases Tipo II , Células HEK293 , Células HeLa , Humanos , Camundongos , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Mitose , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/imunologia , Proteína SUMO-1/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/imunologia
11.
Monoclon Antib Immunodiagn Immunother ; 32(5): 354-61, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24111868

RESUMO

Post-translational modification regulated by conjugation of a small ubiquitin-like modifier (SUMO) is involved in various cellular processes. In this study, we expressed and purified recombinant human SUMO-1 (hSUMO-1). BALB/c mice were immunized with a complex of hSUMO-1 protein and Lipoplex(O) to produce hSUMO-1-specific antibodies. Using conventional hybridoma technology, we obtained four hybridoma clones derived from the mouse with the highest antibody titer against hSUMO-1. Based on Western blot analysis, our hSUMO-1 monoclonal antibody specifically recognizes hSUMO-1, but not other SUMO proteins. These results support that the anti-hSUMO-1 monoclonal antibody produced with the aid of Lipoplex(O) adjuvant is specific and that Lipoplex(O) is useful for development of monoclonal antibodies against recombinant protein. In addition, we analyzed human tissues to examine the distribution of hSUMO-1. Higher expression of hSUMO-1 was detected in normal adrenal gland, esophagus, pancreas, liver, stomach, kidney, and uterus than in corresponding cancer tissues, suggesting a tumor suppressive function of hSUMO-1.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Hibridomas/imunologia , Proteínas Recombinantes/imunologia , Proteína SUMO-1/imunologia , Animais , Western Blotting , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Humanos , Imunização , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Análise em Microsséries
12.
Biochimie ; 92(10): 1306-14, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20561559

RESUMO

SUMO works in a similar way as ubiquitin to alter the biological properties of a target protein by conjugation. The homologous gene of SUMO named BmSmt3 was identified for the first time in silkworm. The expression of BmSmt3 was enhanced in the fat body of silkworm after immune challenge. However, the expression of BmSmt3 after immune challenge was almost invariant in silk gland, which is the nonimmune organ in silkworm. In addition, the expression of BmRelA and CecropinB1 was decreased significantly in pupae after the BmSmt3 was knocked down in vivo. According to our results, BmSmt3 might participate in the immune response through regulating the expression of BmRelA gene, which can further regulate the expression of antibacterial peptide subsequently in silkworm.


Assuntos
Bombyx/imunologia , Proteínas de Insetos/imunologia , Proteína SUMO-1/imunologia , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Regulação da Expressão Gênica/imunologia , Imunidade , Fator de Transcrição RelA/genética
13.
Fish Shellfish Immunol ; 29(2): 233-40, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20382231

RESUMO

B-cell activating factor (BAFF), belonging to the TNF family, is a critical cytokine for B-cell survival, proliferation, maturation and differentiation. In the present study we cloned the cDNA of zebrafish (Danio rerio) BAFF (designated zBAFF) by reverse transcription-PCR (RT-PCR). The open reading frame (ORF) of zBAFF consists of 807 bases encoding a protein of 268 amino acids. The deduced amino acid sequence of its cDNA possessed the TNF family signature, a transmembrane domain, and three cysteine residues, which are the typical characteristics of TNF gene in mammals and birds. Phylogenetic analysis exhibits the highest identity score 67.6, 61.4 and 66.9% with the rainbow trout, tetraodon and salmon counterparts, respectively. The identity to avian and mammalian BAFFs ranges from 49.7 to 53.8%. Recombinant soluble zBAFF (zsBAFF) was fused with a small ubiquitin-related modifier gene (SUMO) to enhance the soluble expression level in Escherichia coli BL21 (DE3). The resulting fused protein SUMO-zsBAFF was highly expressed in BL21 (DE3) with a molecular weight of 38 kDa. The fusing protein was purified using metal chellate affinity chromatography (Ni-NTA) and cleaved by a SUMO-specific protease, then confirmed by SDS-PAGE and Western blotting analysis. In vitro, the MTT assay indicated that the purified zsBAFF as well as SUMO-zsBAFF proteins were able to promote spleen lymphocyte survival in a dose-dependent manner also to co-stimulate the proliferation of mammalian B-cells with anti-IgM. Thus, the fusion protein represents a readily obtainable source of biologically active zsBAFF that may prove useful in further studies on zebrafish BAFF and its receptors.


Assuntos
Fator Ativador de Células B/genética , Fator Ativador de Células B/imunologia , Regulação da Expressão Gênica , Filogenia , Peixe-Zebra/classificação , Peixe-Zebra/fisiologia , Sequência de Aminoácidos , Animais , Fator Ativador de Células B/química , Sequência de Bases , Células Cultivadas , Clonagem Molecular , Perfilação da Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Recombinantes/imunologia , Proteína SUMO-1/imunologia , Alinhamento de Sequência , Peixe-Zebra/genética , Peixe-Zebra/imunologia
14.
J Immunol ; 183(2): 1110-9, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19553542

RESUMO

It is not clear why the development of protective Th2 cells is poor in type 1 diabetes (T1D). c-Maf transactivates the IL-4 gene promoting Th2 cell development; therefore, abnormalities in c-Maf may contribute to reduced IL-4 production by CD4 cells from nonobese diabetic (NOD) mice. In this study we demonstrate that despite normal expression, c-Maf binds poorly to the IL-4 promoter (IL-4p) in NOD CD4 cells. Immunoblotting demonstrates that c-Maf can be modified at lysine 33 by SUMO-1 (small ubiquitin-like modifier 1). Sumoylation is facilitated by direct interaction with the E2-conjugating enzyme Ubc9 and increases following T cell stimulation. In transfected cells, sumoylation decreases c-Maf transactivation of IL-4p-driven luciferase reporter activity, reduces c-Maf binding to the IL-4p in chromatin immunoprecipitation assays, and enhances c-Maf localization into promyelocytic leukemia nuclear bodies. Sumoylation of c-Maf is increased in NOD CD4 cells as compared with CD4 cells from diabetes-resistant B10.D2 mice, suggesting that increased c-Maf sumoylation contributes to immune deviation in T1D by reducing c-Maf access to and transactivation of the IL-4 gene.


Assuntos
Interleucina-4/genética , Proteínas Proto-Oncogênicas c-maf/fisiologia , Proteína SUMO-1/imunologia , Ativação Transcricional/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Imunidade , Leucemia Mieloide/patologia , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-maf/metabolismo
15.
Apoptosis ; 14(4): 447-54, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19199037

RESUMO

Apoptosis plays a pivotal role in tissue homoeostasis both under physiological and pathological conditions and several studies have shown that some characteristic changes in the composition and structure of the inflamed synovial membrane in rheumatoid arthritis (RA) are linked to an altered apoptotic response of synovial cells. As a result, a hyperplastic synovial tissue is generated that mediates the progressive destruction of articular cartilage and bone. In addition to inflammatory cells, these changes most prominently affect resident fibroblast-like cells that have been demonstrated to be of utmost importance for joint destruction. Once activated, these cells pass through prominent molecular changes resulting in an aggressive, invasive behaviour. Research of the past years has identified different mechanisms that prevent synovial cells in RA from apoptosis. They include changes in the mitochondrial pathway as well as altered expression of downstream modulators of death receptors and transcriptional regulators such as NFkappaB. This review summarises our recent progress in understanding aberrant apoptosis in the RA synovial membrane and points to possibilities of intervening specifically with this aspect of the pathogenesis of RA.


Assuntos
Apoptose/genética , Artrite Reumatoide/patologia , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Morte Celular/genética , Fibroblastos/imunologia , Fibroblastos/patologia , Humanos , Modelos Imunológicos , Proteína SUMO-1/imunologia , Membrana Sinovial/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Receptor fas/imunologia
16.
J Immunol ; 181(8): 5646-52, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832723

RESUMO

Efficient clearance of apoptotic cells (AC) by professional phagocytes is crucial for tissue homeostasis and resolution of inflammation. Macrophages respond to AC with an increase in antiinflammatory cytokine production but a diminished release of proinflammatory mediators. Mechanisms to explain attenuated proinflammatory cytokine formation remain elusive. We provide evidence that peroxisome proliferator-activated receptor gamma (PPARgamma) coordinates antiinflammatory responses following its activation by AC. Exposing murine RAW264.7 macrophages to AC before LPS stimulation reduced NF-kappaB transactivation and lowered target gene expression of, that is, TNF-alpha and IL-6 compared with controls. In macrophages overexpressing a dominant negative mutant of PPARgamma, NF-kappaB transactivation in response to LPS was restored, while macrophages from myeloid lineage-specific conditional PPARgamma knockout mice proved that PPARgamma transmitted an antiinflammatory response, which was delivered by AC. Expressing a PPARgamma-Delta aa32-250 deletion mutant, we observed no inhibition of NF-kappaB. Analyzing the PPARgamma domain structures within aa 32-250, we anticipated PPARgamma sumoylation in mediating the antiinflammatory effect in response to AC. Interfering with sumoylation of PPARgamma by mutating the predicted sumoylation site (K77R), or knockdown of the small ubiquitin-like modifier (SUMO) E3 ligase PIAS1 (protein inhibitor of activated STAT1), eliminated the ability of AC to suppress NF-kappaB. Chromatin immunoprecipitation analysis demonstrated that AC prevented the LPS-induced removal of nuclear receptor corepressor (NCoR) from the kappaB site within the TNF-alpha promoter. We conclude that AC induce PPARgamma sumoylation to attenuate the removal of NCoR, thereby blocking transactivation of NF-kappaB. This contributes to an antiinflammatory phenotype shift in macrophages responding to AC by lowering proinflammatory cytokine production.


Assuntos
Apoptose/imunologia , Mediadores da Inflamação/imunologia , Interleucina-6/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , NF-kappa B/imunologia , Proteínas Nucleares/imunologia , PPAR gama/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Proteínas Repressoras/imunologia , Proteína SUMO-1/imunologia , Fator de Necrose Tumoral alfa/imunologia , Sequência de Aminoácidos/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/biossíntese , Células Jurkat , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Correpressor 1 de Receptor Nuclear , PPAR gama/genética , PPAR gama/metabolismo , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Inibidoras de STAT Ativados/imunologia , Proteínas Inibidoras de STAT Ativados/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , Elementos de Resposta/imunologia , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Deleção de Sequência/genética , Deleção de Sequência/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Ativação Transcricional/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
17.
J Immunol ; 180(9): 5983-90, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18424718

RESUMO

The AP-1 family member JunB is a critical regulator of T cell function. JunB is a transcriptional activator of various cytokine genes, such as IL-2, IL-4, and IL-10; however, the post-translational modifications that regulate JunB activity in T cells are poorly characterized. We show here that JunB is conjugated with small ubiquitin-like modifier (SUMO) on lysine 237 in resting and activated primary T cells and T cell lines. Sumoylated JunB associated with the chromatin-containing insoluble fraction of cells, whereas nonsumoylated JunB was also in the soluble fraction. Blocking JunB sumoylation by mutation or use of a dominant-negative form of the SUMO-E2 Ubc-9 diminished its ability to transactivate IL-2 and IL-4 reporter genes. In contrast, nonsumoylable JunB mutants showed unimpaired activity with reporter genes controlled by either synthetic 12-O-tetradecanoylphorbol-13-acetate response elements or NF-AT/AP-1 and CD28RE sites derived from the IL-2 promoter. Ectopic expression of JunB in activated human primary CD4(+) T cells induced activation of the endogenous IL-2 promoter, whereas the nonsumoylable JunB mutant did not. Thus, our work demonstrates that sumoylation of JunB regulates its ability to induce cytokine gene transcription and likely plays a critical role in T cell activation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Ativação Linfocitária/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Proteínas Proto-Oncogênicas c-jun/imunologia , Proteína SUMO-1/imunologia , Transcrição Gênica/imunologia , Linfócitos T CD4-Positivos/metabolismo , Cromatina/genética , Cromatina/imunologia , Cromatina/metabolismo , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Humanos , Células Jurkat , Ativação Linfocitária/genética , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/imunologia , Fatores de Transcrição NFATC/metabolismo , Processamento de Proteína Pós-Traducional/genética , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Elementos de Resposta/genética , Elementos de Resposta/imunologia , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/imunologia , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/genética , Ativação Transcricional/genética , Ativação Transcricional/imunologia , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/imunologia , Enzimas de Conjugação de Ubiquitina/metabolismo
18.
Z Rheumatol ; 66(3): 239-40, 242, 2007 May.
Artigo em Alemão | MEDLINE | ID: mdl-17436005

RESUMO

Apoptosis is a central physiological mechanism for maintaining cellular stability in tissue. Synovial fibroblasts, which play a central role in the pathogenesis of rheumatoid arthritis (RA), show a resistance to apoptosis. Several molecular mechanisms are involved in such resistance. Thus, soluble Fas can bind Fas ligands (Fas-L) and hinder Fas-L induced apoptosis in fibroblasts. SUMO-1 (a small ubiquitin-like modifier) attaches to proteins post-translationally. This appears to be significantly involved in apoptosis resistance in RA fibroblasts. SUMO-1 levels are substantially increased in synovial fibroblasts from RA patients. A change in the post-translational SUMOlation pattern could represent a new target for changing the stable activation of synovial fibroblasts in RA.


Assuntos
Apoptose/imunologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Fibroblastos/imunologia , Modelos Imunológicos , Proteína SUMO-1/imunologia , Membrana Sinovial/imunologia , Animais , Humanos , Membrana Sinovial/patologia
19.
Hepatology ; 41(3): 609-16, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15726652

RESUMO

Serum autoantibodies against components of nuclear dots (anti-NDs), namely PML and Sp100, are specifically detected in 20% to 30% of patients with primary biliary cirrhosis (PBC). Although anti-ND antibodies are nonpathogenic, the mechanisms that lead to this unique reactivity are critical to understanding the loss of immune tolerance in PBC. Importantly, Sp100 and PML are both covalently linked to small ubiquitin-related modifiers (SUMOs). Therefore, we investigated whether SUMO proteins are independent autoantigens in PBC and studied 99 PBC sera samples for reactivity against NDs, PML, and Sp100, as well as against SUMO-2 and SUMO-1 recombinant proteins. Autoantibodies against SUMO-2 and SUMO-1 were found in 42% and 15% of anti-ND-positive PBC sera, respectively. Anti-SUMO reactivity was not observed in anti-ND-negative sera. Anti-SUMO-2 autoantibodies were found in 58% of sera containing autoantibodies against both PML and Sp100 and were detected exclusively in sera containing anti-Sp100 autoantibodies. In conclusion, SUMO proteins constitute a novel and independent class of autoantigens in PBC. Furthermore, we believe our data emphasize the post-translational modification of lysine by either lipoylation in the case of AMA or SUMOylation in the case of specific anti-ND autoantibodies as the pivotal site for autoantibody generation in PBC.


Assuntos
Autoantígenos/imunologia , Cirrose Hepática Biliar/imunologia , Proteína SUMO-1/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Animais , Anticorpos Antinucleares/sangue , Antígenos Nucleares/imunologia , Autoanticorpos/sangue , Galinhas , Imunofluorescência , Humanos , Immunoblotting , Mitocôndrias/imunologia , Proteínas de Neoplasias/imunologia , Proteínas Nucleares/imunologia , Proteína da Leucemia Promielocítica , Proteína SUMO-1/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Fatores de Transcrição/imunologia , Proteínas Supressoras de Tumor
20.
Tsitologiia ; 43(12): 1123-9, 2001.
Artigo em Russo | MEDLINE | ID: mdl-11881150

RESUMO

Nuclear domains 10 (ND10) were first detected occasionally using antibodies to an antigen of unknown nature (Ascoli, Maul, 1991). Further on it was shown that ND10 were sites of locality of the number of proteins (PML, Sp 100, pRB) (Sterndorf et al., 1992; Kamitani et al., 1998), the majority of which are modified with ubiquitin-like small proteins-modifiers (SUMO) (Ishov, Maul, 1996). In addition, it was shown that ND10 were sites of primary localization, transcription and replication of some DNA-viruses (SV40, virus of simple herpes 1, adenovirus 5) (Ishov, Maul, 1996; Ishov et al., 1997). Except for SV40, these viruses produce proteins able to modify ND10, or leading to degradation of ND10-associated proteins (Maul et al., 1993; Maul, Everett, 1994). This degradation is accompanied with protein desumofication and, later, with hydrolysis on the ubiquitin-proteosomal way (Everett et al., 1998, 1999). Cell incubation with interferon leads to augmentation of the number and dimension of ND10 owing to increased expression of Sp100 and PML (Lavau et al., 1995; Grotzinger et al., 1996). In all, these data make it possible to put forward a hypothesis that ND10 may represent a peculiar cell storage ("dépôt") of proteins regulated according to the "accumulation-drop" principle (Ishov et al., 1997; Maul, 1998). However, this hypothesis requires further factual grounds.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Antígenos Nucleares , Proteínas de Transporte/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares/imunologia , Proteína SUMO-1/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Western Blotting , Proteínas de Transporte/genética , Linhagem Celular , Proteínas Correpressoras , Feminino , Humanos , Hibridomas , Camundongos , Camundongos Endogâmicos BALB C , Chaperonas Moleculares , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase , Testes de Precipitina , Proteína da Leucemia Promielocítica , Proteína SUMO-1/genética , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...