Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Poult Sci ; 103(5): 103596, 2024 May.
Article in English | MEDLINE | ID: mdl-38471232

ABSTRACT

This study compared 2 herbal anticoccidiosis drugs (water-soluble and feed-additive drugs) with monensin coccidiostat, toltrazuril (TTZ, anticoccidiosis drug), and Livacox Q (anticoccidiosis vaccine) in terms of their effects on the prevention and treatment of coccidiosis in broilers. In this study, 280 Ross 308 broiler chickens (a mix of both genders) were used in a completely randomized design with 7 treatments and 5 replications each including 8 chickens per replicate. On d 21 of rearing, all experimental groups, except for the negative control group (NC), were challenged with a mixed suspension of common strains of Eimeria, and the intended indices were assessed, including performance indices, number of oocysts per gram (OPG) of feces, intestinal injuries, and the total number of intestinal bacteria. In addition, the NC and the group receiving the monensin had greater body weight gain (BWG) (P < 0.05). At the end of week 6, the monensin group had the highest feed intake (FI), while the water soluble medicine treatment resulted in the lowest feed intake (P < 0.05). Regarding the lesion scores on day 28, the highest and lowest rates of jejunal injuries were observed in the positive control group (PC), the monensin and vaccine group respectively. The rate of oocysts excretion (oocysts per gram of feces = OPG) on different days was higher in the PC group, and the use of monensin could further reduce excretion compared to the other groups (P > 0.05). Based on a comparison of the population of lactic acid bacteria between the NC and both medicinal plant treated groups, the use of these products could increase the population of these types of bacteria. Moreover, the population of Escherichia coli was less considerable in the NC and herbal powder groups (P < 0.05). Overall, similar to commercial medicines, the herbal medicines used in this project can be effective in the prevention and treatment of coccidiosis and can improve profitability in broiler rearing centers by improving intestinal health.


Subject(s)
Animal Feed , Chickens , Coccidiosis , Coccidiostats , Diet , Eimeria , Poultry Diseases , Protozoan Vaccines , Triazines , Animals , Coccidiosis/veterinary , Coccidiosis/prevention & control , Coccidiosis/parasitology , Coccidiostats/pharmacology , Coccidiostats/administration & dosage , Poultry Diseases/prevention & control , Poultry Diseases/parasitology , Triazines/pharmacology , Triazines/administration & dosage , Animal Feed/analysis , Male , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/pharmacology , Eimeria/physiology , Female , Diet/veterinary , Random Allocation , Dietary Supplements/analysis
2.
Front Immunol ; 12: 755792, 2021.
Article in English | MEDLINE | ID: mdl-35003067

ABSTRACT

Toxoplasma gondii can infect almost all endotherm organisms including humans and cause life-threatening toxoplasmosis in immunocompromised individuals, which leads to serious public health problems. Developing an excellent vaccine against this disease is impending. In present study, we formulated a cocktail protein vaccine including the TgMIF, TgCDPK3, and Tg14-3-3 proteins, which play critical roles in T. gondii infection. The recombinant protein vaccines were constructed and assessed by vaccination in BALB/c mice. We organized the mice in various protein combination groups of vaccines, and all mice were immunized with corresponding proteins at 0, 2, and 4 weeks. The specific protective effects of the vaccines on mice against T. gondii were analyzed by the mensuration of cytokines, serum antibodies, splenocyte proliferation assay, survival time, and parasite cyst burden of mice after the challenge. The study indicated that mice immunized with all three multicomponent proteins vaccine triggered a strong immune response with highest levels of IFN-γ production and IgG antibody compared with the other two protein combinations and controls. Moreover, there was an increase in IL-4 production and antigen-specific lymphocyte proliferation. The parasite cysts were significantly reduced (resulting in an 82.7% reduction), and survival time was longer in immunized mice with three multicomponent proteins compared with the other groups of mice. The enhanced humoral and cell-mediated immunity indicated that the protein cocktail vaccine containing three antigens provided effective protection for mice. These results indicated that recombinant TgMIF, TgCDPK3, and Tg14-3-3 multicomponent proteins were potential candidates for vaccine against toxoplasmosis.


Subject(s)
14-3-3 Proteins/immunology , Calcium-Binding Proteins/immunology , Macrophage Migration-Inhibitory Factors/immunology , Protozoan Proteins/immunology , Protozoan Vaccines/pharmacology , Toxoplasmosis, Animal/prevention & control , Animals , Female , Mice , Mice, Inbred BALB C , Protein Kinases/immunology , Recombinant Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology
3.
Poult Sci ; 99(8): 3831-3840, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32731969

ABSTRACT

A total of 960 male Cobb 500 broilers were used in a growth performance study to explore the effect of coccidial vaccination and/or coccidial challenge on blood biochemistry and veterinary postmortem metrics. Day-old chicks were randomly divided into one of the 4 experimental treatments. Treatments were arranged in a 2 × 2 factorial arrangement, with the factors being without or with vaccination (administered on day 1) or coccidial challenge (oral gavage on day 7). Growth performance was monitored on a weekly basis. Blood sample collection, as well as full veterinary necropsies, were carried out on days 6, 8, 13, 20, 27, and 34. Birds that did not receive the vaccination but were challenged with coccidiosis had higher feed conversion ratio, lower body weights, and higher mortality than the other experimental groups, and this effect was particularly evident from day 13 to day 20. Birds challenged with coccidiosis had lower plasma sodium and total carotenoid concentrations and higher potassium and globulin concentrations than nonchallenged birds. Significant interactions between age and experimental treatment for these blood parameters were observed, particularly on day 13. The necropsy results confirmed the effectiveness of the challenge and vaccination treatments, wherein birds that were challenged had higher coccidiosis scores on day 13 and day 27 than birds that were not challenged. These results demonstrate the potential for plasma sodium, potassium, total protein, total carbon dioxide, globulin, and carotenoid analysis for early diagnosis of coccidiosis in growing broiler chickens. Further work is necessary to establish whether the changes in blood biochemistry observed in the present study are transferable to alternative flocks of chicken and whether early diagnosis and intervention may mitigate performance losses associated with this disease.


Subject(s)
Coccidiosis , Eimeria , Poultry Diseases , Protozoan Vaccines , Animal Feed/analysis , Animals , Blood Chemical Analysis , Chickens , Coccidiosis/blood , Coccidiosis/immunology , Coccidiosis/prevention & control , Coccidiosis/veterinary , Diet , Male , Poultry Diseases/blood , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Protozoan Vaccines/blood , Protozoan Vaccines/immunology , Protozoan Vaccines/pharmacology , Random Allocation , Vaccination/veterinary
4.
BMC Infect Dis ; 20(1): 493, 2020 Jul 10.
Article in English | MEDLINE | ID: mdl-32650739

ABSTRACT

BACKGROUND: Toxoplasma gondii is an obligate intracellular parasite that can infect almost all warm-blooded animals, avian species and humans. Toxoplasmosis is asymptomatic in healthy individuals, whereas it may lead to death in immune suppressed or deficient patients. A vaccine against T. gondii is required to prevent consequences of the infection. The aim of this study is to generate a multivalent recombinant protein vaccine against T. gondii. METHODS: 49 previously discovered antigenic proteins of T gondii were evaluated by their expression level in E. coli and by comprehensive bioinformatics analyses to determine antigenic epitopes. Based on these analyses, six vaccine candidate proteins were selected to generate a hexavalent recombinant protein vaccine adjuvanted with Montanide ISA 50 V. Humoral and cellular immune responses were determined by flow cytometry and ELISA. Vaccinated mice were challenged with T. gondii Ankara strain tachyzoites. RESULTS: In mice vaccinated with hexavalent vaccine, strong total IgG (P < 0.0001) and IgG2a (P < 0.001) responses were induced compared to controls, the ratio of CD4+ and CD8+ T lymphocytes secreting IFN-γ increased, and significantly higher extracellular IFN-γ secretion was achieved compared to the controls (P < 0.001). The survival time of the vaccinated mice increased to 8.38 ± 2.13 days which was significantly higher than controls (P < 0.01). CONCLUSIONS: Altogether, these results show that the hexavalent vaccine which is developed for the first time against T. gondii induced strong and balanced Th1 and Th2 immune responses as well as conferred significant protection against challenge with lethal toxoplasmosis in murine model.


Subject(s)
Adjuvants, Immunologic/pharmacology , Mannitol/analogs & derivatives , Protozoan Vaccines/pharmacology , Toxoplasmosis/prevention & control , Vaccines, DNA/pharmacology , Animals , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Epitopes/immunology , Escherichia coli/genetics , Female , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunoglobulin G/blood , Mannitol/pharmacology , Mice , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Toxoplasma/pathogenicity , Toxoplasmosis/immunology , Vaccines, DNA/immunology
5.
Poult Sci ; 99(6): 2967-2975, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32475431

ABSTRACT

Avian coccidiosis causes significant economic losses on the global poultry breeding industry. Exploration of new-concept vaccines against coccidiosis has gradually become a research hotspot. In this study, an Enterococcus faecalis strain (MDXEF-1) showing excellent performance isolated from chicken intestinal tract was used as a vector to deliver Eimeria target protein. The plasmid pTX8048-SP-DCpep-NAΔ3-1E-CWA harboring dendritic cell-targeting peptide (DCpep) fusion with Eimeria tenella NAΔ3-1E gene (3-1E protein-coding gene without start codon ATG and terminator codon TAA) was electrotransformed into MDXEF-1 to generate the recombinant bacteria MDXEF-1/pTX8048-SP-DCpep-NAΔ3-1E-CWA in which NAΔ3-1E protein was covalently anchored to the surface of bacteria cells by cell wall anchor (CWA) sequence. The expression of target fusion protein DCpep-NAΔ3-1E-CWA was detected by Western blot. Each chicken was immunized 3 times at 2-wk intervals with live E. faecalis expressing DCpep-NAΔ3-1E fusion protein (DCpep-NAΔ3-1E group), live E. faecalis expressing NAΔ3-1E protein (NAΔ3-1E group), and live E. faecalis containing empty vector only. The 3 immunized groups were then challenged with homologous E. tenella sporulated oocyst after immunizations, and the immune response and protective efficacy in each group were evaluated. The results showed that serum IgG levels, secretory IgA levels in cecal lavage, proportion of CD4+ and CD8α+ cells in peripheral blood, and mRNA expression levels of IL-2 and IFN-γ in the spleen were significantly higher in chickens in the DCpep-NAΔ3-1E group than in chickens of the NAΔ3-1E group (P < 0.05). Oral immunization to chickens with live E. faecalis expressing DCpep-NAΔ3-1E offered more protective efficacy against homologous challenge including significant improved body weight gain, increased oocyst decrease ratio, and reduced average lesion scores in cecum compared with chickens with live E. faecalis expressing NAΔ3-1E protein. These results suggest that recombinant E. faecalis expressing dendritic cell-targeting peptide fusion with E. tenella 3-1E protein could be a potential approach for prevention of Eimeria infection.


Subject(s)
Chickens , Coccidiosis/veterinary , Eimeria tenella/immunology , Immunization/veterinary , Poultry Diseases/prevention & control , Protozoan Vaccines/pharmacology , Animals , Coccidiosis/immunology , Coccidiosis/prevention & control , Dendritic Cells , Enterococcus faecalis/genetics , Enterococcus faecalis/physiology , Immunity, Cellular , Immunity, Humoral , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/physiology , Peptides/metabolism , Poultry Diseases/immunology , Protozoan Proteins/administration & dosage , Protozoan Proteins/pharmacology , Protozoan Vaccines/administration & dosage , Recombinant Proteins , Specific Pathogen-Free Organisms
6.
Front Immunol ; 11: 578715, 2020.
Article in English | MEDLINE | ID: mdl-33732227

ABSTRACT

Asiatic schistosomiasis caused by Schistosoma japonicum is a neglected tropical disease resulting in significant morbidity to both humans and animals - particularly bovines - in endemic areas. Infection with this parasite leads to less healthy herds, causing problems in communities which rely on bovines for farming, milk and meat production. Additionally, excretion of parasite eggs in feces perpetuates the life cycle and can lead to human infection. We endeavored to develop a minimally purified, inexpensive, and effective vaccine based on the 80 kDa large subunit of the calcium activated neutral protease (calpain) from S. japonicum (Sj-p80). Here we describe the production of veterinary vaccine-grade Sj-p80 at four levels of purity and demonstrate in a pilot study that minimally purified antigen provides protection against infection in mice when paired with a low-cost veterinary adjuvant, Montanide™ ISA61 VG. Preliminary data demonstrate that the vaccine is immunogenic with robust antibody titers following immunization, and vaccination resulted in a reduction of parasite eggs being deposited in the liver (23.4-51.4%) and intestines (1.9-55.1%) depending on antigen purity as well as reducing the ability of these eggs to hatch into miracidia by up to 31.6%. We therefore present Sj-p80 as a candidate vaccine antigen for Asiatic schistosomiasis which is now primed for continued development and testing in bovines in endemic areas. A successful bovine vaccine could play a major role in reducing pathogen transmission to humans by interrupting the parasitic life cycle and improving quality of life for people living in endemic countries.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Helminth/pharmacology , Drug Development , Protozoan Vaccines/pharmacology , Schistosoma japonicum/pathogenicity , Schistosomiasis japonica/prevention & control , Veterinary Drugs/pharmacology , Adjuvants, Immunologic/economics , Animals , Antibodies, Helminth/blood , Antigens, Helminth/economics , Antigens, Helminth/immunology , Cattle , Cost-Benefit Analysis , Disease Models, Animal , Drug Costs , Female , Host-Pathogen Interactions , Immunogenicity, Vaccine , Mice, Inbred C57BL , Parasite Egg Count , Pilot Projects , Protozoan Vaccines/economics , Schistosoma japonicum/immunology , Schistosomiasis japonica/parasitology , Schistosomiasis japonica/transmission , Vaccination , Veterinary Drugs/economics
7.
Front Immunol ; 11: 595039, 2020.
Article in English | MEDLINE | ID: mdl-33414785

ABSTRACT

A parasitic protozoan Trypanosoma cruzi (T. cruzi) is the etiologic agent of Chagas disease. Previously, we have identified T. cruzi antigens TcG2 and TcG4 as potential vaccine candidates, cloned in eukaryotic expression vector pCDNA3.1 (referred as p2/4) and tested their ability to elicit protection from T. cruzi infection. In the present study, we subcloned the two antigens in a nanoplasmid that is optimized for delivery, antigen expression, and regulatory compliance standards, and evaluated the nanovaccine (referred as nano2/4) for prophylactic protection against repeat T. cruzi infections. For this, C57BL/6 mice were immunized with two doses of p2/4 or nano2/4 at 21 days interval, challenged with T. cruzi 21 days after 2nd immunization, and euthanized at 10- and 21-days post-infection (pi) corresponding to parasite dissemination and replication phase, respectively. Some mice were re-challenged 21 days pi and monitored at 7 days after re-infection. Without the help of a vaccine, T. cruzi elicited delayed and sub-par T cell activation and low levels of effector molecules that failed to control tissue dissemination and replication of the parasite and provided no protection against repeat challenge infection. The nano2/4 was most effective in eliciting an early activation and production of IFN-γ by CD4+T effector/effector memory (TEM) cells and cytolytic perforin (PFN) and granzyme B (GZB) molecules by CD4+ and CD8+ TEM subsets at 10 days pi that was followed by robust expansion of CD4+ and CD8+ TEM and TCM cells with further increase in IFN-γ production at 21 days pi. Consequently, nano2/4-immunized mice exhibited potent control of parasite dissemination at 10 days pi, and tissue parasite burden and tissue inflammatory infiltrate and necrosis were barely detectable at 21 days pi. Furthermore, nano2/4-immunized mice responded to re-challenge infection with high levels of effector molecules production by CD4+ and CD8+ TEM subpopulations that offered even better control of tissue parasite burden than was observed after 1st infection. In comparison, non-vaccinated/infected mice exhibited clinical features of sickness and 59% mortality within 7 days after re-infection. In conclusion, we show that delivery of TcG2 and TcG4 in nanoplasmid offers excellent, protective T cell immunity against repeat T. cruzi infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chagas Disease/immunology , Immunity, Cellular/drug effects , Lymphocyte Activation/drug effects , Protozoan Vaccines/pharmacology , Trypanosoma cruzi/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Line , Mice , Protozoan Vaccines/immunology
8.
Fish Shellfish Immunol ; 94: 308-317, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31470140

ABSTRACT

The channel catfish (Ictalurus punctatus) immune response against Ichthyophthirius multifiliis (Ich) after vaccination using plasmid DNA vaccines pcDNA3.1-IAg52a and pcDNA3.1-IAg52b, encoding Ich immobilization antigen genes was studied. Parasite infection level, serum anti-Ich antibodies level, fish mortality after theront challenge, and immune-related gene expression were measured. After in vitro transfection of walking catfish gill cells (G1b) with both pcDNA3.1-IAg52a and pcDNA3.1-IAg52b, antigens IAG52A and IAG52B were detected. During the vaccination trial, 76-fold increase in the Iag52b gene expression was observed in the vaccinated fish group h4 post vaccination. Administration of DNA vaccines by IM injection induced significant gene up-regulation in the head kidney, including immunoglobulin M (IgM), cluster of differentiation 4 (CD4), major histocompatibility I (MHC I), and T cell receptor α (TcR-α) from h4 to d5 post immunization. Fish vaccinated with DNA vaccines or theronts showed increased gene expression of the cytokine interferon (IFN-γ), complement component 3 (C3), and toll-like receptor-1 (TLR-1). Anti-Ich antibodies were detected in fish received pcDNA3.1-IAg52a, pcDNA3.1-IAg52b and the combination of both vaccines d10 post vaccination. Fish vaccinated with pcDNA3.1-IAg52b showed mild parasite infection level, partial survival (20%) and longer mean day to death (MDD) after theront challenge. By contrast, a heavy parasite load, 0% survival and short MDD were observed in the sham vaccinated control fish that received pcDNA3.1 (plasmid without genes encoding Ich immobilization antigen). Further research is needed to improve DNA vaccines for Ich that can induce strong protective immunity in fish. Suggested studies include improved transfection efficiency, use of appropriate adjuvants and including additional parasite antigen genes in the plasmid.


Subject(s)
Ciliophora Infections/veterinary , Fish Diseases/prevention & control , Hymenostomatida/immunology , Ictaluridae , Immunity, Innate , Protozoan Vaccines/pharmacology , Vaccination/veterinary , Adaptive Immunity , Animals , Antigens, Protozoan/pharmacology , Ciliophora Infections/immunology , Ciliophora Infections/prevention & control , Fish Diseases/immunology , Protozoan Proteins/pharmacology , Vaccines, DNA/pharmacology
9.
Front Immunol ; 10: 1456, 2019.
Article in English | MEDLINE | ID: mdl-31293599

ABSTRACT

Background: Chagas cardiomyopathy is caused by Trypanosoma cruzi (Tc). Two antigenic candidates, TcG2 and TcG4, are recognized by antibodies in naturally infected dogs and humans; and these vaccine candidates provided protection from Tc infection in mice and dogs. Trypanosoma rangeli (Tr) is non-pathogenic to mammals and shown to elicit cross-reactive anti-Tc antibodies. In this study, we investigated if fixed Tr (fTr) can further enhance the efficacy of the TcG2/TcG4 DNA vaccine. Methods and Results: C57BL/6 mice were immunized with TcG2/TcG4 DNA vaccine and fTr (delivered as an adjuvant or in prime-boost approach), and challenged with Tc. Serology studies showed that fTr (±quil-A) elicited Tc- and Tr-reactive IgGs that otherwise were not stimulated by TcG2/TcG4 vaccine only, and quil-A had suppressive effects on fTr-induced IgGs. After challenge infection, TcG2/TcG4-vaccinated mice exhibited potent expansion of antigen- and Tc-specific IgGs that were not boosted by fTr±quil-A. Flow cytometry analysis showed that TcG2/TcG4-induced dendritic cells (DC) and macrophages (Mφ) responded to challenge infection by expression of markers of antigen uptake, processing, and presentation, and production of pro-inflammatory cytokines. TcG2/TcG4-induced CD4+T cells acquired Th1 phenotype and expressed markers that orchestrate adaptive immunity. A fraction of vaccine-induced CD4+T cells exhibited iTreg phenotype responsible for aversion of self-injurious immune responses. Further, TcG2/TcG4-vaccinated mice exhibited potent expansion of poly-functional CD8+T cells with TNF-α/IFN-γ production and cytolytic phenotype post-infection. Subsequently, tissue parasites and pathology were hardly detectable in TcG2/TcG4-vaccinated/infected mice. Inclusion of fTr±quil-A had no clear additive effects in improving the Tc-specific adaptive immunity and parasite control than was noted in mice vaccinated with TcG2/TcG4 alone. Non-vaccinated mice lacked sufficient activation of Th1 CD4+/CD8+T cells, and exhibited >10-fold higher levels of tissue parasite burden than was noted in vaccinated/infected mice. Conclusion:TcG2/TcG4 vaccine elicits highly effective immunity, and inclusion of fTr is not required to improve the efficacy of DNA vaccine against acute Tc infection in mice.


Subject(s)
Antigens, Protozoan/pharmacology , Chagas Disease/prevention & control , Immunity, Cellular/drug effects , Immunization, Secondary , Protozoan Vaccines/pharmacology , Th1 Cells/immunology , Trypanosoma cruzi/immunology , Vaccines, DNA/pharmacology , Animals , Antigens, Protozoan/immunology , Chagas Disease/immunology , Chagas Disease/pathology , Female , Mice , Protozoan Vaccines/immunology , Th1 Cells/pathology , Vaccines, DNA/immunology
10.
Avian Dis ; 63(2): 342-350, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31251536

ABSTRACT

The current study was undertaken to assess the vaccine efficacy of Eimeria tenella EF-1α/chicken IL-7 (chIL-7) DNA vaccine when administered with Montanide Gel 01 adjuvant against live Eimeria acervulina challenge in commercial broiler chickens. The criteria used for the evaluation of vaccine efficacy were weight gain, duodenal lesion scores, oocyst counts, humoral antibody response, and duodenal proinflammatory cytokine gene expression. Chickens vaccinated with EF-1α (100 µg)/chIL-7 (20 µg) in Gel 01 PR adjuvant showed body weight gain similar to the uninfected control and higher oocyst shedding, a lower gut lesion score, and higher proinflammatory cytokine gene expression than did the infected controls. Moreover, chickens vaccinated with chIL-7 (20 µg) in Gel 01 PR adjuvant shed fewer oocysts with reduced gut lesion scores and produced higher levels of anti-EF-1α serum antibody than did the infected control. Chickens vaccinated with EF-1α (50 µg)/chIL-7 (20 µg) in Gel 01 PR adjuvant showed higher weight gains than did the infected control and shed significantly fewer oocysts than the infected control. Furthermore, chickens vaccinated with EF-1α (100 µg) in Gel 01 PR adjuvant demonstrated the lowest anti-EF-1α serum antibody levels. This study demonstrated the beneficial effects of using EF-1α and/or host cytokine chIL-7 DNA vaccine together with Gel 01 PR adjuvant to improve T-cell-mediated effector function in broiler chickens challenged with live E. acervulina.


Factor de alargamiento 1α (EF-1α) de Eimeria tenella administrado conjuntamente con vacuna de ADN de interleucina 7 de pollo (chIL-7) emulsionada en adyuvante de Montanide Gel 01 aumentó la respuesta inmune a la infección por E. acervulina en pollos de engorde. Este estudio se realizó para evaluar la eficacia de la vacuna con ADN de Eimeria tenella EF-1α /IL-7 de pollo (chIL-7) cuando se administró con el adyuvante Montanide Gel 01 contra el desafío con Eimeria acervulina viva en pollos de engorde comerciales. Los criterios utilizados para evaluar la eficacia de la vacuna fueron el aumento de peso, las puntuaciones de lesiones duodenales, los recuentos de ooquistes, la respuesta humoral de anticuerpos y la expresión de genes de citoquinas proinflamatorias duodenales. Los pollos vacunados con EF-1α (100 µg)/chIL-7 (20 µg) en el adyuvante Gel 01 PR mostraron un aumento de peso corporal similar al control no infectado y una mayor excreción de ooquistes, una puntuación más baja en la lesión intestinal y una mayor expresión de genes de citoquinas proinflamatorias en comparación con los controles infectados. Además, los pollos vacunados con chIL-7 (20 µg) en adyuvante Gel 01 PR eliminaron menos ooquistes con puntuaciones reducidas de lesión intestinal y produjeron niveles más altos de anticuerpos séricos anti-EF-1α en comparación con el control infectado. Los pollos vacunados con EF-1α (50 µg)/chIL-7 (20 µg) en adyuvante Gel 01 PR mostraron mayores ganancias de peso que el control infectado y eliminaron significativamente menos ooquistes que el control infectado. Además, los pollos vacunados con EF-1α (100 µg) en adyuvante Gel 01 PR demostraron los niveles más bajos de anticuerpos séricos anti-EF-1α. Este estudio demostró los efectos beneficiosos del uso de la vacuna de ADN EF-1α y/o la citoquina del huésped chIL-7 junto con el adyuvante Gel 01 PR para mejorar la función efectora mediada por células T en pollos de engorde desafiados con E. acervulina viva.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chickens , Coccidiosis/veterinary , Eimeria tenella/immunology , Peptide Elongation Factor 1/pharmacology , Poultry Diseases/prevention & control , Protozoan Vaccines/pharmacology , Animals , Coccidiosis/prevention & control , Eimeria/physiology , Immunity, Innate
11.
Sci Rep ; 9(1): 1474, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30728393

ABSTRACT

Almost any warm-blooded creature can be an intermediate host for Toxoplasma gondii. However, sexual reproduction of T. gondii occurs only in felids, wherein fertilisation of haploid macrogametes by haploid microgametes, results in diploid zygotes, around which a protective wall develops, forming unsporulated oocysts. Unsporulated oocysts are shed in the faeces of cats and meiosis gives rise to haploid sporozoites within the oocysts. These, now infectious, sporulated oocysts contaminate the environment as a source of infection for people and their livestock. RNA-Seq analysis of cat enteric stages of T. gondii uncovered genes expressed uniquely in microgametes and macrogametes. A CRISPR/Cas9 strategy was used to create a T. gondii strain that exhibits defective fertilisation, decreased fecundity and generates oocysts that fail to produce sporozoites. Inoculation of cats with this engineered parasite strain totally prevented oocyst excretion following infection with wild-type T. gondii, demonstrating that this mutant is an attenuated, live, transmission-blocking vaccine.


Subject(s)
Protozoan Proteins/genetics , Protozoan Vaccines/administration & dosage , Toxoplasma/genetics , Toxoplasmosis, Animal/prevention & control , Vaccines, Attenuated/administration & dosage , Animals , CRISPR-Cas Systems , Cats , Feces/parasitology , Female , Fertilization/drug effects , Gene Expression Profiling , Gene Expression Regulation , Gene Silencing , Male , Protozoan Vaccines/pharmacology , Sequence Analysis, RNA , Toxoplasma/drug effects , Toxoplasmosis, Animal/transmission , Vaccines, Attenuated/pharmacology
12.
Acta Trop ; 192: 22-29, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30664845

ABSTRACT

Toxoplasma gondii (T. gondii) is an obligatory intracellular parasite that can infect varieties of warm-blooded animals, including humans and birds. Heparan sulfate (HS) is widely distributed on the eukaryotic cell surface of vertebrates and can inhibit T. gondii invasion. In this study, we investigated the transcription and expression of the level of TgROP9, TgMIC3, and TgSAG2 in T. gondii RH strain, and found that the expression levels of these three proteins in invading parasites were higher compared to those free ranging parasites. The recombinant proteins showed specific binding activity to both heparin and host cell surface. Incubation of these proteins with the host cells could block T. gondiiinvasion. Furthermore, protein-specific antibodies also blocked parasite invasion. Antibodies in the sera of T. gondii infected individuals recognized the recombinant TgROP9, TgMIC3, and TgSAG2, which suggested the exposure of these proteins to human immune system. Mice immunized with the three proteins exhibited protective immunity against lethal challenge. The data collectively suggested that these parasitic proteins may be used as candidate antigens for development of anti-toxoplasmosis vaccine.


Subject(s)
Antigens, Protozoan/pharmacology , Heparitin Sulfate/immunology , Immunization/methods , Protozoan Vaccines/pharmacology , Toxoplasma/immunology , Animals , Antibodies, Protozoan , Antimicrobial Cationic Peptides , Blood Proteins , Carrier Proteins , Female , Membrane Glycoproteins/pharmacology , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Protozoan Proteins/pharmacology , Recombinant Proteins , Vaccines, DNA
13.
Microb Pathog ; 126: 172-184, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30399440

ABSTRACT

At present, there is not any available accepted vaccine for prevention of Toxoplasma gondii (T. gondii) in human and animals. We conducted literature search through English (Google Scholar, PubMed, Science Direct, Scopus, EBSCO, ISI Web of Science) scientific paper databases to find the best vaccine candidates against toxoplasmosis among T. gondii antigens. Articles with information on infective stage, pathogenicity, immunogenicity and characterization of antigens were selected. We considered that the ideal and significant vaccines should include different antigens and been expressed in all infective stages of the parasite with a high pathogenicity and immunogenicity. Evaluation within this systematic review indicates that MIC 3, 4, 13, ROP 2, RON 5, GRA 1, 6, 8, 14 are expressed in all three infective stages and have pathogenicity and immunogenicity. MIC 5, ROM 4, GRA 2, 4, 15, ROP 5, 16, 17, 38, RON 4, MIC 1, GRA 10, 12, 16, SAG 3 are expressed in only tachyzoites and bradyzoites stages of T. gondii with pathogenicity/immunogenicity. Some antigens appeared to be expressed in a single stage (tachyzoites) but have high pathogenicity and induce immune response. They include enolase2 (ENO2), SAG 1, SAG5D, HSP 70, ROM 1, ROM 5, AMA 1, ROP 18, RON2 and GRA 24. In conclusion, current vaccination against T. gondii infection is not satisfactory, and with the increasing number of high-risk individuals, the development of an effective and safe specific vaccine is greatly valuable for toxoplasmosis prevention. This systematic review reveals prepare candidates for immunization studies.


Subject(s)
Antigens, Protozoan/immunology , Immunization , Protozoan Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/prevention & control , Animals , Databases, Factual , Humans , Microbial Sensitivity Tests , Protozoan Vaccines/pharmacology , Toxoplasma/pathogenicity , Vaccination , Vaccines, DNA/pharmacology , Virulence
15.
Vet Parasitol ; 261: 73-76, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-30253853

ABSTRACT

Coccidiosis is a major health problem in rabbits. A vaccine using Eimeria with perfect safety and effectiveness seems to be necessary to face this parasitosis. To assess the safety and the efficacy of a vaccine based on the Algerian precocious line of Eimeria magna against rabbit coccidiosis, twenty eight young rabbits from six litters of Coccidia free females were used to monitor oocystal excretion and body weights, they were distributed into four groups (vaccinated-challenged group, double challenged non vaccinated group, simple challenged non vaccinated group and control group). Three other Coccidia free rabbits served for the necropsy in order to compare the effect of the wild and the precocious strains of Eimeria magna at the histological level. Following the challenge inoculation, a statistically significant decrease of about 97% in the oocyst excretion was noticed in the vaccinated rabbits as a sign of a good immune response acquired by the vaccination associated to a good growth rate. Moreover, a statistically significant increase in oocyst output following the challenge in both double challenged non vaccinated group and simple challenged non vaccinated one was noticed: (1.2 × 108 and 1.5 × 108vs 4.6 × 106 oocysts/rabbit respectively). Taking the control group showing a steady growth as a reference, the vaccinated rabbits showed a good growth during the experiment (p < 0.05). Globally the challenged groups showed a normal growth compared with the control group except for a temporary decrease in weights. No case of diarrhea was recorded in the vaccinated - challenged group and the control one (neither vaccinated nor challenged) whereas more than 50% of the young rabbits from both simple and double challenged - non vaccinated groups presented diarrhea. Consequently, the Algerian precocious strain of Eimeria magna constitute a good candidate for anticoccidian vaccine in the future.


Subject(s)
Coccidiosis/veterinary , Eimeria/immunology , Protozoan Vaccines/pharmacology , Rabbits , Vaccination/veterinary , Algeria , Animals , Coccidiosis/parasitology , Coccidiosis/prevention & control , Female , Protozoan Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/pharmacology
16.
Front Immunol ; 9: 1173, 2018.
Article in English | MEDLINE | ID: mdl-29896197

ABSTRACT

Recombinant filamentous fd bacteriophages (rfd) expressing antigenic peptides were shown to induce cell-mediated immune responses in the absence of added adjuvant, being a promising delivery system for vaccination. Here, we tested the capacity of rfd phages to protect against infection with the human protozoan Trypanosoma cruzi, the etiologic agent of Chagas Disease. For this, C57BL/6 (B6) and Tlr9-/- mice were vaccinated with rfd phages expressing the OVA257-264 peptide or the T. cruzi-immunodominant peptides PA8 and TSKB20 and challenged with either the T. cruzi Y-OVA or Y-strain, respectively. We found that vaccination with rfd phages induces anti-PA8 and anti-TSKB20 IgG production, expansion of Ag-specific IFN-γ, TNF-α, and Granzyme B-producing CD8+ T cells, as well as in vivo Ag-specific cytotoxic responses. Moreover, the fd-TSKB20 vaccine was able to protect against mortality induced by a high-dose inoculum of the parasite. Although vaccination with rfd phages successfully reduced both parasitemia and parasite load in the myocardium of WT B6 mice, Tlr9-/- animals were not protected against infection. Thus, our data extend previous studies, demonstrating that rfd phages induce Ag-specific IgG and CD8+ T cell-mediated responses and confer protection against an important human parasite infection, through a TLR9-dependent mechanism.


Subject(s)
Bacteriophage M13 , Chagas Disease , Gene Expression Regulation , Protozoan Vaccines , Toll-Like Receptor 9 , Trypanosoma cruzi , Vaccination , Animals , Bacteriophage M13/genetics , Bacteriophage M13/immunology , Chagas Disease/genetics , Chagas Disease/immunology , Chagas Disease/prevention & control , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Mice , Mice, Knockout , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Protozoan Vaccines/pharmacology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Trypanosoma cruzi/genetics , Trypanosoma cruzi/immunology
17.
BMC Vet Res ; 14(1): 145, 2018 May 02.
Article in English | MEDLINE | ID: mdl-29716583

ABSTRACT

BACKGROUND: The tick-borne protozoan parasite Theileria parva causes a usually fatal cattle disease known as East Coast fever in sub-Saharan Africa, with devastating consequences for poor small-holder farmers. Immunity to T. parva, believed to be mediated by a cytotoxic T lymphocyte (CTL) response, is induced following natural infection and after vaccination with a live vaccine, known as the Infection and Treatment Method (ITM). The most commonly used version of ITM is a combination of parasites derived from three isolates (Muguga, Kiambu 5 and Serengeti-transformed), known as the "Muguga cocktail". The use of a vaccine comprising several strains is believed to be required to induce a broad immune response effective against field challenge. In this study we investigated whether immunization with the Muguga cocktail induces a broader CTL response than immunization with a single strain (Muguga). RESULTS: Four MHC haplotype-matched pairs of cattle were immunized with either the trivalent Muguga cocktail or the single Muguga strain. CTL specificity was assessed on a panel of five different strains, and clonal responses to these strains were also assessed in one of the MHC-matched pairs. We did not find evidence for a broader CTL response in animals immunized with the Muguga cocktail compared to those immunized with the Muguga strain alone, in either the bulk or clonal CTL analyses. This was supported by an in vivo trial in which all vaccinated animals survived challenge with a lethal dose of the Muguga cocktail vaccine stabilate. CONCLUSION: We did not observe any substantial differences in the immunity generated from animals immunized with either Muguga alone or the Muguga cocktail in the animals tested here, corroborating earlier results showing limited antigenic diversity in the Muguga cocktail. These results may warrant further field studies using single T. parva strains as future vaccine candidates.


Subject(s)
Protozoan Vaccines/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Theileria parva/immunology , Theileriasis/prevention & control , Animals , Cattle , Genes, MHC Class I/immunology , Haplotypes , Major Histocompatibility Complex/immunology , Protozoan Vaccines/immunology , Species Specificity , T-Lymphocytes, Cytotoxic/drug effects , Theileriasis/immunology
18.
Vet Parasitol ; 254: 82-94, 2018 Apr 30.
Article in English | MEDLINE | ID: mdl-29657017

ABSTRACT

The aim of this study was to evaluate the immunogenic and immunoprotective activities and to determine the neuroprotective capacity of the tetravalent vaccine containing selected recombinant T. gondii antigens (ROP2 + ROP4 + SAG1 + MAG1) administered with safe adjuvants (MPL and alum) using male and female inbred mice. The tested antigenic combination provided partial protection against brain cyst formation, especially in males (reduction in cyst burden by 72%). The decrease in cyst burden was observed for the whole brain as well as for specified brain regions associated with natural defensive behaviors, emotion processing and integration of motor and sensory stimuli. The vaccine triggered a strong, specific immune response, regardless of sex, which was characterized by the antigen-specific in vitro synthesis of cytokines (IL-2, IFN-γ and IL-10) and in vivo production of systemic IgG1 and IgG2a immunoglobulins. Immunization prior to the parasite challenge seemed to influence T. gondii - associated behavioral and neurochemical changes, although the impact of vaccination strongly depended on sex and time post-infection. Interestingly, in the vaccinated and T. gondii infected mice there was a significant delay in the parasite-induced loss of aversion toward cat smell (cats are the definitive hosts of the parasite). The regained attraction toward feline scent in vaccinated males, observed during chronic parasite invasion, correlated with the increase in the dopamine metabolism.


Subject(s)
Protozoan Vaccines/pharmacology , Toxoplasma/immunology , Toxoplasmosis, Animal/prevention & control , Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Animals , Antigens, Protozoan/immunology , Female , Male , Mice , Toxoplasmosis, Animal/immunology , Vaccines, Subunit/immunology
19.
Front Immunol ; 9: 317, 2018.
Article in English | MEDLINE | ID: mdl-29515595

ABSTRACT

Toxoplasmosis is a major public health problem and the development of a human vaccine is of high priority. Efficient vaccination against Toxoplasma gondii requires both a mucosal and systemic Th1 immune response. Moreover, dendritic cells play a critical role in orchestrating the innate immune functions and driving specific adaptive immunity to T. gondii. In this study, we explore an original vaccination strategy that combines administration via mucosal and systemic routes of fusion proteins able to target the major T. gondii surface antigen SAG1 to DCs using an antibody fragment single-chain fragment variable (scFv) directed against DEC205 endocytic receptor. Our results show that SAG1 targeting to DCs by scFv via intranasal and subcutaneous administration improved protection against chronic T. gondii infection. A marked reduction in brain parasite burden is observed when compared with the intranasal or the subcutaneous route alone. DC targeting improved both local and systemic humoral and cellular immune responses and potentiated more specifically the Th1 response profile by more efficient production of IFN-γ, interleukin-2, IgG2a, and nasal IgA. This study provides evidence of the potential of DC targeting for the development of new vaccines against a range of Apicomplexa parasites.


Subject(s)
Antigens, Protozoan/pharmacology , Dendritic Cells/immunology , Drug Delivery Systems , Immunogenicity, Vaccine , Protozoan Vaccines/pharmacology , Toxoplasma/immunology , Toxoplasmosis/prevention & control , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Dendritic Cells/pathology , Female , Mice , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Toxoplasma/genetics , Toxoplasmosis/genetics , Toxoplasmosis/immunology , Toxoplasmosis/pathology
20.
Adv Healthc Mater ; 7(13): e1701494, 2018 07.
Article in English | MEDLINE | ID: mdl-29602254

ABSTRACT

Nanomedicine approaches have the potential to transform the battle against parasitic worm (helminth) infections, a major global health scourge from which billions are currently suffering. It is anticipated that the intersection of two currently disparate fields, nanomedicine and helminth biology, will constitute a new frontier in science and technology. This progress report surveys current innovations in these research fields and discusses research opportunities. In particular, the focus is on: (1) major challenges that helminth infections impose on mankind; (2) key aspects of helminth biology that inform future research directions; (3) efforts to construct nanodelivery platforms to target drugs and genes to helminths hidden in their hosts; (4) attempts in applying nanotechnology to enable vaccination against helminth infections; (5) outlooks in utilizing nanoparticles to enhance immunomodulatory activities of worm-derived factors to cure allergy and autoimmune diseases. In each section, achievements are summarized, limitations are explored, and future directions are assessed.


Subject(s)
Helminthiasis/therapy , Helminths/pathogenicity , Host-Parasite Interactions/immunology , Nanomedicine/methods , Protozoan Vaccines/pharmacology , Animals , Drug Delivery Systems/methods , Drug Resistance , Gene Transfer Techniques , Helminthiasis/prevention & control , Helminths/drug effects , Helminths/physiology , Humans , Immunotherapy , Nanoparticles/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...