Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Front Microbiol ; 15: 1441495, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39296289

RESUMO

Introduction: The severity of Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is often dictated by a range of comorbidities. A considerable literature suggests iron deficiency and iron overload may contribute to increased infection, inflammation and disease severity, although direct causal relationships have been difficult to establish. Methods: Here we generate iron deficient and iron loaded C57BL/6 J mice by feeding standard low and high iron diets, with mice on a normal iron diet representing controls. All mice were infected with a primary SARS-CoV-2 omicron XBB isolate and lung inflammatory responses were analyzed by histology, immunohistochemistry and RNA-Seq. Results: Compared with controls, iron deficient mice showed no significant changes in lung viral loads or histopathology, whereas, iron loaded mice showed slightly, but significantly, reduced lung viral loads and histopathology. Transcriptional changes were modest, but illustrated widespread dysregulation of inflammation signatures for both iron deficient vs. controls, and iron loaded vs. controls. Some of these changes could be associated with detrimental outcomes, whereas others would be viewed as beneficial. Discussion: Diet-associated iron deficiency or overload thus induced modest modulations of inflammatory signatures, but no significant histopathologically detectable disease exacerbations.

2.
Vaccine ; 42(25): 126136, 2024 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-39004524

RESUMO

Getah virus (GETV) is an emerging mosquito-borne virus with economic impact on the livestock industry in East Asia. In this study, we successfully produced GETV virus-like particles (VLPs) in insect cells using the baculovirus expression vector system. We show that the GETV envelope glycoproteins were successfully expressed at the surface of the insect cell and were glycosylated. VLPs were isolated from the culture fluid as enveloped particles of 60-80 nm in diameter. Two 1 µg vaccinations with this GETV VLP vaccine, without adjuvant, generated neutralizing antibody responses and protected wild-type C57/BL6 mice against GETV viremia and arthritic disease. The GETV VLP vaccine may find application as a horse and/or pig vaccine in the future.


Assuntos
Alphavirus , Anticorpos Neutralizantes , Anticorpos Antivirais , Artrite , Camundongos Endogâmicos C57BL , Vacinas de Partículas Semelhantes a Vírus , Viremia , Animais , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Viremia/prevenção & controle , Viremia/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Camundongos , Artrite/imunologia , Artrite/prevenção & controle , Alphavirus/imunologia , Alphavirus/genética , Infecções por Alphavirus/prevenção & controle , Infecções por Alphavirus/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Feminino , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Baculoviridae/genética , Baculoviridae/imunologia , Células Sf9
3.
PLoS One ; 17(7): e0270584, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35881628

RESUMO

Granzyme K (GzmK) is a tryptic member of the granzyme family of chymotrypsin-like serine proteases produced by cells of the immune system. Previous studies have indicated that GzmK activates protease-activated receptor 1 (PAR1) enhancing activation of monocytes and wound healing in endothelial cells. Here, we show using peptides and full length proteins that GzmK and, to a lesser extent the related protease GzmA, are capable of activating PAR1 and PAR2. These cleavage events occur at the canonical arginine P1 residue and involve exosite interactions between protease and receptor. Despite cleaving PAR2 at the same point as trypsin, GzmK does not induce a classical Ca2+ flux but instead activates a distinct signalling cascade, involving recruitment of ß-arrestin and phosphorylation of ERK. In epithelial A549 cells, PAR2 activation by GzmK results in the release of inflammatory cytokines IL-6 and IL-8. These data suggest that during an immune response GzmK acts as a pro-inflammatory regulator, rather than as a cytotoxin.


Assuntos
Receptor PAR-1 , Receptor PAR-2 , Endopeptidases/metabolismo , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Granzimas/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo
4.
ACS Cent Sci ; 8(5): 527-545, 2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35647275

RESUMO

Heparan sulfate (HS) is a cell surface polysaccharide recently identified as a coreceptor with the ACE2 protein for the S1 spike protein on SARS-CoV-2 virus, providing a tractable new therapeutic target. Clinically used heparins demonstrate an inhibitory activity but have an anticoagulant activity and are supply-limited, necessitating alternative solutions. Here, we show that synthetic HS mimetic pixatimod (PG545), a cancer drug candidate, binds and destabilizes the SARS-CoV-2 spike protein receptor binding domain and directly inhibits its binding to ACE2, consistent with molecular modeling identification of multiple molecular contacts and overlapping pixatimod and ACE2 binding sites. Assays with multiple clinical isolates of SARS-CoV-2 virus show that pixatimod potently inhibits the infection of monkey Vero E6 cells and physiologically relevant human bronchial epithelial cells at safe therapeutic concentrations. Pixatimod also retained broad potency against variants of concern (VOC) including B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 (Delta), and B.1.1.529 (Omicron). Furthermore, in a K18-hACE2 mouse model, pixatimod significantly reduced SARS-CoV-2 viral titers in the upper respiratory tract and virus-induced weight loss. This demonstration of potent anti-SARS-CoV-2 activity tolerant to emerging mutations establishes proof-of-concept for targeting the HS-Spike protein-ACE2 axis with synthetic HS mimetics and provides a strong rationale for clinical investigation of pixatimod as a potential multimodal therapeutic for COVID-19.

5.
mBio ; 12(5): e0181321, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34634927

RESUMO

Vaccines pave the way out of the SARS-CoV-2 pandemic. Besides mRNA and adenoviral vector vaccines, effective protein-based vaccines are needed for immunization against current and emerging variants. We have developed a virus-like particle (VLP)-based vaccine using the baculovirus-insect cell expression system, a robust production platform known for its scalability, low cost, and safety. Baculoviruses were constructed encoding SARS-CoV-2 spike proteins: full-length S, stabilized secreted S, or the S1 domain. Since subunit S only partially protected mice from SARS-CoV-2 challenge, we produced S1 for conjugation to bacteriophage AP205 VLP nanoparticles using tag/catcher technology. The S1 yield in an insect-cell bioreactor was ∼11 mg/liter, and authentic protein folding, efficient glycosylation, partial trimerization, and ACE2 receptor binding was confirmed. Prime-boost immunization of mice with 0.5 µg S1-VLPs showed potent neutralizing antibody responses against Wuhan and UK/B.1.1.7 SARS-CoV-2 variants. This two-component nanoparticle vaccine can now be further developed to help alleviate the burden of COVID-19. IMPORTANCE Vaccination is essential to reduce disease severity and limit the transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Protein-based vaccines are useful to vaccinate the world population and to boost immunity against emerging variants. Their safety profiles, production costs, and vaccine storage temperatures are advantageous compared to mRNA and adenovirus vector vaccines. Here, we use the versatile and scalable baculovirus expression vector system to generate a two-component nanoparticle vaccine to induce potent neutralizing antibody responses against SARS-CoV-2 variants. These nanoparticle vaccines can be quickly adapted as boosters by simply updating the antigen component.


Assuntos
Anticorpos Neutralizantes/metabolismo , Nanopartículas/metabolismo , SARS-CoV-2/metabolismo , Animais , COVID-19/imunologia , Feminino , Glicosilação , Camundongos , Camundongos Endogâmicos BALB C , SARS-CoV-2/imunologia , Células Sf9 , Vacinas Virais/imunologia
6.
PLoS Pathog ; 17(7): e1009788, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34310650

RESUMO

Zika virus (ZIKV) strains are classified into the African and Asian genotypes. The higher virulence of the African MR766 strain, which has been used extensively in ZIKV research, in adult IFNα/ß receptor knockout (IFNAR-/-) mice is widely viewed as an artifact associated with mouse adaptation due to at least 146 passages in wild-type suckling mouse brains. To gain insights into the molecular determinants of MR766's virulence, a series of genes from MR766 were swapped with those from the Asian genotype PRVABC59 isolate, which is less virulent in IFNAR-/- mice. MR766 causes 100% lethal infection in IFNAR-/- mice, but when the prM gene of MR766 was replaced with that of PRVABC59, the chimera MR/PR(prM) showed 0% lethal infection. The reduced virulence was associated with reduced neuroinvasiveness, with MR766 brain titers ≈3 logs higher than those of MR/PR(prM) after subcutaneous infection, but was not significantly different in brain titers of MR766 and MR/PR(prM) after intracranial inoculation. MR/PR(prM) also showed reduced transcytosis when compared with MR766 in vitro. The high neuroinvasiveness of MR766 in IFNAR-/- mice could be linked to the 10 amino acids that differ between the prM proteins of MR766 and PRVABC59, with 5 of these changes affecting positive charge and hydrophobicity on the exposed surface of the prM protein. These 10 amino acids are highly conserved amongst African ZIKV isolates, irrespective of suckling mouse passage, arguing that the high virulence of MR766 in adult IFNAR-/- mice is not the result of mouse adaptation.


Assuntos
Proteínas do Envelope Viral/genética , Virulência/genética , Infecção por Zika virus/virologia , Zika virus/genética , Zika virus/patogenicidade , Animais , Barreira Hematoencefálica , Permeabilidade Capilar , Genótipo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Zika virus/metabolismo
7.
PLoS Biol ; 19(4): e3001201, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33872300

RESUMO

Most vertebrate RNA viruses show pervasive suppression of CpG and UpA dinucleotides, closely resembling the dinucleotide composition of host cell transcriptomes. In contrast, CpG suppression is absent in both invertebrate mRNA and RNA viruses that exclusively infect arthropods. Arthropod-borne (arbo) viruses are transmitted between vertebrate hosts by invertebrate vectors and thus encounter potentially conflicting evolutionary pressures in the different cytoplasmic environments. Using a newly developed Zika virus (ZIKV) model, we have investigated how demands for CpG suppression in vertebrate cells can be reconciled with potentially quite different compositional requirements in invertebrates and how this affects ZIKV replication and transmission. Mutant viruses with synonymously elevated CpG or UpA dinucleotide frequencies showed attenuated replication in vertebrate cell lines, which was rescued by knockout of the zinc-finger antiviral protein (ZAP). Conversely, in mosquito cells, ZIKV mutants with elevated CpG dinucleotide frequencies showed substantially enhanced replication compared to wild type. Host-driven effects on virus replication attenuation and enhancement were even more apparent in mouse and mosquito models. Infections with CpG- or UpA-high ZIKV mutants in mice did not cause typical ZIKV-induced tissue damage and completely protected mice during subsequent challenge with wild-type virus, which demonstrates their potential as live-attenuated vaccines. In contrast, the CpG-high mutants displayed enhanced replication in Aedes aegypti mosquitoes and a larger proportion of mosquitoes carried infectious virus in their saliva. These findings show that mosquito cells are also capable of discriminating RNA based on dinucleotide composition. However, the evolutionary pressure on the CpG dinucleotides of viral genomes in arthropod vectors directly opposes the pressure present in vertebrate host cells, which provides evidence that an adaptive compromise is required for arbovirus transmission. This suggests that the genome composition of arbo flaviviruses is crucial to maintain the balance between high-level replication in the vertebrate host and persistent replication in the mosquito vector.


Assuntos
Evolução Molecular , Genoma Viral/genética , Interações Hospedeiro-Patógeno/genética , Zika virus/genética , Células A549 , Aedes/virologia , Animais , Composição de Bases/fisiologia , Sequência de Bases/genética , Linhagem Celular , Chlorocebus aethiops , Ilhas de CpG/fisiologia , Fosfatos de Dinucleosídeos/análise , Fosfatos de Dinucleosídeos/genética , Adaptação ao Hospedeiro/genética , Humanos , Masculino , Mamíferos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mosquitos Vetores/genética , Mosquitos Vetores/virologia , RNA Viral/química , RNA Viral/genética , Seleção Genética/fisiologia , Células Vero , Infecção por Zika virus/genética , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia
8.
NPJ Vaccines ; 5(1): 44, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32550013

RESUMO

The Sementis Copenhagen Vector (SCV) is a new vaccinia virus-derived, multiplication-defective, vaccine technology assessed herein in non-human primates. Indian rhesus macaques (Macaca mulatta) were vaccinated with a multi-pathogen recombinant SCV vaccine encoding the structural polyproteins of both Zika virus (ZIKV) and chikungunya virus (CHIKV). After one vaccination, neutralising antibody responses to ZIKV and four strains of CHIKV, representative of distinct viral genotypes, were generated. A second vaccination resulted in significant boosting of neutralising antibody responses to ZIKV and CHIKV. Following challenge with ZIKV, SCV-ZIKA/CHIK-vaccinated animals showed significant reductions in viremias compared with animals that had received a control SCV vaccine. Two SCV vaccinations also generated neutralising and IgG ELISA antibody responses to vaccinia virus. These results demonstrate effective induction of immunity in non-human primates by a recombinant SCV vaccine and illustrates the utility of SCV as a multi-disease vaccine platform capable of delivering multiple large immunogens.

9.
PLoS Pathog ; 15(6): e1007880, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31211814

RESUMO

The largest ever recorded epidemic of the Chikungunya virus (CHIKV) broke out in 2004 and affected four continents. Acute symptomatic infections are typically associated with the onset of fever and often debilitating polyarthralgia/polyarthritis. In this study, a systems biology approach was adopted to analyze the blood transcriptomes of adults acutely infected with the CHIKV. Gene signatures that were associated with viral RNA levels and the onset of symptoms were identified. Among these genes, the putative role of the Eukaryotic Initiation Factor (eIF) family genes and apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC3A) in the CHIKV replication process were displayed. We further compared these signatures with signatures induced by the Dengue virus infection and rheumatoid arthritis. Finally, we demonstrated that the CHIKV in vitro infection of murine bone marrow-derived macrophages induced IL-1 beta production in a mechanism that is significantly dependent on the inflammasome NLRP3 activation. The observations provided valuable insights into virus-host interactions during the acute phase and can be instrumental in the investigation of new and effective therapeutic interventions.


Assuntos
Artrite/imunologia , Febre de Chikungunya/imunologia , Vírus Chikungunya/fisiologia , Citidina Desaminase/imunologia , Proteínas/imunologia , Replicação Viral/imunologia , Adulto , Animais , Artrite/patologia , Artrite/virologia , Febre de Chikungunya/patologia , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Feminino , Febre/imunologia , Febre/patologia , Febre/virologia , Seguimentos , Humanos , Interleucina-1beta/imunologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
10.
Nat Microbiol ; 4(5): 876-887, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30886357

RESUMO

Arboviruses cycle between, and replicate in, both invertebrate and vertebrate hosts, which for Zika virus (ZIKV) involves Aedes mosquitoes and primates1. The viral determinants required for replication in such obligate hosts are under strong purifying selection during natural virus evolution, making it challenging to resolve which determinants are optimal for viral fitness in each host. Herein we describe a deep mutational scanning (DMS) strategy2-5 whereby a viral cDNA library was constructed containing all codon substitutions in the C-terminal 204 amino acids of ZIKV envelope protein (E). The cDNA library was transfected into C6/36 (Aedes) and Vero (primate) cells, with subsequent deep sequencing and computational analyses of recovered viruses showing that substitutions K316Q and S461G, or Q350L and T397S, conferred substantial replicative advantages in mosquito and primate cells, respectively. A 316Q/461G virus was constructed and shown to be replication-defective in mammalian cells due to severely compromised virus particle formation and secretion. The 316Q/461G virus was also highly attenuated in human brain organoids, and illustrated utility as a vaccine in mice. This approach can thus imitate evolutionary selection in a matter of days and identify amino acids key to the regulation of virus replication in specific host environments.


Assuntos
Análise Mutacional de DNA/métodos , Tropismo Viral , Infecção por Zika virus/virologia , Zika virus/fisiologia , Aedes/virologia , Animais , Evolução Biológica , Chlorocebus aethiops , Feminino , Especificidade de Hospedeiro , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Mosquitos Vetores/virologia , Mutação , Seleção Genética , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Zika virus/química , Zika virus/genética
11.
Nat Commun ; 9(1): 1230, 2018 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-29581442

RESUMO

Zika and chikungunya viruses have caused major epidemics and are transmitted by Aedes aegypti and/or Aedes albopictus mosquitoes. The "Sementis Copenhagen Vector" (SCV) system is a recently developed vaccinia-based, multiplication-defective, vaccine vector technology that allows manufacture in modified CHO cells. Herein we describe a single-vector construct SCV vaccine that encodes the structural polyprotein cassettes of both Zika and chikungunya viruses from different loci. A single vaccination of mice induces neutralizing antibodies to both viruses in wild-type and IFNAR-/- mice and protects against (i) chikungunya virus viremia and arthritis in wild-type mice, (ii) Zika virus viremia and fetal/placental infection in female IFNAR-/- mice, and (iii) Zika virus viremia and testes infection and pathology in male IFNAR-/- mice. To our knowledge this represents the first single-vector construct, multi-pathogen vaccine encoding large polyproteins, and offers both simplified manufacturing and formulation, and reduced "shot burden" for these often co-circulating arboviruses.


Assuntos
Febre de Chikungunya/prevenção & controle , Vírus Chikungunya/imunologia , Vetores Genéticos , Vaccinia virus/genética , Vacinas Virais/genética , Vacinas Virais/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Células CHO , Febre de Chikungunya/imunologia , Chlorocebus aethiops , Cricetulus , Ensaio de Imunoadsorção Enzimática , Feminino , Células HeLa , Humanos , Masculino , Troca Materno-Fetal , Camundongos Endogâmicos C57BL , Gravidez , Receptor de Interferon alfa e beta/genética , Células Vero , Vacinas Virais/administração & dosagem , Infecção por Zika virus/imunologia
12.
Nat Microbiol ; 2(10): 1435-1445, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28848230

RESUMO

Mosquito-borne viruses can cause severe inflammatory diseases and there are limited therapeutic solutions targeted specifically at virus-induced inflammation. Chikungunya virus (CHIKV), a re-emerging alphavirus responsible for several outbreaks worldwide in the past decade, causes debilitating joint inflammation and severe pain. Here, we show that CHIKV infection activates the NLRP3 inflammasome in humans and mice. Peripheral blood mononuclear cells isolated from CHIKV-infected patients showed elevated NLRP3, caspase-1 and interleukin-18 messenger RNA expression and, using a mouse model of CHIKV infection, we found that high NLRP3 expression was associated with peak inflammatory symptoms. Inhibition of NLRP3 activation using the small-molecule inhibitor MCC950 resulted in reduced CHIKV-induced inflammation and abrogated osteoclastogenic bone loss and myositis, but did not affect in vivo viral replication. Mice treated with MCC950 displayed lower expression levels of the cytokines interleukin-6, chemokine ligand 2 and tumour necrosis factor in joint tissue. Interestingly, MCC950 treatment abrogated disease signs in mice infected with a related arthritogenic alphavirus, Ross River virus, but not in mice infected with West Nile virus-a flavivirus. Here, using mouse models of alphavirus-induced musculoskeletal disease, we demonstrate that NLRP3 inhibition in vivo can reduce inflammatory pathology and that further development of therapeutic solutions targeting inflammasome function could help treat arboviral diseases.


Assuntos
Alphavirus/imunologia , Febre de Chikungunya/tratamento farmacológico , Vírus Chikungunya/efeitos dos fármacos , Inflamassomos/farmacologia , Inflamação , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Alphavirus/efeitos dos fármacos , Animais , Caspase 1 , Quimiocinas/metabolismo , Febre de Chikungunya/patologia , Chlorocebus aethiops , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Leucócitos Mononucleares/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miosite/patologia , RNA Mensageiro/metabolismo , Ross River virus/efeitos dos fármacos , Células Vero , Vírus do Nilo Ocidental/efeitos dos fármacos
13.
PLoS One ; 11(4): e0153975, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27100888

RESUMO

Ingenol mebutate is approved for the topical treatment of actinic keratoses and may ultimately also find utility in treating skin cancers. Here we show that relapse rates of subcutaneous B16 melanoma tumours treated topically with ingenol mebutate were not significantly different in C57BL/6 and Rag1-/- mice, suggesting B and T cells do not play a major role in the anti-cancer efficacy of ingenol mebutate. Relapse rates were, however, significantly increased in MyD88-/- mice and in C57BL/6 mice treated with the anti-IL-1 agent, anakinra. Ingenol mebutate treatment induces a pronounced infiltration of neutrophils, which have been shown to have anti-cancer activity in mice. Herein we provide evidence that IL-1 promotes neutrophil recruitment to the tumour, decreases apoptosis of infiltrating neutrophils and increases neutrophil tumour killing activity. These studies suggest IL-1, via its action on neutrophils, promotes the anti-cancer efficacy of ingenol mebutate, with ingenol mebutate treatment causing both IL-1ß induction and IL-1α released from keratinocytes.


Assuntos
Antineoplásicos/uso terapêutico , Diterpenos/uso terapêutico , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Interleucina-1/imunologia , Melanoma/tratamento farmacológico , Animais , Feminino , Deleção de Genes , Imunidade Celular/efeitos dos fármacos , Melanoma/genética , Melanoma/imunologia , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/imunologia
14.
PLoS One ; 10(6): e0130136, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26083412

RESUMO

SerpinB2 (PAI-2), a member of the clade B family of serine protease inhibitors, is one of the most upregulated proteins following cellular stress. Originally described as an inhibitor of urokinase plasminogen activator, its predominant cytoplasmic localisation suggests an intracellular function. SerpinB2 has been reported to display cytoprotective properties in neurons and to interact with intracellular proteins including components of the ubiquitin-proteasome system (UPS). In the current study we explored the potential role of SerpinB2 as a modulator of proteotoxic stress. Initially, we transiently transfected wild-type SerpinB2 and SerpinB2-/- murine embryonic fibroblasts (MEFs) with Huntingtin exon1-polyglutamine (fused C-terminally to mCherry). Inclusion body formation as result of Huntingtin aggregation was evident in the SerpinB2 expressing cells but significantly impaired in the SerpinB2-/- cells, the latter concomitant with loss in cell viability. Importantly, recovery of the wild-type phenotype and cell viability was rescued by retroviral transduction of SerpinB2 expression. SerpinB2 modestly attenuated Huntingtin and amyloid beta fibril formation in vitro and was able to bind preferentially to misfolded proteins. Given the modest chaperone-like activity of SerpinB2 we tested the ability of SerpinB2 to modulate UPS and autophagy activity using a GFP reporter system and autophagy reporter, respectively. Activity of the UPS was reduced and autophagy was dysregulated in SerpinB2-/- compared to wild-type MEFs. Moreover, we observed a non-covalent interaction between ubiquitin and SerpinB2 in cells using GFP-pulldown assays and bimolecular fluorescence complementation. We conclude that SerpinB2 plays an important role in proteostasis as its loss leads to a proteotoxic phenotype associated with an inability to compartmentalize aggregating proteins and a reduced capacity of the UPS.


Assuntos
Citoproteção/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Corpos de Inclusão/efeitos dos fármacos , Corpos de Inclusão/metabolismo , Inibidor 2 de Ativador de Plasminogênio/metabolismo , Inibidor 2 de Ativador de Plasminogênio/farmacologia , Dobramento de Proteína/efeitos dos fármacos , Peptídeos beta-Amiloides/química , Animais , Éxons/genética , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Camundongos , Peptídeos/metabolismo , Gravidez , Agregados Proteicos , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Ubiquitina/metabolismo
15.
J Gen Virol ; 96(Pt 3): 507-512, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25480927

RESUMO

Chikungunya virus (CHIKV) is a mosquito-borne pathogen responsible for epidemics of debilitating arthritic disease. The recent outbreak (2004-2014) resulted in an estimated 1.4-6.5 million cases, with imported cases reported in nearly 40 countries. The development of CHIKV-specific diagnostics and research tools is thus highly desirable. Herein we describe the generation and characterization of the first mAbs specific for the capsid protein (CP) of CHIKV. The antibodies recognized isolates representing the major genotypes of CHIKV, as well as several other alphaviruses, and were reactive in a range of assays including ELISA, Western blot, immunofluorescence and immunohistochemistry (IHC). We have also used the anti-CP mAb 5.5G9 in IHC studies to show that capsid antigen is persistently expressed 30 days post-infection in cells with macrophage morphology in a mouse model of chronic CHIKV disease. These antibodies may thus represent useful tools for further research, including investigations into the structure and function of CHIKV CP, and as valuable reagents for CHIKV detection in a range of settings.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas do Capsídeo/imunologia , Vírus Chikungunya/imunologia , Animais , Especificidade de Anticorpos , Western Blotting , Células COS , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Febre de Chikungunya/imunologia , Febre de Chikungunya/virologia , Vírus Chikungunya/genética , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Regulação Viral da Expressão Gênica , Humanos , Imuno-Histoquímica , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
16.
Cancer Med ; 3(3): 500-13, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24644264

RESUMO

Expression of SerpinB2 (plasminogen activator inhibitor type 2/PAI-2) by certain cancers is associated with a favorable prognosis. Although tumor-associated host tissues can express SerpinB2, no significant differences in the growth of a panel of different tumors in SerpinB2(-/-) and SerpinB2(+/+) mice were observed. SerpinB2 expression by cancer cells (via lentiviral transduction) also had no significant effect on the growth of panel of mouse and human tumor lines in vivo or in vitro. SerpinB2 expression by cancer cells did, however, significantly reduce the number of metastases in a B16 metastasis model. SerpinB2-expressing B16 cells also showed reduced migration and increased length of invadopodia-like structures, supporting the classical view that that tumor-derived SerpinB2 is inhibiting extracellular urokinase. Importantly, although SerpinB2 is usually poorly secreted, we found that SerpinB2 effectively reaches the extracellular milieu on the surface of 0.5-1 µm microparticles (MPs), where it was able to inhibit urokinase. We also provide evidence that annexins mediate the binding of SerpinB2 to phosphatidylserine, a lipid characteristically exposed on the surface of MPs. The presence of SerpinB2 on the surface of MPs provides a physiological mechanism whereby cancer cell SerpinB2 can reach the extracellular milieu and access urokinase plasminogen activator (uPA). This may then lead to inhibition of metastasis and a favorable prognosis.


Assuntos
Melanoma Experimental/genética , Metástase Neoplásica/genética , Inibidor 2 de Ativador de Plasminogênio/biossíntese , Animais , Micropartículas Derivadas de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma Experimental/patologia , Camundongos , Metástase Neoplásica/patologia , Fosfatidilserinas/metabolismo , Inibidor 2 de Ativador de Plasminogênio/genética , Prognóstico , Ligação Proteica , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
17.
Clin Immunol ; 149(3): 487-97, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24239837

RESUMO

Chikungunya virus (CHIKV) recently caused the largest epidemic ever recorded for this virus involving an estimated 1.4-6.5million cases, with imported cased reported in over 40 countries. The number of monoclonal antibodies specific for this re-emerging alphavirus is currently limited. Herein we describe the generation and characterisation of five monoclonal antibodies specific for the E2 glycoprotein of CHIKV. The antibodies detected a range of CHIKV isolates in several assays including ELISA, Western blot, immunofluorescence assay (IFA) and immunohistochemistry (IHC) without evidence of cross-reactivity with other alphaviruses. Four antibodies also neutralised CHIKV in vitro, two of which provided complete protection against arthritis in a CHIKV mouse model when administered prior to infection. Given the current shortage of widely available reagents for CHIKV, these specific antibodies will be useful not only in research, but may also provide the basis for new diagnostics and treatments.


Assuntos
Infecções por Alphavirus/prevenção & controle , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/farmacologia , Artrite Experimental/prevenção & controle , Proteínas do Envelope Viral/imunologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/isolamento & purificação , Artrite Experimental/imunologia , Artrite Experimental/virologia , Células COS , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/imunologia , Chlorocebus aethiops , Feminino , Humanos , Hibridomas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células Vero , Proteínas do Envelope Viral/genética
18.
PLoS Negl Trop Dis ; 7(9): e2423, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24069479

RESUMO

The mosquito-borne alphavirus, chikungunya virus (CHIKV), has recently reemerged, producing the largest epidemic ever recorded for this virus, with up to 6.5 million cases of acute and chronic rheumatic disease. There are currently no licensed vaccines for CHIKV and current anti-inflammatory drug treatment is often inadequate. Here we describe the isolation and characterization of two human monoclonal antibodies, C9 and E8, from CHIKV infected and recovered individuals. C9 was determined to be a potent virus neutralizing antibody and a biosensor antibody binding study demonstrated it recognized residues on intact CHIKV VLPs. Shotgun mutagenesis alanine scanning of 98 percent of the residues in the E1 and E2 glycoproteins of CHIKV envelope showed that the epitope bound by C9 included amino-acid 162 in the acid-sensitive region (ASR) of the CHIKV E2 glycoprotein. The ASR is critical for the rearrangement of CHIKV E2 during fusion and viral entry into host cells, and we predict that C9 prevents these events from occurring. When used prophylactically in a CHIKV mouse model, C9 completely protected against CHIKV viremia and arthritis. We also observed that when administered therapeutically at 8 or 18 hours post-CHIKV challenge, C9 gave 100% protection in a pathogenic mouse model. Given that targeting this novel neutralizing epitope in E2 can potently protect both in vitro and in vivo, it is likely to be an important region both for future antibody and vaccine-based interventions against CHIKV.


Assuntos
Infecções por Alphavirus/prevenção & controle , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Vírus Chikungunya/imunologia , Proteínas do Envelope Viral/imunologia , Infecções por Alphavirus/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/isolamento & purificação , Febre de Chikungunya , Modelos Animais de Doenças , Mapeamento de Epitopos , Humanos , Imunização Passiva , Camundongos , Camundongos Endogâmicos C57BL , Resultado do Tratamento
19.
PLoS One ; 8(2): e57343, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23460840

RESUMO

SerpinB2, also known as plasminogen activator inhibitor type 2, is a major product of activated monocytes/macrophages and is often strongly induced during infection and inflammation; however, its physiological function remains somewhat elusive. Herein we show that SerpinB2 is induced in peripheral blood mononuclear cells following infection of pigtail macaques with CCR5-utilizing (macrophage-tropic) SIVmac239, but not the rapidly pathogenic CXCR4-utilizing (T cell-tropic) SHIVmn229. To investigate the role of SerpinB2 in lentiviral infections, SerpinB2(-/-) mice were infected with EcoHIV, a chimeric HIV in which HIV gp120 has been replaced with gp80 from ecotropic murine leukemia virus. EcoHIV infected SerpinB2(-/-) mice produced significantly lower anti-gag IgG1 antibody titres than infected SerpinB2(+/+) mice, and showed slightly delayed clearance of EcoHIV. Analyses of published microarray studies showed significantly higher levels of SerpinB2 mRNA in monocytes from HIV-1 infected patients when compared with uninfected controls, as well as a significant negative correlation between SerpinB2 and T-bet mRNA levels in peripheral blood mononuclear cells. These data illustrate that SerpinB2 can be induced by lentiviral infection in vivo and support the emerging notion that a physiological role of SerpinB2 is modulation of Th1/Th2 responses.


Assuntos
Infecções por Lentivirus/imunologia , Inibidor 2 de Ativador de Plasminogênio/metabolismo , Células Th1/imunologia , Células Th2/imunologia , Animais , Movimento Celular , Regulação da Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/fisiologia , Humanos , Imunoglobulina G/imunologia , Infecções por Lentivirus/genética , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Macaca nemestrina/imunologia , Macaca nemestrina/virologia , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Inibidor 2 de Ativador de Plasminogênio/deficiência , Inibidor 2 de Ativador de Plasminogênio/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral/fisiologia
20.
Arch Dermatol Res ; 305(1): 79-83, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22871992

RESUMO

Ingenol mebutate has recently been approved by the Federal Drug Administration (USA) as a topical treatment for actinic keratoses. Herein, we describe the efficacy of ingenol mebutate for the topical treatment of squamous cell carcinoma (SCC) using a wild-type mouse model (SKH1) and the UV-induced mouse SCC cell line, T7. Daily treatment for 2 days with 0.25 % ingenol mebutate gel produced a cure rate of 70 %, with 0 % for placebo gel. Electron microscopy revealed swelling of cancer cell mitochondria within 1 h, with disruption of the inner mitochondrial membranes evident at 6 h post treatment. Primary necrosis of cancer cells was clearly evident by 24 h. Treatment was associated with local haemorrhage and a prodigious neutrophil infiltrate, with anti-T7 antibodies also detected. This is the first report of the successful treatment of SCC tumours with ingenol mebutate gel in wild-type mice, and supports the view that ingenol mebutate induces primary necrosis and activates the immune system.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Diterpenos/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Administração Cutânea , Animais , Antineoplásicos/administração & dosagem , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/ultraestrutura , Diterpenos/administração & dosagem , Feminino , Géis , Masculino , Camundongos , Camundongos Pelados , Microscopia Eletrônica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Dilatação Mitocondrial/efeitos dos fármacos , Necrose , Infiltração de Neutrófilos/efeitos dos fármacos , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/ultraestrutura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA