Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
MAbs ; 16(1): 2297451, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38170638

RESUMO

The development of specific, safe, and potent monoclonal antibodies (Abs) has led to novel therapeutic options for infectious disease. In addition to preventing viral infection through neutralization, Abs can clear infected cells and induce immunomodulatory functions through engagement of their crystallizable fragment (Fc) with complement proteins and Fc receptors on immune cells. Little is known about the role of Fc effector functions of neutralizing Abs in the context of encephalitic alphavirus infection. To determine the role of Fc effector function in therapeutic efficacy against Venezuelan equine encephalitis virus (VEEV), we compared the potently neutralizing anti-VEEV human IgG F5 (hF5) Ab with intact Fc function (hF5-WT) or containing the loss of function Fc mutations L234A and L235A (hF5-LALA) in the context of VEEV infection. We observed significantly reduced binding to complement and Fc receptors, as well as differential in vitro kinetics of Fc-mediated cytotoxicity for hF5-LALA compared to hF5-WT. The in vivo efficacy of hF5-LALA was comparable to hF5-WT at -24 and + 24 h post infection, with both Abs providing high levels of protection. However, when hF5-WT and hF5-LALA were administered + 48 h post infection, there was a significant decrease in the therapeutic efficacy of hF5-LALA. Together these results demonstrate that optimal therapeutic Ab treatment of VEEV, and possibly other encephalitic alphaviruses, requires neutralization paired with engagement of immune effectors via the Fc region.


Assuntos
Anticorpos Antivirais , Vírus da Encefalite Equina Venezuelana , Animais , Cavalos , Humanos , Vírus da Encefalite Equina Venezuelana/genética , Anticorpos Neutralizantes/farmacologia , Receptores Fc , Imunoglobulina G
2.
Front Immunol ; 13: 801182, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154115

RESUMO

Leishmania donovani is a protozoan parasite that causes visceral leishmaniasis, provoking liver and spleen tissue destruction that is lethal unless treated. The parasite replicates in macrophages and modulates host microbicidal responses. We have previously reported that neutrophil elastase (NE) is required to sustain L. donovani intracellular growth in macrophages through the induction of interferon beta (IFN-ß). Here, we show that the gene expression of IFN-ß by infected macrophages was reduced by half when TLR4 was blocked by pre-treatment with neutralizing antibodies or in macrophages from tlr2-/- mice, while the levels in macrophages from myd88-/- mice were comparable to those from wild-type C57BL/6 mice. The neutralization of TLR4 in tlr2-/- macrophages completely abolished induction of IFN-ß gene expression upon parasite infection, indicating an additive role for both TLRs. Induction of type I interferon (IFN-I), OASL2, SOD1, and IL10 gene expression by L. donovani was completely abolished in macrophages from NE knock-out mice (ela2-/-) or from protein kinase R (PKR) knock-out mice (pkr-/-), and in C57BL/6 macrophages infected with transgenic L. donovani expressing the inhibitor of serine peptidase 2 (ISP2). Parasite intracellular growth was impaired in pkr-/- macrophages but was fully restored by the addition of exogenous IFN-ß, and parasite burdens were reduced in the spleen of pkr-/- mice at 7 days, as compared to the 129Sv/Ev background mice. Furthermore, parasites were unable to grow in macrophages lacking TLR3, which correlated with lack of IFN-I gene expression. Thus, L. donovani engages innate responses in infected macrophages via TLR2, TLR4, and TLR3, via downstream PKR, to induce the expression of pro-survival genes in the host cell, and guarantee parasite intracellular development.


Assuntos
Interferon-alfa/metabolismo , Interferon beta/metabolismo , Leishmania donovani/imunologia , Leishmaniose Visceral/imunologia , Macrófagos Peritoneais/imunologia , Transdução de Sinais/genética , Receptor 2 Toll-Like/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , eIF-2 Quinase/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Feminino , Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Interferon-alfa/genética , Interferon beta/genética , Leishmaniose Visceral/parasitologia , Elastase de Leucócito/antagonistas & inibidores , Elastase de Leucócito/genética , Elastase de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Sulfonamidas/farmacologia , Receptor 2 Toll-Like/genética , Receptor 3 Toll-Like/genética , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/imunologia , eIF-2 Quinase/genética
3.
Sci Rep ; 12(1): 700, 2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35027600

RESUMO

Venezuelan equine encephalitis virus (VEEV) is a mosquito borne alphavirus which leads to high viremia in equines followed by lethal encephalitis and lateral spread to humans. In addition to naturally occurring outbreaks, VEEV is a potential biothreat agent with no approved human vaccine or therapeutic currently available. Single domain antibodies (sdAb), also known as nanobodies, have the potential to be effective therapeutic agents. Using an immune phage display library derived from a llama immunized with an equine vaccine that included inactivated VEEV, five sdAb sequence families were identified that showed varying ability to neutralize VEEV. One of the sequence families had been identified previously in selections against chikungunya virus, a related alphavirus of public health concern. A key advantage of sdAb is the ability to optimize properties such as neutralization capacity through protein engineering. Neutralization of VEEV was improved by two orders of magnitude by genetically linking sdAb. One of the bivalent constructs showed effective neutralization of both VEEV and chikungunya virus. Several of the bivalent constructs neutralized VEEV in cell-based assays with reductions in the number of plaques by 50% at protein concentrations of 1 ng/mL or lower, making future evaluation of their therapeutic potential compelling.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Encefalomielite Equina Venezuelana/virologia , Doenças dos Cavalos/prevenção & controle , Doenças dos Cavalos/virologia , Anticorpos de Domínio Único/uso terapêutico , Animais , Anticorpos Neutralizantes/farmacologia , Cavalos , Humanos , Engenharia de Proteínas , Anticorpos de Domínio Único/farmacologia
4.
Viruses ; 13(7)2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34372553

RESUMO

The recent spreading of new SARS-CoV-2 variants, carrying several mutations in the spike protein, could impact immune protection elicited by natural infection or conferred by vaccination. In this study, we evaluated the neutralizing activity against the viral variants that emerged in the United Kingdom (B.1.1.7), Brazil (P.1), and South Africa (B.1.351) in human serum samples from hospitalized patients infected by SARS-CoV-2 during the first pandemic wave in Italy in 2020. Of the patients studied, 59.5% showed a decrease (≥2 fold) in neutralizing antibody titer against B.1.1.7, 83.3% against P.1, and 90.5% against B.1.351 with respect to the original strain. The reduction in antibody titers against all analyzed variants, and in particular P.1 and B.1.351, suggests that previous symptomatic infection might be not fully protective against exposure to SARS-CoV-2 variants carrying a set of relevant spike mutations.


Assuntos
Anticorpos Neutralizantes/farmacologia , Tratamento Farmacológico da COVID-19 , COVID-19/imunologia , SARS-CoV-2/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Brasil/epidemiologia , COVID-19/epidemiologia , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Chlorocebus aethiops , Mutação , Pandemias , SARS-CoV-2/genética , SARS-CoV-2/imunologia , África do Sul/epidemiologia , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Reino Unido/epidemiologia , Vacinação , Células Vero
5.
Biochem J ; 478(3): 597-617, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33600595

RESUMO

A sequence of interconnected events known as the metastatic cascade promotes tumor progression by regulating cellular and molecular interactions between tumor, stromal, endothelial, and immune cells both locally and systemically. Recently, a new concept has emerged to better describe this process by defining four attributes that metastatic cells should undergo. Every individual hallmark represents a unique trait of a metastatic cell that impacts directly in the outcome of the metastasis process. These critical features, known as the hallmarks of metastasis, include motility and invasion, modulation of the microenvironment, cell plasticity and colonization. They are hierarchically regulated at different levels by several factors, including galectins, a highly conserved family of ß-galactoside-binding proteins abundantly expressed in tumor microenvironments and sites of metastasis. In this review, we discuss the role of galectins in modulating each hallmark of metastasis, highlighting novel therapeutic opportunities for treating the metastatic disease.


Assuntos
Galectinas/fisiologia , Metástase Neoplásica/prevenção & controle , Proteínas de Neoplasias/fisiologia , Imunidade Adaptativa , Animais , Anticorpos Neutralizantes/farmacologia , Aptâmeros de Nucleotídeos/farmacologia , Carboidratos/farmacologia , Movimento Celular , Ensaios Clínicos Fase I como Assunto , Transição Epitelial-Mesenquimal/fisiologia , Matriz Extracelular/metabolismo , Galectinas/antagonistas & inibidores , Humanos , Imunidade Inata , Camundongos , Invasividade Neoplásica , Metástase Neoplásica/imunologia , Metástase Neoplásica/fisiopatologia , Proteínas de Neoplasias/antagonistas & inibidores , Células Neoplásicas Circulantes , Neovascularização Patológica/metabolismo , Oligopeptídeos/farmacologia , Peptídeos/farmacologia , Polissacarídeos/fisiologia , RNA Interferente Pequeno/farmacologia , Células Estromais/metabolismo , Microambiente Tumoral/fisiologia
6.
Anaerobe ; 69: 102326, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33508438

RESUMO

Beta toxins (CPB) produced by Clostridium perfringens type B and C cause various diseases in animals, and the use of toxoids is an important prophylactic measure against such diseases. Promising recombinant toxoids have been developed recently. However, both soluble and insoluble proteins expressed in Escherichia coli can interfere with the production and immunogenicity of these antigens. In this context, bioinformatics tools have been used to design new versions of the beta toxin, and levels of expression and solubility were evaluated in different strains of E. coli. The immunogenicity in sheep was assessed using the molecule with the greatest potential that was selected on analyzing these results. In silico analyzes, greater mRNA stability (-169.70 kcal/mol), solubility (-0.755), and better tertiary structure (-0.12) were shown by rCPB-C. None of the strains of E. coli expressed rFH8-CPB, but a high level of expression and solubility was shown by rCPB-C. Higher levels of total and neutralizing anti-CPB antibodies were observed in sheep inoculated with bacterins containing rCPB-C. Thus, this study suggests that due to higher productivity of rCPB-C in E. coli and immunogenicity, it is considered as the most promising molecule for the production of a recombinant vaccine against diseases caused by the beta toxin produced by C. perfringens type B and C.


Assuntos
Anticorpos Neutralizantes/farmacologia , Infecções por Clostridium/prevenção & controle , Infecções por Clostridium/veterinária , Clostridium perfringens/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Toxoides/farmacologia , Vacinas Sintéticas/farmacologia , Animais , Imunogenicidade da Vacina , Ovinos
7.
Angiogenesis ; 24(1): 1-5, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33006019

RESUMO

Galectins, a family of highly conserved ß-galactoside-binding proteins, control tumor progression by modulating different hallmarks of cancer. Galectin-1 (Gal-1), a proto-type member of this family, plays essential roles in tumor angiogenesis and immunosuppression by cross-linking glycosylated receptors on the surface of endothelial and immune cells. Targeted disruption of Gal-1 suppresses tumor growth by counteracting aberrant angiogenesis and reinforcing antitumor immunity in several experimental settings. Given the multiple therapeutic benefits associated with Gal-1 blockade, several Gal-1 inhibitors, including glycan-based competitors, antagonistic peptides, aptamers and neutralizing monoclonal antibodies, have been designed and evaluated in pre-clinical tumor models. Here we report the biochemical and functional characterization of a newly developed neutralizing anti-human Gal-1 monoclonal antibody (Gal-1-mAb3), which specifically recognizes a unique epitope in Gal-1 protein and exerts both angioregulatory and immunomodulatory activities. Blockade of Gal-1 function using Gal-1-mAb3, might be relevant not only in cancer but also in other pathologic conditions characterized by aberrant angiogenesis and uncontrolled immunosuppression.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Galectina 1/imunologia , Fatores Imunológicos/farmacologia , Neovascularização Fisiológica , Animais , Fenômenos Biofísicos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos Endogâmicos BALB C , Neovascularização Fisiológica/efeitos dos fármacos
8.
PLoS Pathog ; 15(12): e1008157, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31790515

RESUMO

There are no FDA licensed vaccines or therapeutics for Venezuelan equine encephalitis virus (VEEV) which causes a debilitating acute febrile illness in humans that can progress to encephalitis. Previous studies demonstrated that murine and macaque monoclonal antibodies (mAbs) provide prophylactic and therapeutic efficacy against VEEV peripheral and aerosol challenge in mice. Additionally, humanized versions of two neutralizing mAbs specific for the E2 glycoprotein, 1A3B-7 and 1A4A-1, administered singly protected mice against aerosolized VEEV. However, no studies have demonstrated protection in nonhuman primate (NHP) models of VEEV infection. Here, we evaluated a chimeric antibody 1A3B-7 (c1A3B-7) containing mouse variable regions on a human IgG framework and a humanized antibody 1A4A-1 containing a serum half-life extension modification (Hu-1A4A-1-YTE) for their post-exposure efficacy in NHPs exposed to aerosolized VEEV. Approximately 24 hours after exposure, NHPs were administered a single bolus intravenous mAb. Control NHPs had typical biomarkers of VEEV infection including measurable viremia, fever, and lymphopenia. In contrast, c1A3B-7 treated NHPs had significant reductions in viremia and lymphopenia and on average approximately 50% reduction in fever. Although not statistically significant, Hu-1A4A-1-YTE administration did result in reductions in viremia and fever duration. Delay of treatment with c1A3B-7 to 48 hours post-exposure still provided NHPs protection from severe VEE disease through reductions in viremia and fever. These results demonstrate that post-exposure administration of c1A3B-7 protected macaques from development of severe VEE disease even when administered 48 hours following aerosol exposure and describe the first evaluations of VEEV-specific mAbs for post-exposure prophylactic use in NHPs. Viral mutations were identified in one NHP after c1A3B-7 treatment administered 24 hrs after virus exposure. This suggests that a cocktail-based therapy, or an alternative mAb against an epitope that cannot mutate without resulting in loss of viral fitness may be necessary for a highly effective therapeutic.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Neutralizantes/farmacologia , Encefalomielite Equina Venezuelana/imunologia , Vacinas Virais/farmacologia , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Neutralizantes/imunologia , Modelos Animais de Doenças , Encefalomielite Equina Venezuelana/prevenção & controle , Humanos , Macaca fascicularis , Vacinas Virais/imunologia
9.
Drug Des Devel Ther ; 13: 3813-3821, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31806932

RESUMO

PURPOSE: To evaluate the efficacy of a therapy on improving characteristics of laser-induced choroidal neovascularization (CNV) via single intravitreal injection of a humanized anti-human VEGF monoclonal antibody (PRO-169) versus bevacizumab in a rhesus monkey model. METHODS: To induce experimental CNV, small high-energy laser spots were used to treat several areas, around the macula in the retinas of monkeys at Day -21. Eighteen rhesus monkeys were used for CNV induction. The efficacy endpoints were fluorescein leakage by FFA and retinal thickness by OCT. FFA examinations were performed 19 days after induction. Appropriate animals were enrolled for treatment and randomly divided into 3 groups: bevacizumab (n=5, 7 eyes), PRO-169 (n=5, 7 eyes), and vehicle controls (n=4, 7 eyes). RESULTS: In 25 of 36 (69.4%) eyes, CNV lesions were identified. The average percent change of retinal thickness in the eyes of bevacizumab group was -159.3±62.2% and -154.0±45.1% (p<0.01 vs Vehicle) at Day 14 and Day 28, respectively; in the eyes of PRO-169 group it was -131.6±68.7% and -131.5±63.8% (p<0.01 vs Vehicle), respectively. The average percent change of leakage area in the eyes of bevacizumab group was -75.3±49.4% and -78.0±42.6% (p<0.01 vs Vehicle) at Day 14 and Day 28, respectively; in the eyes of PRO-169 group it was -82.0±19.3% and -81.4±21.0% (p<0.01 vs Vehicle), respectively. There were no abnormalities found in behavior, skin and hair, excretion and overall eye appearance before and after treatment in all groups. CONCLUSION: After photocoagulation, the eyes enrolled in this studio showed CNV related characteristics including increased retinal thickness, and fluorescein leakage at laser spots. PRO-169 (1.25 mg per eye) can reduce the retinal thickness and fluorescein leakage area after treatment for 14 and 28 days in this rhesus monkeys model, without toxic effect or adverse events. These findings suggested that PRO-169 can inhibit CNV.


Assuntos
Inibidores da Angiogênese/farmacologia , Anticorpos Neutralizantes/farmacologia , Bevacizumab/farmacologia , Neovascularização de Coroide/tratamento farmacológico , Modelos Animais de Doenças , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Neovascularização de Coroide/metabolismo , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos , Angiofluoresceinografia , Lasers , Macaca mulatta , Estrutura Molecular , Relação Estrutura-Atividade , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Mol Neurobiol ; 56(11): 7774-7788, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31119556

RESUMO

Galectin-8 (Gal-8) is a glycan-binding protein that modulates a variety of cellular processes interacting with cell surface glycoproteins. Neutralizing anti-Gal-8 antibodies that block Gal-8 functions have been described in autoimmune and inflammatory disorders, likely playing pathogenic roles. In the brain, Gal-8 is highly expressed in the choroid plexus and accordingly has been detected in human cerebrospinal fluid. It protects against central nervous system autoimmune damage through its immune-suppressive potential. Whether Gal-8 plays a direct role upon neurons remains unknown. Here, we show that Gal-8 protects hippocampal neurons in primary culture against damaging conditions such as nutrient deprivation, glutamate-induced excitotoxicity, hydrogen peroxide (H2O2)-induced oxidative stress, and ß-amyloid oligomers (Aßo). This protective action is manifested even after 2 h of exposure to the harmful condition. Pull-down assays demonstrate binding of Gal-8 to selected ß1-integrins, including α3 and α5ß1. Furthermore, Gal-8 activates ß1-integrins, ERK1/2, and PI3K/AKT signaling pathways that mediate neuroprotection. Hippocampal neurons in primary culture produce and secrete Gal-8, and their survival decreases upon incubation with human function-blocking Gal-8 autoantibodies obtained from lupus patients. Despite the low levels of Gal-8 expression detected by real-time PCR in hippocampus, compared with other brain regions, the complete lack of Gal-8 in Gal-8 KO mice determines higher levels of apoptosis upon H2O2 stereotaxic injection in this region. Therefore, endogenous Gal-8 likely contributes to generate a neuroprotective environment in the brain, which might be eventually counteracted by human function-blocking autoantibodies.


Assuntos
Anticorpos Neutralizantes/farmacologia , Autoanticorpos/farmacologia , Encéfalo/metabolismo , Galectinas/metabolismo , Neuroproteção , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipocampo/patologia , Humanos , Peróxido de Hidrogênio/metabolismo , Integrina beta1/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neuroproteção/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
11.
Nanotechnology ; 30(35): 355101, 2019 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-31082814

RESUMO

Metallic nanorods are promising agents for a wide range of biomedical applications. We report an optical hyperthermia method capable of inducing slowdown tumor progression of an experimental in vivo CT-2A glioblastoma tumor. The tumor model used in this research is based on the transplantation of mouse astrocytoma CT-2A cells in the striatum of mice by intracranial stereotaxic surgery. Two weeks after cell implant, the resulting tumor is treated by irradiating intratumoral injected gold nanorods, biofunctionalized with CD133 antibody (B-GNRs), using a continuous wave laser. Nanoparticles convert the absorbed light into localized heat (reaching up to 44 °C) due to the effect of surface plasmon resonance. A significant slowdown in CT-2A tumor progression is evident, by histology and magnetic resonance imaging, at one (p = 0.03) and two weeks (p = 0.008) after irradiation treatment. A notable deceleration in tumor size (15%-75%) as compared to the control untreated groups, it is observed. Thus, laser irradiation of B-GNRs is found to be effective for the treatment of CT-2A tumor progression. Similarities between the pre-clinical CT-2A tumor model and the human astrocytoma disease, in terms of anatomy, metastatic behavior and histopathology, suggest that hyperthermic treatment by laser irradiation of B-GNRs administered into high-grade human astrocytoma might constitute a promising alternative treatment to limit the progression of this deadly disease.


Assuntos
Astrocitoma/terapia , Neoplasias Encefálicas/terapia , Ouro/farmacologia , Hipertermia Induzida/métodos , Terapia a Laser/métodos , Nanotubos/química , Antígeno AC133/antagonistas & inibidores , Antígeno AC133/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Astrocitoma/imunologia , Astrocitoma/patologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Ouro/administração & dosagem , Ouro/química , Humanos , Injeções Intralesionais , Lasers , Camundongos , Camundongos Endogâmicos C57BL , Nanotubos/ultraestrutura , Transplante de Neoplasias , Técnicas Estereotáxicas , Ressonância de Plasmônio de Superfície , Carga Tumoral/efeitos da radiação
12.
Toxins (Basel) ; 11(2)2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30759862

RESUMO

Human accidents with spiders of the genus Loxosceles are an important health problem affecting thousands of people worldwide. Patients evolve to severe local injuries and, in many cases, to systemic disturbances as acute renal failure, in which cases antivenoms are considered to be the most effective treatment. However, for antivenom production, the extraction of the venom used in the immunization process is laborious and the yield is very low. Thus, many groups have been exploring the use of recombinant Loxosceles toxins, particularly phospholipases D (PLDs), to produce the antivenom. Nonetheless, some important venom activities are not neutralized by anti-PLD antibodies. Astacin-like metalloproteases (ALMPs) are the second most expressed toxin acting on the extracellular matrix, indicating the importance of its inclusion in the antigen's formulation to provide a better antivenom. Here we show the construction of a hybrid recombinant immunogen, called LgRec1ALP1, composed of hydrophilic regions of the PLD and the ALMP toxins from Loxosceles gaucho. Although the LgRec1ALP1 was expressed as inclusion bodies, it resulted in good yields and it was effective to produce neutralizing antibodies in mice. The antiserum neutralized fibrinogenolytic, platelet aggregation and dermonecrotic activities elicited by L. gaucho, L. laeta, and L. intermedia venoms, indicating that the hybrid recombinant antigen may be a valuable source for the production of protective antibodies against Loxosceles ssp. venoms. In addition, the hybrid recombinant toxin approach may enrich and expand the alternative antigens for antisera production for other venoms.


Assuntos
Anticorpos Neutralizantes/farmacologia , Antivenenos/farmacologia , Diester Fosfórico Hidrolases/toxicidade , Venenos de Aranha/toxicidade , Animais , Antivenenos/metabolismo , Edema/induzido quimicamente , Edema/tratamento farmacológico , Humanos , Masculino , Metaloproteases/metabolismo , Camundongos Endogâmicos BALB C , Necrose/induzido quimicamente , Necrose/tratamento farmacológico , Diester Fosfórico Hidrolases/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Coelhos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Venenos de Aranha/metabolismo , Aranhas
13.
Toxins (Basel) ; 10(12)2018 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-30513821

RESUMO

Shiga toxin (Stx) producing Escherichia coli (STEC) is responsible for causing hemolytic uremic syndrome (HUS), a life-threatening thrombotic microangiopathy characterized by thrombocytopenia, hemolytic anemia, and acute renal failure after bacterially induced hemorrhagic diarrhea. Until now, there has been neither an effective treatment nor method of prevention for the deleterious effects caused by Stx intoxication. Antibodies are well recognized as affinity components of therapeutic drugs; thus, a previously obtained recombinant human FabC11:Stx2 fragment was used to neutralize Stx2 in vitro in a Vero cell viability assay. Herein, we demonstrated that this fragment neutralized, in a dose-dependent manner, the cytotoxic effects of Stx2 on human glomerular endothelial cells, on human proximal tubular epithelial cells, and prevented the morphological alterations induced by Stx2. FabC11:Stx2 protected mice from a lethal dose of Stx2 by toxin-antibody pre-incubation. Altogether, our results show the ability of a new encouraging molecule to prevent Stx-intoxication symptoms during STEC infection.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Toxina Shiga II/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Humanos , Glomérulos Renais/citologia , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/farmacologia
14.
Sci Transl Med ; 10(468)2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30463919

RESUMO

Andes hantavirus (ANDV) is an etiologic agent of hantavirus cardiopulmonary syndrome (HCPS), a severe disease characterized by fever, headache, and gastrointestinal symptoms that may progress to hypotension, pulmonary failure, and cardiac shock that results in a 25 to 40% case-fatality rate. Currently, there is no specific treatment or vaccine; however, several studies have shown that the generation of neutralizing antibody (Ab) responses strongly correlates with survival from HCPS in humans. In this study, we screened 27 ANDV convalescent HCPS patient sera for their capacity to bind and neutralize ANDV in vitro. One patient who showed high neutralizing titer was selected to isolate ANDV-glycoprotein (GP) Abs. ANDV-GP-specific memory B cells were single cell sorted, and recombinant immunoglobulin G antibodies were cloned and produced. Two monoclonal Abs (mAbs), JL16 and MIB22, potently recognized ANDV-GPs and neutralized ANDV. We examined the post-exposure efficacy of these two mAbs as a monotherapy or in combination therapy in a Syrian hamster model of ANDV-induced HCPS, and both mAbs protected 100% of animals from a lethal challenge dose. These data suggest that monotherapy with mAb JL16 or MIB22, or a cocktail of both, could be an effective post-exposure treatment for patients infected with ANDV-induced HCPS.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Infecções por Hantavirus/tratamento farmacológico , Infecções por Hantavirus/prevenção & controle , Orthohantavírus/fisiologia , Proteínas Recombinantes/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Neutralizantes/uso terapêutico , Linfócitos B/efeitos dos fármacos , Glicoproteínas/imunologia , Células HEK293 , Orthohantavírus/efeitos dos fármacos , Infecções por Hantavirus/sangue , Infecções por Hantavirus/imunologia , Humanos , Memória Imunológica/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Sobreviventes
15.
Immunol Res ; 66(3): 392-405, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29855993

RESUMO

Tumor necrosis factor alpha (TNFα) is a pro-inflammatory cytokine that mediates the homeostasis of immune responses; its exacerbated production is associated with the pathogenesis of autoimmune and chronic inflammatory diseases. Anti-TNFα drugs have revolutionized the treatment of inflammatory conditions such as rheumatoid arthritis and Crohn's disease. Currently, a worldwide race is on stage for the production of biosimilars moved by patent expiration of monoclonal antibodies (mAbs), such as anti-TNFα adalimumab. Our goal was to develop the first stage of an adalimumab biosimilar candidate with potential for national production, through the generation of a productive and stable cell line and assess its functionality. The robotic system ClonePix was used for screening and isolation of colonies from transfected CHO-S stable pools plated in semisolid medium. Selected clones were expanded based on growth and productivity. Purified mAbs from different clones were tested for binding and functional activity. The binding affinity of the denominated adabut clones to TNFα and FcRγ did not differ statistically when compared to reference adalimumab. One functional activity assay demonstrated the antibody neutralization capacity of the cytotoxicity induced by TNFα in L929 murine fibroblasts. A second assay confirmed adabut as an antagonist of the TNFα activity by the inhibition of the cell adhesion molecule expression in HUVEC cultures. The binding and functional activity analyses performed with selected adabut clones in comparison to reference adalimumab represent an important status of "non-inferiority," part of the process required for a biosimilar development. We generated and selected high-quality adabut clones which mAbs may be further developed as the first in-house made Brazilian biosimilar, demonstrating a success case for our incipient biotechnology industry, or also modified as biobetters, thus representing an innovative strategy for the patients' welfare.


Assuntos
Adalimumab/imunologia , Anticorpos Monoclonais/imunologia , Medicamentos Biossimilares , Proteínas Recombinantes de Fusão/imunologia , Fator de Necrose Tumoral alfa/imunologia , Adalimumab/genética , Adalimumab/metabolismo , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Células CHO , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Camundongos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
Mol Neurobiol ; 55(10): 8014-8037, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29498005

RESUMO

Neurogenesis plays a significant role during adulthood, and the observation that neural stem cells reside in the central nervous system and the olfactory epithelium has attracted attention due to their importance in neuronal regeneration. In addition, soluble factors (SFs) release by neural stem cells may modulate the neurogenic process. Thus, in this study, we identified the SFs released by olfactory human neural stem/progenitor cells (hNS/PCs-OE). These cells express Ki67, nestin, and ßIII-tubulin, indicating their neural lineage. The hNS/PCs-OE also express PSD95 and tau proteins during proliferation, but increased levels are observed after differentiation. Thus, we evaluated the effects of SFs from hNS/PCs-OE on the viability, proliferation, and differentiation potential of adult murine hippocampal neural precursor cells (AHPCs). SFs from hNS/PCs-OE maintain cells in the precursor and proliferative stages and mainly promote the astrocytic differentiation of AHPCs. These effects involved the activation, as measured by phosphorylation, of several proteins (Erk1/2; Akt/PRAS40/GSK3ß and JAK/STAT) involved in key events of the neurogenic process. Moreover, according to the results from the antibody-based microarray approach, among the soluble factors, hNS/PCs-OE produce interleukin-6 (IL-6) and neurotrophin 4 (NT4). However, residual epidermal growth factor (EGF) was also detected. These proteins partially reproduced the effects of SFs from hNS/PCs-OE on AHPCs, and the mechanism underlying these effects is mediated by Src proteins, which have been implicated in EGF-induced transactivation of TrkB receptor. The results of the present study suggest the potential use of SFs from hNS/PCs-OE in controlling the differentiation potential of AHPCs. Thus, the potential clinical relevance of hNS/PCs-OE is worth pursuing.


Assuntos
Linhagem da Célula , Hipocampo/citologia , Células-Tronco Neurais/citologia , Mucosa Olfatória/citologia , Adulto , Animais , Anticorpos Neutralizantes/farmacologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Citocinas/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Fosforilação/efeitos dos fármacos , Receptor trkB/metabolismo , Solubilidade , Ativação Transcricional/efeitos dos fármacos
17.
Am J Reprod Immunol ; 79(1)2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29154408

RESUMO

PROBLEM: We hypothesized that trophoblast expression of Ccl25 attracts a specific leukocyte cell population to the implantation site for local regulation. METHOD OF STUDY: Mice blastocysts, ectoplacental cones, and decidua at gestational days 3.5-7.5 were evaluated for Ccl25 and Ccr9 expressions. Peripheral availability and characterization of Ccr9+ leukocytes were determined by flow cytometry. Leukocyte chemotaxis was assessed in the presence of Ccl25 recombinant protein and embryos using antisense oligomers (ODNs) to Ccl25 and Ccr9 neutralizing antibody. RESULTS: Ccl25 was expressed by embryonic cells, whereas Ccr9 expression was strong at the maternal compartment and in PBMC. Immunolocalization confirmed this expression. In vitro, chemotaxis assays showed that the embryonic Ccl25 signals to Ccr9+ PBMCs. Maternal Ccr9+α4ß7+ monocytes switch from an anti-inflammatory phenotype (F4/80+11b+Ly6C-TGF-ß+ cells, pre-implantation) to an inflammatory profile (F4/80+11b+Ly6C+TNF-α+ cells, post-implantation). CONCLUSION: Our data support the establishment of a CCL25/CCR9-axis at the maternal-fetal interface in mice, which may be involved in immune regulatory mechanisms during embryo implantation.


Assuntos
Blastocisto/metabolismo , Quimiocinas CC/metabolismo , Implantação do Embrião , Leucócitos Mononucleares/fisiologia , Monócitos/fisiologia , Receptores CCR/metabolismo , Trofoblastos/patologia , Animais , Anticorpos Neutralizantes/farmacologia , Antígenos de Diferenciação/metabolismo , Diferenciação Celular , Células Cultivadas , Quimiotaxia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos , Oligodesoxirribonucleotídeos Antissenso/genética , Transporte Proteico , Receptores CCR/imunologia , Fator de Crescimento Transformador beta/metabolismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-27590117

RESUMO

In this work, we examined some biochemical and biological activities of Bothrops fonsecai venom, a pitviper endemic to southeastern Brazil, and assessed their neutralization by commercial bothropic antivenom (CAv). Cross-reactivity of venom with CAv was also assessed by immunoblotting and size-exclusion high performance chromatography (SE-HPLC). Bothrops fonsecai venom had PLA2, proteolytic and esterase activities that were neutralized to varying extents by venom:antivenom ratios of 5:1 and 5:2 (PLA2 and esterase activities) or not significantly by either venom:antivenom ratio (proteolytic activity). The minimum hemorrhagic dose (69.2µg) was totally neutralized by both ratios. Clotting time in rat citrated plasma was 33±10.5s (mean±SD; n=5) and was completely neutralized by a 5:2 ratio. Edema formation was dose-dependent (1-30µg/site) and significantly inhibited by both ratios. Venom (10-300µg/mL) caused neuromuscular blockade in extensor digitorum longus preparations; this blockade was inhibited best by a 5:2 ratio. Venom caused myonecrosis and creatine kinase release in vivo (gastrocnemius muscle) and in vitro (extensor digitorum longus) that was effectively neutralized by both venom:antivenom ratios. Immunoblotting showed that venom components of ~25-100kDa interacted with CAv. SE-HPLC profiles for venom incubated with CAv or specific anti-B. fonsecai antivenom raised in rabbits (SAv) indicated that CAv had a higher binding capacity than SAv, whereas SAv had higher affinity than CAv. These findings indicate that B. fonsecai venom contains various activities that are neutralized to different extents by CAv and suggest that CAv could be used to treat envenoming by B. fonsecai.


Assuntos
Anticorpos Neutralizantes/imunologia , Antídotos , Antivenenos/imunologia , Bothrops/imunologia , Venenos de Crotalídeos/imunologia , Proteínas de Répteis/imunologia , Mordeduras de Serpentes/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Antídotos/farmacologia , Antivenenos/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Western Blotting , Bothrops/metabolismo , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Reações Cruzadas , Venenos de Crotalídeos/enzimologia , Venenos de Crotalídeos/toxicidade , Relação Dose-Resposta a Droga , Edema/induzido quimicamente , Edema/prevenção & controle , Eletroforese em Gel Bidimensional , Esterases/imunologia , Esterases/metabolismo , Fosfolipases A2 do Grupo II/imunologia , Fosfolipases A2 do Grupo II/metabolismo , Hemorragia/sangue , Hemorragia/induzido quimicamente , Hemorragia/prevenção & controle , Masculino , Camundongos , Junção Neuromuscular/efeitos dos fármacos , Peptídeo Hidrolases/imunologia , Peptídeo Hidrolases/metabolismo , Proteólise , Ratos Wistar , Proteínas de Répteis/metabolismo , Proteínas de Répteis/toxicidade , Mordeduras de Serpentes/tratamento farmacológico , Mordeduras de Serpentes/enzimologia , Fatores de Tempo
19.
BMC Cancer ; 16(1): 731, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27619675

RESUMO

BACKGROUND: For several decades now an antagonism between Trypanosoma cruzi infection and tumor development has been detected. The molecular basis of this phenomenon remained basically unknown until our proposal that T. cruzi Calreticulin (TcCRT), an endoplasmic reticulum-resident chaperone, translocated-externalized by the parasite, may mediate at least an important part of this effect. Thus, recombinant TcCRT (rTcCRT) has important in vivo antiangiogenic and antitumor activities. However, the relevant question whether the in vivo antitumor effect of T. cruzi infection is indeed mediated by the native chaperone (nTcCRT), remains open. Herein, by using specific modified anti-rTcCRT antibodies (Abs), we have neutralized the antitumor activity of T. cruzi infection and extracts thereof, thus identifying nTcCRT as a valid mediator of this effect. METHODS: Polyclonal anti-rTcCRT F(ab')2 Ab fragments were used to reverse the capacity of rTcCRT to inhibit EAhy926 endothelial cell (EC) proliferation, as detected by BrdU uptake. Using these F(ab')2 fragments, we also challenged the capacity of nTcCRT, during T. cruzi infection, to inhibit the growth of an aggressive mammary adenocarcinoma cell line (TA3-MTXR) in mice. Moreover, we determined the capacity of anti-rTcCRT Abs to reverse the antitumor effect of an epimastigote extract (EE). Finally, the effects of these treatments on tumor histology were evaluated. RESULTS: The rTcCRT capacity to inhibit ECs proliferation was reversed by anti-rTcCRT F(ab')2 Ab fragments, thus defining them as valid probes to interfere in vivo with this important TcCRT function. Consequently, during infection, these Ab fragments also reversed the in vivo experimental mammary tumor growth. Moreover, anti-rTcCRT Abs also neutralized the antitumor effect of an EE, again identifying the chaperone protein as an important mediator of this anti mammary tumor effect. Finally, as determined by conventional histological parameters, in infected animals and in those treated with EE, less invasive tumors were observed while, as expected, treatment with F(ab')2 Ab fragments increased malignancy. CONCLUSION: We have identified translocated/externalized nTcCRT as responsible for at least an important part of the anti mammary tumor effect of the chaperone observed during experimental infections with T. cruzi.


Assuntos
Calreticulina/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Trypanosoma cruzi/metabolismo , Tripanossomíase/parasitologia , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/farmacologia , Calreticulina/antagonistas & inibidores , Calreticulina/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Feminino , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/farmacologia
20.
Cell Death Dis ; 7: e2232, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27195678

RESUMO

Chagas disease is caused by infection with the protozoan Trypanosoma cruzi. CD8 T-lymphocytes help to control infection, but apoptosis of CD8 T cells disrupts immunity and efferocytosis can enhance parasite infection within macrophages. Here, we investigate how apoptosis of activated CD8 T cells affects M1 and M2 macrophage phenotypes. First, we found that CD8 T-lymphocytes and inflammatory monocytes/macrophages infiltrate peritoneum during acute T. cruzi infection. We show that treatment with anti-Fas ligand (FasL) prevents lymphocyte apoptosis, upregulates type-1 responses to parasite antigens, and reduces infection in macrophages cocultured with activated CD8 T cells. Anti-FasL skews mixed M1/M2 macrophage profiles into polarized M1 phenotype, both in vitro and following injection in infected mice. Moreover, inhibition of T-cell apoptosis induces a broad reprogramming of cytokine responses and improves macrophage-mediated immunity to T. cruzi. The results indicate that disposal of apoptotic CD8 T cells increases M2-macrophage differentiation and contributes to parasite persistence.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doença de Chagas/imunologia , Proteína Ligante Fas/antagonistas & inibidores , Interações Hospedeiro-Parasita , Imunidade Celular/efeitos dos fármacos , Macrófagos/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Apoptose/efeitos dos fármacos , Assialoglicoproteínas/genética , Assialoglicoproteínas/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/parasitologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/parasitologia , Comunicação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Doença de Chagas/tratamento farmacológico , Doença de Chagas/genética , Doença de Chagas/parasitologia , Técnicas de Cocultura , Proteína Ligante Fas/genética , Proteína Ligante Fas/imunologia , Regulação da Expressão Gênica , Subunidade p35 da Interleucina-12/genética , Subunidade p35 da Interleucina-12/imunologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Ativação Linfocitária , Macrófagos/efeitos dos fármacos , Macrófagos/parasitologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Trypanosoma cruzi/crescimento & desenvolvimento , Trypanosoma cruzi/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA