Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.357
Filtrar
1.
Cancer Res Commun ; 4(4): 1050-1062, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38592453

RESUMO

The ability to temporally regulate gene expression and track labeled cells makes animal models powerful biomedical tools. However, sudden expression of xenobiotic genes [e.g., GFP, luciferase (Luc), or rtTA3] can trigger inadvertent immunity that suppresses foreign protein expression or results in complete rejection of transplanted cells. Germline exposure to foreign antigens somewhat addresses these challenges; however, native fluorescence and bioluminescence abrogates the utility of reporter proteins and highly spatiotemporally restricted expression can lead to suboptimal xenoantigen tolerance. To overcome these unwanted immune responses and enable reliable cell tracking/gene regulation, we developed a novel mouse model that selectively expresses antigen-intact but nonfunctional forms of GFP and Luc, as well as rtTA3, after CRE-mediated recombination. Using tissue-specific CREs, we observed model and sex-based differences in immune tolerance to the encoded xenoantigens, illustrating the obstacles of tolerizing animals to foreign genes and validating the utility of these "NoGlow" mice to dissect mechanisms of central and peripheral tolerance. Critically, tissue unrestricted NoGlow mice possess no detectable background fluorescence or luminescence and exhibit limited adaptive immunity against encoded transgenic xenoantigens after vaccination. Moreover, we demonstrate that NoGlow mice allow tracking and tetracycline-inducible gene regulation of triple-transgenic cells expressing GFP/Luc/rtTA3, in contrast to transgene-negative immune-competent mice that eliminate these cells or prohibit metastatic seeding. Notably, this model enables de novo metastasis from orthotopically implanted, triple-transgenic tumor cells, despite high xenoantigen expression. Altogether, the NoGlow model provides a critical resource for in vivo studies across disciplines, including oncology, developmental biology, infectious disease, autoimmunity, and transplantation. SIGNIFICANCE: Multitolerant NoGlow mice enable tracking and gene manipulation of transplanted tumor cells without immune-mediated rejection, thus providing a platform to investigate novel mechanisms of adaptive immunity related to metastasis, immunotherapy, and tolerance.


Assuntos
Antígenos Heterófilos , Rastreamento de Células , Animais , Camundongos , Regulação da Expressão Gênica , Camundongos Transgênicos , Modelos Animais de Doenças
2.
Biotechnol J ; 19(1): e2300254, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37750498

RESUMO

The novel mosquito-borne Tembusu virus (TMUV, family Flaviviridae) was discovered as the cause of a severe outbreak of egg-drop syndrome affecting ducks in Southeast Asia in 2010. TMUV infection can also lead to high mortality in various additional avian species such as geese, pigeons, and chickens. This study describes the construction of an infectious cDNA clone of a contemporary duck-isolate (TMUV WU2016). The virus recovered after transfection of BHK-21 cells shows enhanced virus replication compared to the mosquito-derived MM1775 strain. Next, the WU2016 cDNA clone was modified to create a SP6 promoter-driven, self-amplifying mRNA (replicon) capable of expressing a range of different reporter genes (Renilla luciferase, mScarlet, mCherry, and GFP) and viral (glyco)proteins of avian influenza virus (AIV; family Orthomyxoviridae), infectious bursal disease virus (IDBV; family Bunyaviridae) and infectious bronchitis virus (IBV; family Coronaviridae). The current study demonstrates the flexibility of the TMUV replicon system, to produce different heterologous proteins over an extended period of time and its potential use as a platform technology for novel poultry vaccines.


Assuntos
Culicidae , Infecções por Flavivirus , Flavivirus , Doenças das Aves Domésticas , Animais , Infecções por Flavivirus/veterinária , Infecções por Flavivirus/genética , Aves Domésticas/genética , Genes Reporter/genética , DNA Complementar , Antígenos Heterófilos , Doenças das Aves Domésticas/genética , Galinhas , Flavivirus/genética , Patos/genética , Células Clonais , Replicon
3.
Xenotransplantation ; 30(6): e12834, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37971870

RESUMO

Pig liver xenotransplantation is limited by a thrombocytopenic coagulopathy that occurs immediately following graft reperfusion. In vitro and ex vivo studies from our lab suggested that the thrombocytopenia may be the result of a species incompatibility in platelet glycosylation. Realization that platelet α-granules contain antibodies caused us to reevaluate whether the thrombocytopenia in liver xenotransplantation could occur because IgM and IgG from inside platelet α-granules bound to pig liver sinusoidal endothelial cells (LSECs). Our in vitro analysis of IgM and IgG from inside α-granules showed that platelets do carry xenoreactive antibodies that can bind to known xenoantigens. This study suggests that thrombocytopenia occurring following liver xenotransplantation could occur because of xenoreactive antibodies tethering human platelets to the pig LSEC enabling the platelet to be phagocytosed. These results suggest genetic engineering strategies aimed at reducing xenoantigens on the surface of pig LSEC will be effective in eliminating the thrombocytopenia that limits survival in liver xenotransplantation.


Assuntos
Células Endoteliais , Trombocitopenia , Suínos , Animais , Humanos , Transplante Heterólogo/métodos , Fígado , Plaquetas , Trombocitopenia/etiologia , Antígenos Heterófilos , Imunoglobulina G , Imunoglobulina M
4.
Front Immunol ; 14: 1275658, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38022676

RESUMO

Introduction: Microbial infections are associated with the occurrence of autoimmune diseases, but the mechanisms of microbial infection inducing autoimmune diseases are not fully understood. The existence of heterophilic antigens between microorganisms and human tissues may explain part of the pathogenesis of autoimmune diseases. Here, we investigate the distribution of heterophilic antigens and its relationship with autoimmune diseases. Methods: Monoclonal antibodies against a variety of microorganisms were prepared. The titer, subclass and reactivity of antibodies with microorganisms were identified, and heterophilic antibodies that cross-reacted with human tissues were screened by human tissue microarray. The reactivity of these heterophilic antibodies with different individuals and different species was further examined by immunohistochemistry. Results: In this study, 21 strains of heterophilic antibodies were screened. The results showed that these heterophilic antibodies were produced due to the existence of heterophilic antigens between microorganism and human body and the distribution of heterophilic antigens had individual, tissue and species differences. Conclusion: Our study showed that heterophilic antigens exist widely between microorganisms and human body, and the heterophilic antigens carried by microorganisms may break the immune tolerance of the body through carrier effect and initiate immune response, which may be one of the important mechanisms of infection inducing autoimmune diseases.


Assuntos
Antígenos Heterófilos , Doenças Autoimunes , Humanos , Anticorpos Monoclonais , Anticorpos Heterófilos , Imuno-Histoquímica
5.
Biomaterials ; 302: 122297, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37666102

RESUMO

The dysfunction of tumor infiltrating lymphocytes (TILs) directly correlates with out of control of tumor growth and metastasis. New approaches and insightful clarity for rescuing TILs dysfunction are urgently needed. Here, we design two heterogenous antigens based on MHC-I epitope and MHC-II epitope from tumor, and assemble heterogenous antigens by electrostatic interactions and π-π stacking into heteroantigen-assembled nanovaccine (HANV). HANV not only significantly increases the abundance of CD8+ and CD4+ TILs, but also elicits stronger polyfunctionality of CD8+ and CD4+ TILs in vivo. Enhanced polyfunctionality of CD8+ and CD4+ TILs positively correlate to suppression of tumor growth and metastasis in melanoma-bearing mouse models. We also validate that nucleotide-binding oligomerization domain-containing protein 2 (NOD2) dominantly enhances anti-tumor capacity of TILs in a temporal immunoregulation manner. This work presents a new insight in developing HANV as a rational strategy to shape TILs polyfunctionality for tumor growth and metastasis.


Assuntos
Linfócitos do Interstício Tumoral , Melanoma , Animais , Camundongos , Linfócitos do Interstício Tumoral/patologia , Melanoma/patologia , Epitopos , Antígenos Heterófilos , Linfócitos T CD8-Positivos
6.
Cancer Rep (Hoboken) ; 6(8): e1831, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37265054

RESUMO

BACKGROUND AND OBJECTIVES: Hepatocellular carcinoma (HCC) has a high recurrence rate even after radical hepatectomy. More optimal biomarkers may help improve recurrence and prognosis. METHODS: We investigated whether the oncological properties of N-glycolylneuraminic acid (NeuGc) can participate in the prognosis of HCC. We evaluated the NeuGc antigen (Ag) expression in the HCC tissues and measured the preoperative anti-NeuGc IgG antibodies (Abs) in the sera of the patients with HCC. We compared the clinical characteristics and survival rate in the hepatectomized patients (initial; n = 66, recurrent; n = 34) with and without the NeuGc Ag or Abs. RESULTS: Multivariate analyses showed positive expression of NeuGc Ag in HCC tissues (Odds ratio; initial = 6.3, recurrent = 14.0) and higher titers of preoperative anti-NeuGc Ab (Odds ratio; initial = 4.9; recurrent = 3.8), which could be the predictive factors related to early recurrence. Both the NeuGc Ag-positive and Ab-positive groups in the initial hepatectomized patients exhibited significantly shorter recurrent free survival compared to those in the negative groups. CONCLUSIONS: Our findings suggested that anti-NeuGc Ab titers and NeuGc Ag expression in the HCC tissues can be used as the predictive factors for the postoperative recurrence and prognosis of HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/cirurgia , Antígenos Heterófilos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/cirurgia , Ácidos Neuramínicos/análise , Ácidos Neuramínicos/metabolismo , Biomarcadores Tumorais
7.
Appl Environ Microbiol ; 89(3): e0204722, 2023 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-36809058

RESUMO

Bacterial outer membrane vesicles (OMVs) are considered a promising vaccine platform for their high built-in adjuvanticity and ability to efficiently induce immune responses. OMVs can be engineered with heterologous antigens based on genetic engineering strategies. However, several critical issues should still be validated, including optimal exposure to the OMV surface, increased production of foreign antigens, nontoxicity, and induction of powerful immune protection. In this study, engineered OMVs with the lipoprotein transport machinery (Lpp) were designed to present SaoA antigen as a vaccine platform against Streptococcus suis. The results suggest that Lpp-SaoA fusions can be delivered on the OMV surface and do not have significant toxicity. Moreover, they can be engineered as lipoprotein and significantly accumulated in OMVs at high levels, thus accounting for nearly 10% of total OMV proteins. Immunization with OMVs containing Lpp-SaoA fusion antigen induced strong specific antibody responses and high levels of cytokines, as well as a balanced Th1/Th2 immune response. Furthermore, the decorated OMV vaccination significantly enhanced microbial clearance in a mouse infection model. It was found that antiserum against lipidated OMVs significantly promoted the opsonophagocytic uptake of S. suis in RAW246.7 macrophages. Lastly, OMVs engineered with Lpp-SaoA induced 100% protection against a challenge with 8× the 50% lethal dose (LD50) of S. suis serotype 2 and 80% protection against a challenge with 16× the LD50 in mice. Altogether, the results of this study provide a promising versatile strategy for the engineering of OMVs and suggest that Lpp-based OMVs may be a universal adjuvant-free vaccine platform for important pathogens. IMPORTANCE Bacterial outer membrane vesicles (OMVs) have become a promising vaccine platform due to their excellent built-in adjuvanticity properties. However, the location and amount of the expression of the heterologous antigen in the OMVs delivered by the genetic engineering strategies should be optimized. In this study, we exploited the lipoprotein transport pathway to engineer OMVs with heterologous antigen. Not only did lapidated heterologous antigen accumulate in the engineered OMV compartment at high levels, but also it was engineered to be delivered on the OMV surface, thus leading to the optimal activation of antigen-specific B cells and T cells. Immunization with engineered OMVs induced a strong antigen-specific antibodies in mice and conferred 100% protection against S. suis challenge. In general, the data of this study provide a versatile strategy for the engineering of OMVs and suggest that OMVs engineered with lipidated heterologous antigens may be a vaccine platform for significant pathogens.


Assuntos
Streptococcus suis , Vacinas , Animais , Camundongos , Streptococcus suis/genética , Streptococcus suis/metabolismo , Antígenos Heterófilos , Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Externa Bacteriana/metabolismo , Lipoproteínas/genética , Anticorpos Antibacterianos , Vacinas Bacterianas/genética
8.
Cell Transplant ; 32: 9636897221149444, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36644879

RESUMO

Clinically, xenotransplantation often leads to T-cell-mediated graft rejection. Immunosuppressive agents including polyclonal regulatory T cells (poly-Tregs) promote global immunosuppression, resulting in serious infections and malignancies in patients. Xenoantigen-expanded Tregs (xeno-Tregs) have become a promising immune therapy strategy to protect xenografts with fewer side effects. In this study, we aimed to identify an efficient and stable subset of xeno-Tregs. We enriched CD27+ xeno-Tregs using cell sorting and evaluated their suppressive functions and stability in vitro via mixed lymphocyte reaction (MLR), real-time polymerase chain reaction, inflammatory induction assay, and Western blotting. A STAT5 inhibitor was used to investigate the relationship between the function and stability of CD27+ xeno-Tregs and the JAK3-STAT5 signaling pathway. A humanized xenotransplanted mouse model was used to evaluate the function of CD27+ xeno-Tregs in vivo. Our results show that CD27+ xeno-Tregs express higher levels of Foxp3, cytotoxic T-lymphocyte antigen-4 (CTLA4), and Helios and lower levels of interleukin-17 (IL-17) than their CD27- counterparts. In addition, CD27+ xeno-Tregs showed enhanced suppressive function in xeno-MLR at ratios of 1:4 and 1:16 of Tregs:responder cells. Under inflammatory conditions, a lower percentage of CD27+ xeno-Tregs secretes IL-17 and interferon-γ (IFN-γ). CD27+ xeno-Tregs demonstrated an upregulated JAK3-STAT5 pathway compared with that of CD27- xeno-Tregs and showed decreased Foxp3, Helios, and CTLA4 expression after addition of STAT5 inhibitor. Mice that received porcine skin grafts showed a normal tissue phenotype and less leukocyte infiltration after reconstitution with CD27+ xeno-Tregs. Taken together, these data indicate that CD27+ xeno-Tregs may suppress immune responses in a xenoantigen-specific manner, which might be related to the activation of the JAK3-STAT5 signaling pathway.


Assuntos
Interleucina-17 , Linfócitos T Reguladores , Transplante Heterólogo , Animais , Humanos , Camundongos , Antígenos Heterófilos/metabolismo , Antígeno CTLA-4/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Interleucina-17/metabolismo , Ativação Linfocitária , Fator de Transcrição STAT5/metabolismo , Suínos , Linfócitos T Reguladores/imunologia
9.
J Virol ; 97(2): e0168422, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36651747

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant is becoming a dominant circulator and has several mutations in the spike glycoprotein, which may cause shifts of immunogenicity, so as to result in immune escape and breakthrough infection among the already infected or vaccinated populations. It is unclear whether infection with Omicron could generate adequate cross-variant protection. To investigate this possibility, we used Syrian hamsters as an animal model for infection of SARS-CoV-2. The serum from Omicron BA.1 variant-infected hamsters showed a significantly lower neutralization effect against infection of the same or different SARS-CoV-2 variants than the serum from Beta variant-infected hamsters. Furthermore, the serum from Omicron BA.1 variant-infected hamsters were insufficient to protect against rechallenge of SARS-CoV-2 Prototype, Beta and Delta variants and itself. Importantly, we found that rechallenge with different SARS-CoV-2 lineages elevated cross-variant serum neutralization titers. Overall, our findings indicate a weakened immunogenicity feature of Omicron BA.1 variant that can be overcome by rechallenge of a different SARS-CoV-2 lineages. Our results may lead to a new guideline in generation and use of the vaccinations to combat the pandemic of SARS-CoV-2 Omicron variant and possible new variants. IMPORTANCE The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant causes breakthrough infections among convalescent patients and vaccinated populations. However, Omicron does not generate robust cross-protective responses. Here, we investigate whether heterologous SARS-CoV-2 challenge is able to enhance antibody response in a sensitive animal model, namely, Syrian hamster. Of note, a heterologous challenge of Beta and Omicron BA.1 variant significantly broadens the breadth of SARS-CoV-2 neutralizing responses against the prototype, Beta, Delta, and Omicron BA.1 variants. Our findings confirm that vaccination strategy with heterologous antigens might be a good option to protect against the evolving SARS-CoV-2.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Cricetinae , Anticorpos Neutralizantes , Anticorpos Antivirais , Antígenos Heterófilos/imunologia , Infecções Irruptivas , COVID-19/prevenção & controle , Mesocricetus , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Imunogenicidade da Vacina
10.
Hum Immunol ; 84(1): 18-26, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35817653

RESUMO

BACKGROUND: Antibody-mediated rejection is a major cause of graft injury and contributes to failure of pig xenografts in nonhuman primates (NHPs). Most 'natural' or elicited antibodies found in humans and NHPs are directed against pig glycan antigens, but antibodies binding to swine leukocyte antigens (SLA) have also been detected. Of clinical importance is (i) whether the presence of high levels of antibodies directed towards human leukocyte antigens (HLA) (i.e., high panel-reactive antibodies) would be detrimental to the outcome of a pig organ xenograft; and (ii) whether, in the event of sensitization to pig antigens, a subsequent allotransplant would be at increased risk of graft failure due to elicited anti-pig antibodies that cross-react with human HLA or other antigens. SUMMARY: A literature review of pig-to-primate studies indicates that relatively few highly-HLA-sensitized humans have antibodies that cross-react with pigs, predicting that most would not be at increased risk of rejecting an organ xenograft. Furthermore, the existing evidence indicates that sensitization to pig antigens will probably not elicit increased alloantibody titers; if so, 'bridging' with a pig organ could be carried out without increased risk of subsequent antibody-mediated allograft failure. KEY MESSAGE: These issues have important implications for the design and conduct of clinical xenotransplantation trials.


Assuntos
Antígenos Heterófilos , Isoantígenos , Animais , Humanos , Transplante Heterólogo , Primatas , Antígenos , Antígenos HLA , Isoanticorpos , Rejeição de Enxerto
11.
Curr Protoc ; 2(10): e575, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36300895

RESUMO

The Sleeping Beauty (SB) transposon system is an efficient non-viral tool for gene transfer into a variety of cells, including human cells. Through a cut-and-paste mechanism, your favorite gene (YFG) is integrated into AT-rich regions within the genome, providing stable long-term expression of the transfected gene. The SB system is evolving and has become a powerful tool for gene therapy. There are no safety concerns using this system, the handling is easy, and the time required to obtain a stable cell line is significantly reduced compared to other systems currently available. Here, we present a novel application of this system to generate, within 8 days, a stable producer HEK293T cell line capable of constitutively delivering enveloped virus-like particles (eVLPs) for vaccination. We provide step-by-step protocols for generation of the SB transposon constructs, transfection procedures, and validation of the produced eVLPs. We next describe a method to pseudotype the constitutively produced eVLPs using the Spike protein derived from the SARS-CoV-2 virus (by coating the eVLP capsid with the heterologous antigen). We also describe optimization methods to scale up the production of pseudotyped eVLPs in a laboratory setting (from 100 µg to 5 mg). © Published 2022. This article is a U.S. Government work and is in the public domain in the USA. Basic Protocol 1: Generation of the SB plasmids Basic Protocol 2: Generation of a stable HEK293T cell line constitutively secreting MLV-based eVLPs Basic Protocol 3: Evaluation of the SB constructs by immunofluorescence assay Basic Protocol 4: Validation of eVLPs by denaturing PAGE and western blot Alternate Protocol 1: Analysis of SARS-CoV-2 Spike protein oligomerization using blue native gel electrophoresis and western blot Alternate Protocol 2: Evaluation of eVLP quality by electron microscopy (negative staining) Basic Protocol 5: Small-scale production of eVLPs Alternate Protocol 3: Large-scale production of eVLPs (up to about 1 to 3 mg VLPs) Alternate Protocol 4: Large-scale production of eVLPs (up to about 3 to 5 mg VLPs) Support Protocol: Quantification of total protein concentration by Bradford assay.


Assuntos
COVID-19 , Glicoproteína da Espícula de Coronavírus , Humanos , Glicoproteína da Espícula de Coronavírus/genética , SARS-CoV-2/genética , Células HEK293 , COVID-19/prevenção & controle , Vacinação , Antígenos Heterófilos
12.
Toxins (Basel) ; 14(10)2022 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-36287930

RESUMO

Ustilaginoidins are a class of bis-naphtho-γ-pyrone mycotoxins produced by the pathogen Villosiclava virens of rice false smut, which has recently become one of the most devastating diseases in rice-growing regions worldwide. In this research, the nanobody phage display library was established after an alpaca was immunized with the hemiustilaginoidin F-hapten coupled with bovine serum albumin (BSA). Heterologous antigen selection and combing trypsin with competition alternant elution methods were performed for nanobody screening. Two nanobodies, namely, Nb-B15 and Nb-C21, were selected for the establishment of indirect competitive enzyme-linked immunosorbent assays (ic-ELISAs). For Nb-B15 and Nb-C21, their IC50 values were 11.86 µg/mL and 11.22 µg/mL, and the detection ranges were at 3.41-19.98 µg/mL and 1.17-32.13 µg/mL, respectively. Two nanobodies had a broad spectrum to quantify the contents of total ustilaginoidins in rice samples according to cross-reactivity. The recognition mechanisms of Nb-B15 and Nb-C21 against ustilaginoidin A were elucidated by molecular modeling and docking. The key amino acid sites for the binding of Nb-B15 or Nb-C21 to ustilaginoidin A were mainly located in the FR1 and CDR1 regions. As Nb-B15 was superior to Nb-C21 in the aspects of protein expression, ELISA titer, and tolerance to organic solvents, it was selected for application in the detection of actual contaminated rice samples. The total ustilaginoidin contents of rice samples were analyzed by Nb-B15-based ic-ELISA and HPLC-DAD, between which the results were found to be consistent. The developed immunoassay based on the nanobody from the alpaca can be employed as a rapid and effective method for detection of total utilaginoidins in contaminated rice samples.


Assuntos
Camelídeos Americanos , Micotoxinas , Oryza , Anticorpos de Domínio Único , Animais , Oryza/química , Pironas , Soroalbumina Bovina , Tripsina , Micotoxinas/análise , Imunoensaio , Ensaio de Imunoadsorção Enzimática/métodos , Solventes , Haptenos , Aminoácidos , Antígenos Heterófilos
13.
J Environ Sci Health B ; 57(11): 883-889, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36217592

RESUMO

The heterologous strategy could improve the sensitivity of competitive enzyme-linked immunosorbent assay (ELISA) for detection of chemical contaminants in food samples. In this study, the heterologous coating antigen ELISA was developed to evaluate its sensitivity for mebendazole (MBZ). Results showed that the heterologous ELISA had a linear range of (IC20-IC80) 0.34-10.54 ng/mL, an IC50 value of 1.83 ng/mL, and a limit of detection (LOD) of 0.13 ng/mL, in which the sensitivity of ELISA improved 1.7- and 2-fold (IC50 value dropping from 7.41 and 3.65 ng/mL to 4.27 and 1.83 ng/mL) than that of rabbit IgG- and chicken IgY-based homologous ELISA for MBZ, respectively. The heterologous coating antigen ELISA showed negligible cross reactivity (<0.2%) with its structural analogues, including hydroxy-MBZ, albendazole, oxfendazole, fenbendazole, and flubendazole, except the value of 72.6% for amino-MBZ. The average recoveries of MBZ spiked in pork and chicken muscle samples by the assay ranged from 83.7% to 109.8% and agreed well with those of high-performance liquid chromatography. The results suggested that using heterologous coating antigen could distinctly improve the sensitivity of ELISA for routine screening of MBZ residues in food samples.


Assuntos
Antígenos Heterófilos , Mebendazol , Animais , Coelhos , Antígenos Heterófilos/análise , Ensaio de Imunoadsorção Enzimática/métodos , Cromatografia Líquida de Alta Pressão , Limite de Detecção
14.
Front Immunol ; 13: 910136, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35911728

RESUMO

We assessed if immune responses are enhanced in CD-1 mice by heterologous vaccination with two different nucleic acid-based COVID-19 vaccines: a next-generation human adenovirus serotype 5 (hAd5)-vectored dual-antigen spike (S) and nucleocapsid (N) vaccine (AdS+N) and a self-amplifying and -adjuvanted S RNA vaccine (AAHI-SC2) delivered by a nanostructured lipid carrier. The AdS+N vaccine encodes S modified with a fusion motif to increase cell-surface expression and an N antigen modified with an Enhanced T-cell Stimulation Domain (N-ETSD) to direct N to the endosomal/lysosomal compartment and increase MHC class I and II stimulation potential. The S sequence in the AAHI-SC2 vaccine comprises the D614G mutation, two prolines to stabilize S in the prefusion conformation, and 3 glutamines in the furin cleavage region to confer protease resistance. CD-1 mice received vaccination by homologous and heterologous prime > boost combinations. Humoral responses to S were the highest with any regimen that included the AAHI-SC2 vaccine, and IgG bound to wild type and Delta (B.1.617.2) variant S1 at similar levels. An AAHI-SC2 prime followed by an AdS+N boost particularly enhanced CD4+ and CD8+ T-cell responses to both wild type and Delta S peptides relative to all other vaccine regimens. Sera from mice receiving AAHI-SC2 homologous or heterologous vaccination were found to be highly neutralizing for all pseudovirus strains tested: Wuhan, Beta, Delta, and Omicron strains. The findings here, taken in consideration with the availability of both vaccines in thermostable formulations, support the testing of heterologous vaccination by an AAHI-SC2 > AdS+N regimen in animal models of SARS-CoV-2 infection to assess its potential to provide increased protection against emerging SARS-CoV-2 variants particularly in regions of the world where the need for cold-chain storage has limited the distribution of other vaccines.


Assuntos
COVID-19 , Vacinas Virais , Animais , Anticorpos Neutralizantes , Antígenos Heterófilos , COVID-19/prevenção & controle , Vacinas contra COVID-19 , DNA , Humanos , Camundongos , SARS-CoV-2 , Vacinação , Vacinas Sintéticas , Vacinas de mRNA
15.
J Appl Microbiol ; 133(4): 2484-2500, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35858677

RESUMO

AIMS: This study aimed to provide a safe, stable and efficient SARS-CoV-2 oral vaccine development strategy based on the type III secretion system of attenuated Salmonella and a reference for the development of a SARS-CoV-2 vaccine. METHODS AND RESULTS: The attenuated Salmonella mutant ΔhtrA-VNP was used as a vector to secrete the antigen SARS-CoV-2 based on the type III secretion system (T3SS). The Salmonella pathogenicity island 2 (SPI-2)-encoded T3SS promoter (sifB) was screened to express heterologous antigens (RBD, NTD, S2), and the SPI-2-encoded secretion system (sseJ) was employed to secrete this molecule (psifB-sseJ-antigen, abbreviated BJ-antigen). Both immunoblotting and fluorescence microscopy revealed effective expression and secretion of the antigen into the cytosol of macrophages in vitro. The mixture of the three strains (BJ-RBD/NTD/S2, named AisVax) elicited a marked increase in the induction of IgA or IgG S-protein Abs after oral gavage, intraperitoneal and subcutaneous administration. Flow cytometric analysis proved that AisVax caused T-cell activation, as shown by a significant increase in CD44 and CD69 expression. Significant production of IgA or IgG N-protein Abs was also detected by using psifB-sseJ-N(FL), indicating the universality of this strategy. CONCLUSIONS: Delivery of multiple SARS-CoV-2 antigens using the type III secretion system of attenuated Salmonella ΔhtrA-VNP is a potential COVID-19 vaccine strategy. SIGNIFICANCE AND IMPACT OF THE STUDY: The attenuated Salmonella strain ΔhtrA-VNP showed excellent performance as a vaccine vector. The Salmonella SPI-2-encoded T3SS showed highly efficient delivery of SARS-COV-2 antigens. Anti-loss elements integrated into the plasmid stabilized the phenotype of the vaccine strain. Mixed administration of antigen-expressing strains improved antibody induction.


Assuntos
COVID-19 , Sistemas de Secreção Tipo III , Antígenos Heterófilos/metabolismo , Proteínas de Bactérias/metabolismo , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Imunoglobulina A/metabolismo , Imunoglobulina G , SARS-CoV-2/genética , Salmonella typhimurium/genética , Sistemas de Secreção Tipo III/genética , Desenvolvimento de Vacinas
16.
BMC Genom Data ; 23(1): 54, 2022 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-35820824

RESUMO

BACKGROUND: Previous studies have identified the carbohydrate epitope Galα1-3Galß1-4GlcNAc-R (termed the α-galactosyl epitope), known as the α-Gal antigen as the primary xenoantigen recognized by the human immune system. The α-Gal antigen is regulated by galactosyltransferase (GGTA1), and α-Gal antigen-deficient mice have been widely used in xenoimmunological studies, as well as for the immunogenic risk evaluation of animal-derived medical devices. The objective of this study was to develop α-Gal antigen-deficient rabbits by GGTA1 gene editing with the CRISPR/Cas9 system. RESULTS: The mutation efficiency of GGTA1 gene-editing in rabbits was as high as 92.3% in F0 pups. Phenotype analysis showed that the α-Gal antigen expression in the major organs of F0 rabbits was decreased by more than 99.96% compared with that in wild-type (WT) rabbits, and the specific anti-Gal IgG and IgM antibody levels in F1 rabbits increased with increasing age, peaking at approximately 5 or 6 months. Further study showed that GGTA1 gene expression in F2-edited rabbits was dramatically reduced compared to that in WT rabbits. CONCLUSIONS: α-Gal antigen-deficient rabbits were successfully generated by GGTA1 gene editing via the CRISPR/Cas9 system in this study. The feasibility of using these α-Gal antigen-deficient rabbits for the in situ implantation and residual immunogenic risk evaluation of animal tissue-derived medical devices was also preliminarily confirmed.


Assuntos
Antígenos Heterófilos , Sistemas CRISPR-Cas , Animais , Sistemas CRISPR-Cas/genética , Edição de Genes , Humanos , Lactente , Camundongos , Coelhos
17.
Front Immunol ; 13: 893985, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35874735

RESUMO

Porcine islets surviving the acute injury caused by humoral rejection and IBMIR will be subjected to cellular xenograft rejection, which is predominately mediated by CD4+ T cells and is characterised by significant infiltration of macrophages, B cells and T cells (CD4+ and CD8+). Overall, the response is different compared to the alloimmune response and more difficult to suppress. Activation of CD4+ T cells is both by direct and indirect antigen presentation. After activation they recruit macrophages and direct B cell responses. Although they are less important than CD4+ T cells in islet xenograft rejection, macrophages are believed to be a major effector cell in this response. Rodent studies have shown that xenoantigen-primed and CD4+ T cell-activated macrophages were capable of recognition and rejection of pancreatic islet xenografts, and they destroyed a graft via the secretion of various proinflammatory mediators, including TNF-α, reactive oxygen and nitrogen species, and complement factors. B cells are an important mediator of islet xenograft rejection via xenoantigen presentation, priming effector T cells and producing xenospecific antibodies. Depletion and/or inhibition of B cells combined with suppressing T cells has been suggested as a promising strategy for induction of xeno-donor-specific T- and B-cell tolerance in islet xenotransplantation. Thus, strategies that expand the influence of regulatory T cells and inhibit and/or reduce macrophage and B cell responses are required for use in combination with clinical applicable immunosuppressive agents to achieve effective suppression of the T cell-initiated xenograft response.


Assuntos
Transplante das Ilhotas Pancreáticas , Animais , Antígenos Heterófilos , Rejeição de Enxerto , Xenoenxertos , Humanos , Imunidade Celular , Suínos , Transplante Heterólogo
18.
Front Immunol ; 13: 860165, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35493484

RESUMO

After producing triple (Gal, H-D and Sda)-KO pigs, hyperacute rejection appeared to no longer be a problem. However, the origin of xeno-rejection continues to be a controversial topic, including small amounts of antibodies and subsequent activation of the graft endothelium, the complement recognition system and the coagulation systems. The complement is activated via the classical pathway by non-Gal/H-D/Sda antigens and by ischemia-reperfusion injury (IRI), via the alternative pathway, especially on islets, and via the lectin pathway. The complement system therefore is still an important recognition and effector mechanism in xeno-rejection. All complement regulatory proteins (CRPs) regulate complement activation in different manners. Therefore, to effectively protect xenografts against xeno-rejection, it would appear reasonable to employ not only one but several CRPs including anti-complement drugs. The further assessment of antigens continues to be an important issue in the area of clinical xenotransplantation. The above conclusions suggest that the expression of sufficient levels of human CRPs on Triple-KO grafts is necessary. Moreover, multilateral inhibition on local complement activation in the graft, together with the control of signals between macrophages and lymphocytes is required.


Assuntos
Proteínas do Sistema Complemento , Rejeição de Enxerto , Animais , Antígenos Heterófilos , Ativação do Complemento , Proteínas do Sistema Complemento/fisiologia , Humanos , Suínos , Transplante Heterólogo
19.
J Helminthol ; 96: e21, 2022 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-35297359

RESUMO

Polycystic echinococcosis (PE) is a zoonosis endemic in the Neotropical region of the Americas. It is caused by the larval stage of the cestode Echinococcus vogeli, which develops as harmful cysts that slowly grow in the liver, lungs and other organs of humans and other host species. Human PE diagnosis is usually based on clinical and epidemiological aspects and imaging techniques, often requiring confirmation by immunological assays. The currently available serological tests for detecting antibodies against Echinococcus spp. are mostly based on complex, variable and poorly characterized mixtures of native parasite antigens, which impairs specificity and/or sensitivity. In this scenario, the evaluation of well-characterized alternative antigens is urgently needed for the improvement of PE diagnosis. Here, two subunits (AgB8/1 and AgB8/2) of the major secretory antigen from Echinococcus granulosus (antigen B (AgB)), of diagnostic value for cystic echinococcosis, were validated for PE diagnosis. These antigens, produced as pure recombinant proteins (rAgB8/1 and rAgB8/2) in Escherichia coli, allowed detecting specific immunoglobulin G antibodies in sera from PE patients in an enzyme-linked immunosorbent assay, with sensitivities of 83.72% and 81.40%, respectively, and specificities of 83.12% and 80.09%, respectively. The use of recombinant proteins overcomes difficulties to obtain parasite material and reduced non-specific reactions and costs. Our results demonstrated reproducibility and accuracy high enough to be considered valid according to the acceptance criteria for Food and Drug Administration assay validation. This qualifies rAgB8/1 and rAgB8/2 as potential substitutes for the currently used parasite crude or partially purified antigens.


Assuntos
Antígenos Heterófilos , Equinococose , Animais , Anticorpos Anti-Helmínticos , Antígenos de Helmintos/genética , Equinococose/parasitologia , Humanos , Reprodutibilidade dos Testes
20.
Transbound Emerg Dis ; 69(5): 2779-2787, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34919790

RESUMO

West Nile virus (WNV) and Usutu virus (USUV) are mosquito-borne viruses that belong to the Japanese encephalitis virus serocomplex within the genus Flavivirus. Due to climate change and the expansion of mosquito vectors, flaviviruses are becoming endemic in increasing numbers of countries. WNV infections are reported with symptoms ranging from mild fever to severe neuro-invasive disease. Until now, only a few USUV infections have been reported in humans, mostly with mild symptoms. The serological diagnosis and differentiation between flavivirus infections, in general, and between WNV and USUV, in particular, are challenging due to the high degree of cross-reacting antibodies, especially of those directed against the conserved fusion loop (FL) domain of the envelope (E) protein. We have previously shown that E proteins containing four amino-acid mutations in and near the FL strongly reduce the binding of cross-reactive antibodies leading to diagnostic technologies with improved specificities. Here, we expanded the technology to USUV and analyzed the differentiation of USUV- and WNV-induced antibodies in humans. IgG ELISAs modified by an additional competition step with the heterologous antigen resulted in overall specificities of 93.94% for WNV Equad and 92.75% for USUV Equad. IgM antibodies against WNV could be differentiated from USUV IgM in a direct comparison using both antigens. The data indicate the potential of the system to diagnose antigenically closely related flavivirus infections.


Assuntos
Infecções por Flavivirus , Flavivirus , Febre do Nilo Ocidental , Vírus do Nilo Ocidental , Animais , Anticorpos Antivirais , Antígenos Heterófilos , Epitopos , Flavivirus/genética , Infecções por Flavivirus/diagnóstico , Infecções por Flavivirus/epidemiologia , Infecções por Flavivirus/veterinária , Humanos , Imunoglobulina G , Imunoglobulina M , Febre do Nilo Ocidental/diagnóstico , Febre do Nilo Ocidental/epidemiologia , Febre do Nilo Ocidental/veterinária , Vírus do Nilo Ocidental/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...