Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Parasitol Res ; 116(11): 3175-3188, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29026995

RESUMO

Schistosomiasis remains a major global health problem. Despite large-scale schistosomiasis control efforts, clear limitations such as possible emergence of drug resistance and reinfection rates highlight the need for an effective schistosomiasis vaccine. Schistosoma mansoni large subunit of calpain (Sm-p80)-based vaccine formulations have shown remarkable efficacy in protecting against S. mansoni challenge infections in mice and baboons. In this study, we evaluated the cross-species protective efficacy of Sm-p80 vaccine against S. japonicum and S. haematobium challenge infections in rodent models. We also elucidated the expression of Sm-p80 and Sm-p80 ortholog proteins in different developmental stages of S. mansoni, S. haematobium, and S. japonicum. Immunization with Sm-p80 vaccine reduced worm burden by 46.75% against S. japonicum challenge infection in mice. DNA prime/protein boost (1 + 1 dose administered on a single day) resulted in 26.95% reduction in worm burden in S. haematobium-hamster infection/challenge model. A balanced Th1 (IFN-γ, TNF-α, IL-2, and IL-12) and Th2 (IL-4, IgG1) type of responses were observed following vaccination in both S. japonicum and S. haematobium challenge trials and these are associated with the prophylactic efficacy of Sm-p80 vaccine. Immunohistochemistry demonstrated that Sm-p80/Sm-p80 ortholog proteins are expressed in different life cycle stages of the three major human species of schistosomes studied. The data presented in this study reinforce the potential of Sm-p80-based vaccine for both hepatic/intestinal and urogenital schistosomiasis occurring in different geographical areas of the world. Differential expression of Sm-p80/Sm-p80 protein orthologs in different life cycle makes this vaccine potentially useful in targeting different levels of infection, disease, and transmission.


Assuntos
Antígenos de Helmintos/imunologia , Vacinas Protozoárias/imunologia , Schistosoma haematobium/imunologia , Schistosoma japonicum/imunologia , Schistosoma mansoni/imunologia , Esquistossomose Urinária/prevenção & controle , Esquistossomose Japônica/prevenção & controle , Esquistossomose mansoni/prevenção & controle , Animais , Anticorpos Anti-Helmínticos/imunologia , Calpaína/imunologia , Cricetinae , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina G/imunologia , Interleucina-12/biossíntese , Interleucina-2/biossíntese , Interleucina-4/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Papio , Schistosoma haematobium/crescimento & desenvolvimento , Schistosoma japonicum/crescimento & desenvolvimento , Schistosoma mansoni/crescimento & desenvolvimento , Esquistossomose Urinária/imunologia , Esquistossomose Urinária/parasitologia , Esquistossomose Japônica/imunologia , Esquistossomose Japônica/parasitologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia , Fator de Necrose Tumoral alfa/biossíntese , Vacinação , Vacinas de DNA/imunologia
2.
Exp Parasitol ; 153: 139-50, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25819297

RESUMO

Many chronic inflammatory diseases can be improved by helminth infection, but the mechanisms are poorly understood. Allergy and helminthiasis are both associated with Th2-like immune responses; thus, defining how infection with parasites leads to reduced allergy has been particularly challenging. We sought to better understand this conundrum by evaluating host-parasite interactions involved in Th2 immunity in human schistosomiasis. Immune cells were cultured with schistosomes and the effect on CD23, an IgE receptor associated with resistance in schistosomiasis, was evaluated. Cells treated with schistosomes demonstrated reduced surface CD23 levels with a parallel accumulation of soluble (s) CD23 suggesting this IgE receptor is proteolytically cleaved by the parasite. Consistent with this hypothesis, a schistosome-generated (SG)-sCD23 fragment of 15 kDa was identified. SG-sCD23 inhibited IgE from binding to CD23 and FcεRI, but lacked the ability to bind CD21. These results suggested that schistosomes target IgE-mediated immunity in immuno-evasive tactics. Based on its characteristics, we predicted that SG-sCD23 would function as an efficacious allergy preventative. Treatment of human FcεRI-transgenic mice with recombinant (r) SG-sCD23 reduced the ability of human IgE to induce an acute allergic response in vivo. In addition, an optimized form of rSG-sCD23 with an introduced point mutation at Asp258 (D258E)to stabilize IgE binding had increased efficacy compared to native rSG-sCD23. Schistosome infection may thus inhibit allergic-like protective immune responses by increasing soluble IgE decoy receptors. Allergy treatments based on this naturally occurring phenomenon may be highly effective and have fewer side effects with long-term use.


Assuntos
Hipersensibilidade/prevenção & controle , Evasão da Resposta Imune , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Adulto , Animais , Interações Hospedeiro-Parasita , Humanos , Hipersensibilidade/imunologia , Imunoglobulina E/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Receptores de IgE/administração & dosagem , Receptores de IgE/genética , Receptores de IgE/imunologia , Schistosoma mansoni/genética , Esquistossomose mansoni/genética , Esquistossomose mansoni/parasitologia , Células Th2/imunologia , Adulto Jovem
3.
J Infect Dis ; 209(12): 1929-40, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24436452

RESUMO

No vaccines are available for human use for any parasitic infections, including the helminthic disease schistosomiasis. Sm-p80, the large subunit of Schistosoma mansoni calpain, is a leading antigen candidate for a schistosomiasis vaccine. Prophylactic and antifecundity efficacies of Sm-p80 have been tested using a variety of vaccine approaches in both rodent and nonhuman primate models. However, the therapeutic efficacy of a Sm-p80-based vaccine had not been determined. In this study, we evaluated the therapeutic efficacy of Sm-p80 by using 2 different strategies and 3 Sm-p80-based vaccine formulations in baboons. Vaccine formulations were able to decrease established adult worms by 10%-36%, reduce retention of eggs in tissues by 10%-57%, and decrease egg excretion in feces by 13%-33%, compared with control formulations. Marked differences were observed in B and T cell immune correlates between vaccinated and control animals. This is the first report of killing of established adult schistosome worms by a vaccine. In addition to distinct prophylactic efficacy of Sm-p80, this study adds to the evidence that Sm-p80 is a potentially important antigen with both substantial prophylactic and therapeutic efficacies. These data reinforce that Sm-p80 should be moved forward along the path toward human clinical trials.


Assuntos
Antígenos de Helmintos/imunologia , Calpaína/imunologia , Papio/parasitologia , Esquistossomose mansoni/tratamento farmacológico , Vacinas/imunologia , Animais , Anticorpos Anti-Helmínticos/sangue , Anticorpos Anti-Helmínticos/imunologia , Linfócitos B/imunologia , Modelos Animais de Doenças , Fezes/parasitologia , Feminino , Interferon gama/sangue , Interleucina-17/sangue , Interleucina-4/sangue , Leucócitos Mononucleares/imunologia , Masculino , Contagem de Ovos de Parasitas , Schistosoma mansoni/efeitos dos fármacos , Linfócitos T/imunologia , Vacinação
4.
Parasitol Res ; 113(3): 1195-200, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24452916

RESUMO

Prophylactic efficacy of Sm-p80 was tested in the mouse model using DNA priming and boosting with protein approach. However, the novelty of the approach utilized in this study is that both the DNA priming and protein boosting was performed on a single day and no further vaccine inoculations were given to mice; the animals were challenged 1 month after the initial vaccine administration. Using this approach, significant reduction in worm burden (33 to 57 %) and marked decrease in egg retention in tissues (34 to 66%) was observed. Robust antibody titers and upregulation of cytokines (IL-1α/ß, IL-12α, and IFN-γ) appears to correlate with the protection. This approach of administering vaccine on a single day could be greatly helpful in the field setting because it will eliminate the compliance issues that may arise with multiple boosters that may be required for optimal efficacy for some vaccines.


Assuntos
Antígenos de Helmintos/imunologia , Proteínas de Helminto/imunologia , Esquistossomose mansoni/prevenção & controle , Vacinas de DNA/imunologia , Animais , Anticorpos Anti-Helmínticos/sangue , Antígenos de Helmintos/genética , Citocinas/imunologia , Modelos Animais de Doenças , Feminino , Proteínas de Helminto/genética , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/administração & dosagem , Contagem de Ovos de Parasitas , Schistosoma mansoni , Baço/imunologia , Vacinação/métodos , Vacinas de DNA/administração & dosagem
5.
Parasitol Res ; 113(6): 2239-50, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24728521

RESUMO

Based on data obtained using vaccine efficacy studies in mice, hamsters, and baboons, the credentials of Sm-p80 as a first tier vaccine candidate for schistosomiasis have been well established. Sm-p80-based vaccine formulation(s) have consistently exhibited potent prophylactic efficacy in reducing adult worm burden following cercarial challenge and induce killing of established adult worms in chronic infection. This vaccine is protective against both intestinal and urinary schistosomiasis. In this study, the longevity of Sm-p80-specific antibody responses was studied in mice and in baboons. Robust antibody titers were detected in mice for up to 60 weeks following vaccination with Sm-p80 recombinant vaccine (Sm-p80 + GLA-SE). In the follow-up experiments to our published studies, Sm-p80-specific IgG was also detected in baboons 5-8 years following the initial vaccination with an Sm-p80 DNA vaccine. In one baboon, transfer of Sm-p80-specific antibody was detected in umbilical cord blood and in the baby. These long-lasting humoral immune response data coupled with the vaccine efficacy data in rodents and nonhuman primates further strengthens the case for Sm-p80 to be moved forward through development leading to human clinical trials.


Assuntos
Anticorpos Anti-Helmínticos/sangue , Schistosoma mansoni/imunologia , Esquistossomose mansoni/prevenção & controle , Vacinas/imunologia , Animais , Antígenos de Helmintos/imunologia , Biomphalaria/parasitologia , Cricetinae , Feminino , Sangue Fetal/química , Sangue Fetal/imunologia , Imunidade Materno-Adquirida , Imunoglobulina G/sangue , Camundongos , Papio , Gravidez , Schistosoma mansoni/genética , Vacinação , Vacinas de DNA/imunologia
6.
Immunother Adv ; 3(1): ltad010, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37538934

RESUMO

Control of schistosomiasis depends on a single drug, praziquantel, with variable cure rates, high reinfection rates, and risk of drug resistance. A vaccine could transform schistosomiasis control. Preclinical data show that vaccine development is possible, but conventional vaccine efficacy trials require high incidence, long-term follow-up, and large sample size. Controlled human infection studies (CHI) can provide early efficacy data, allowing the selection of optimal candidates for further trials. A Schistosoma CHI has been established in the Netherlands but responses to infection and vaccines differ in target populations in endemic countries. We aim to develop a CHI for Schistosoma mansoni in Uganda to test candidate vaccines in an endemic setting. This is an open-label, dose-escalation trial in two populations: minimal, or intense, prior Schistosoma exposure. In each population, participants will be enrolled in sequential dose-escalating groups. Initially, three volunteers will be exposed to 10 cercariae. If all show infection, seven more will be exposed to the same dose. If not, three volunteers in subsequent groups will be exposed to higher doses (20 or 30 cercariae) following the same algorithm, until all 10 volunteers receiving a particular dose become infected, at which point the study will be stopped for that population. Volunteers will be followed weekly after infection until CAA positivity or to 12 weeks. Once positive, they will be treated with praziquantel and followed for one year. The trial registry number is ISRCTN14033813 and all approvals have been obtained. The trial will be subjected to monitoring, inspection, and/or audits.

7.
J Infect Dis ; 204(9): 1437-49, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21921206

RESUMO

The prophylactic efficacy of a schistosome antigen (Sm-p80) was tested in a nonhuman primate model, the baboon. Using a total of 28 baboons, different vaccination strategies were used including recombinant Sm-p80 protein formulated in Toll-like receptor 7 and Toll-like receptor 9 agonists, and DNA priming followed by boosting with protein plus adjuvants. Recombinant protein approaches provided levels of prophylactic efficacy of 52%-58%, whereas prime-boost approaches conferred 38%-47% protection in baboons. An appropriately balanced pro-inflammatory (T-helper 17 [Th17] and Th1) and anti-inflammatory (Th2) type of response was generated; the Th1 and Th17 types of immune responses appear to be indicative of increased prophylactic efficacy. Production and expression of several cytokines (interleukin 2 [IL-2], interferon γ, IL-12α, IL-1ß, IL-6, and IL-22) were up-regulated in vaccinated animals. Human correlate studies revealed Sm-p80 reactivity with immunoglobulin G in human serum samples from schistosome-infected individuals. In addition, a complete lack of prevailing Sm-p80-specific immunoglobulin E in a high-risk or infected population was observed, thus minimizing the risk of hypersensitivity reaction following vaccination with Sm-p80 in humans. This study provided the proof of concept to move Sm-p80 forward into further preclinical development leading to human clinical trials.


Assuntos
Anticorpos Anti-Helmínticos/sangue , Antígenos de Helmintos/imunologia , Doenças Endêmicas , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/prevenção & controle , Vacinas de DNA/imunologia , Animais , Antígenos de Helmintos/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Masculino , Papio , Doenças dos Primatas/imunologia , Doenças dos Primatas/prevenção & controle , Soro/imunologia , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
8.
Hum Vaccin ; 7(11): 1192-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22048120

RESUMO

Schistosomiasis is a major neglected tropical disease of public health importance to a billion people. An estimated 200 million people are currently infected; an additional 779 million individuals are at risk to acquire the infection in 74 countries. Despite many years of implementation of mass anti-parasitic drug therapy programs and other control measures, this disease has not been contained and continues to spread to new geographic areas. The discovery of a protective vaccine still remains the most potentially effective means for the control of this disease, especially if the vaccine provides long-term immunity against the infection. A vaccine would contribute to the reduction of schistosomiasis morbidity through induced immune responses leading to decrease in parasite load and reduced egg production. This vaccine could be administered to children between the ages of 3 and 12 years to prevent severe infection in a particularly high risk population. This review summarizes the current status of schistosomiasis vaccine development.


Assuntos
Antígenos de Helmintos/imunologia , Esquistossomose/prevenção & controle , Vacinas/imunologia , Animais , Criança , Pré-Escolar , Humanos , Doenças Negligenciadas , Carga Parasitária , Esquistossomose/imunologia , Esquistossomose/parasitologia , Medicina Tropical , Vacinas/administração & dosagem
9.
J Infect Dis ; 201(7): 1105-12, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20187746

RESUMO

To date, no vaccine is available to prevent human schistosomiasis. We have targeted a protein of Schistosoma mansoni that plays an important role in the surface membrane renewal process, a mechanism widely believed to be utilized by the parasite as an immune evasion strategy. Sm-p80 antigen is a promising vaccine target because of its documented immunogenicity, protective efficacy, and antifecundity effects observed in both experimental murine and nonhuman primate models of this infectious disease. In the present study, we report that, in a vector approved for human use (VR1020), an Sm-p80-based DNA vaccine formulation confers a 46% reduction in the worm burden in a baboon (Papio anubis) model. Baboons vaccinated with Sm-p80-VR1020 had a 28% decrease in egg production after challenge with the infectious parasite. Sm-p80-VR1020 vaccine elicited robust immune responses to specific antigen Sm-p80, including immunoglobulin (Ig) G, its subtypes IgG1 and IgG2, and IgA and IgM in vaccinated animals. When stimulated in vitro with recombinant Sm-p80, peripheral blood mononuclear cells and splenocytes from baboons vaccinated with Sm-p80-VR1020 produced considerably higher levels of T helper 1 response-enhancing cytokines (interleukin [IL]-2 and interferon-gamma) than T helper 2 (Th2) response-enhancing cytokines (IL-4 and IL-10). Peripheral blood mononuclear cells produced a significantly higher number of spot-forming units for interferon-gamma than for IL-4 in enzyme-linked immunosorbent spot assays. A mixed T helper 1/T helper 2 type of humoral and T cell responses was generated after immunization with Sm-p80-VR1020. These findings again highlight the potential of Sm-p80 as a promising vaccine candidate for schistosomiasis.


Assuntos
Antígenos de Helmintos/imunologia , Papio/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/prevenção & controle , Vacinas de DNA/imunologia , Animais , Anticorpos Anti-Helmínticos/sangue , Antígenos de Helmintos/genética , Células CHO , Células COS , Proliferação de Células , Chlorocebus aethiops , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Imunoglobulina G/sangue , Interleucina-4/metabolismo , Intestinos/parasitologia , Leucócitos Mononucleares/metabolismo , Fígado/parasitologia , Contagem de Ovos de Parasitas , Schistosoma mansoni/genética , Esquistossomose mansoni/imunologia , Linfócitos T/imunologia , Vacinas de DNA/administração & dosagem
10.
Front Immunol ; 12: 635985, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33746974

RESUMO

Despite mass drug administration programmes with praziquantel, the prevalence of schistosomiasis remains high. A vaccine is urgently needed to control transmission of this debilitating disease. As some promising schistosomiasis vaccine candidates are moving through pre-clinical and clinical testing, we review the immunological challenges that these vaccine candidates may encounter in transitioning through the clinical trial phases in endemic settings. Prior exposure of the target population to schistosomes and other infections may impact vaccine response and efficacy and therefore requires considerable attention. Schistosomes are known for their potential to induce T-reg/IL-10 mediated immune suppression in populations which are chronically infected. Moreover, endemicity of schistosomiasis is focal whereby target and trial populations may exhibit several degrees of prior exposure as well as in utero exposure which may increase heterogeneity of vaccine responses. The age dependent distribution of exposure and development of acquired immunity, and general differences in the baseline immunological profile, adds to the complexity of selecting suitable trial populations. Similarly, prior or concurrent infections with other parasitic helminths, viral and bacterial infections, may alter immunological responses. Consequently, treatment of co-infections may benefit the immunogenicity of vaccines and may be considered despite logistical challenges. On the other hand, viral infections leave a life-long immunological imprint on the human host. Screening for serostatus may be needed to facilitate interpretation of vaccine responses. Co-delivery of schistosome vaccines with PZQ is attractive from a perspective of implementation but may complicate the immunogenicity of schistosomiasis vaccines. Several studies have reported PZQ treatment to induce both transient and long-term immuno-modulatory effects as a result of tegument destruction, worm killing and subsequent exposure of worm antigens to the host immune system. These in turn may augment or antagonize vaccine immunogenicity. Understanding the complex immunological interactions between vaccine, co-infections or prior exposure is essential in early stages of clinical development to facilitate phase 3 clinical trial design and implementation policies. Besides well-designed studies in different target populations using schistosome candidate vaccines or other vaccines as models, controlled human infections could also help identify markers of immune protection in populations with different disease and immunological backgrounds.


Assuntos
Desenvolvimento de Medicamentos , Doenças Endêmicas/prevenção & controle , Vacinas Protozoárias/uso terapêutico , Schistosoma/imunologia , Esquistossomose/prevenção & controle , Animais , Coinfecção , Desenho de Fármacos , Interações Hospedeiro-Parasita , Humanos , Imunogenicidade da Vacina , Praziquantel/uso terapêutico , Vacinas Protozoárias/efeitos adversos , Vacinas Protozoárias/imunologia , Schistosoma/patogenicidade , Esquistossomose/epidemiologia , Esquistossomose/imunologia , Esquistossomose/transmissão , Esquistossomicidas/uso terapêutico
11.
Methods Mol Biol ; 2151: 229-262, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32452009

RESUMO

Vaccination has greatly reduced the burden of human diseases caused by infectious pathogens. Systematic development of vaccine targets requires established protocols to assess immunogenicity and efficacy of such vaccine candidates. Using a leading schistosomiasis vaccine candidate, Sm-p80, as an example, we describe standardized approaches for testing the immunogenicity and efficacy of Schistosoma mansoni vaccine targets. Unlike other parasite systems in which sterile immunity is required, the goal of S. mansoni vaccine targets is overall reduction in morbidity. Methods related to the parasitological parameters described in this chapter allow for the testing of the prophylactic (reduction in adult worm burden), anti-pathology (liver and intestine egg retention), and transmission blocking (fecal egg expulsion and egg hatching rates) efficacies for the vaccine target. The RNA sequencing approaches provide basis for identification of molecular signatures predictive of desirable outcomes for schistosomiasis vaccines.


Assuntos
Schistosoma mansoni/imunologia , Vacinas/imunologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Fezes/parasitologia , Feminino , Imunidade Celular , Imunidade Humoral , Camundongos Endogâmicos C57BL , Óvulo/fisiologia , Parasitos/imunologia , Perfusão , Primatas
12.
Artigo em Inglês | MEDLINE | ID: mdl-32154190

RESUMO

For decades, mass drug treatment with praziquantel (PZQ) has been utilized to treat schistosomiasis, yet reinfection and the risk of drug resistance are among the various factors precluding successful elimination of schistosomiasis. Tractable models that replicate "real world" field conditions are crucial to effectively evaluate putative schistosomiasis vaccines. Herein, we describe the cellular immune responses and cytokine expression profiles under field conditions that include prior infection with schistosomes followed by treatment with PZQ. Baboons were exposed to Schistosoma mansoni cercariae through trickle infection over 5 weeks, allowed for chronic disease to develop, and then treated with PZQ. Peripheral blood mononuclear cells (PBMCs) were monitored for cellular immune response(s) at each disease stage and PZQ therapy. After initial infection and during chronic disease, there was an increase in non-classical monocytes, NK and NKT cells while the CD4:CD8 T cell ratio inverted from a 2:1 to 1:2.5. The cytokine expressions of PBMCs after trickle infections were polarized more toward a Th2 response with a gradual increase in Th1 cytokine expression at chronic disease stage. Following PZQ treatment, with the exception of an increase in B cells, immune cell populations reverted back toward naïve levels; however, expression of almost all Th1, Th2, and Th17 cytokines was significantly increased. This preliminary study is the first to follow the cellular immune response and cytokine expression profiles in a non-human primate model simulating field conditions of schistosomiasis and PZQ therapy, providing a promising reference in predicting the immune response to future vaccines for schistosomiasis.


Assuntos
Anti-Helmínticos , Esquistossomose , Animais , Anti-Helmínticos/uso terapêutico , Leucócitos Mononucleares , Praziquantel/uso terapêutico , Primatas , Esquistossomose/tratamento farmacológico , Esquistossomose/veterinária
13.
Front Immunol ; 11: 578715, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33732227

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos de Helmintos/farmacologia , Desenvolvimento de Medicamentos , Vacinas Protozoárias/farmacologia , Schistosoma japonicum/patogenicidade , Esquistossomose Japônica/prevenção & controle , Drogas Veterinárias/farmacologia , Adjuvantes Imunológicos/economia , Animais , Anticorpos Anti-Helmínticos/sangue , Antígenos de Helmintos/economia , Antígenos de Helmintos/imunologia , Bovinos , Análise Custo-Benefício , Modelos Animais de Doenças , Custos de Medicamentos , Feminino , Interações Hospedeiro-Patógeno , Imunogenicidade da Vacina , Camundongos Endogâmicos C57BL , Contagem de Ovos de Parasitas , Projetos Piloto , Vacinas Protozoárias/economia , Schistosoma japonicum/imunologia , Esquistossomose Japônica/parasitologia , Esquistossomose Japônica/transmissão , Vacinação , Drogas Veterinárias/economia
14.
Parasit Vectors ; 13(1): 109, 2020 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-32111243

RESUMO

BACKGROUND: Schistosomiasis continues to inflict significant morbidity and mortality in the tropical and subtropical regions of the world. The disease endemicity overlaps with the transmission of other parasitic diseases. Despite the ubiquity of polyparasitism in tropical regions, particularly in rural communities, little is known about the impact of multiple helminth infections on disease progression. In this pilot study, we describe the influence of chronic Trichuris trichiura infection on Schistosoma mansoni egg-induced hepatopathology in infected baboons. METHODS: Baboons with or without underlying whipworm infection were challenged with S. mansoni cercariae to establish schistosomiasis. Adult S. mansoni worms were recovered by perfusion and enumerated, hepatic granulomas were quantified via light microscopy, and transcriptional profiling of tissues were completed using RNA sequencing technologies. RESULTS: Co-infection with both S. mansoni and T. trichiura resulted in higher female schistosome worm burden and significantly larger liver granuloma sizes. Systems biology analyses of peripheral blood mononuclear cells (PBMC) revealed pathways associated with increased liver damage in co-infected baboons. CONCLUSIONS: Underlying chronic whipworm infection intensified schistosome egg-induced liver pathology in infected baboons. RNA-Seq analysis provided insight into pathways associated with increased liver damage, corroborating histological findings.


Assuntos
Coinfecção/patologia , Coinfecção/veterinária , Hepatopatias Parasitárias/patologia , Hepatopatias Parasitárias/veterinária , Esquistossomose/patologia , Esquistossomose/veterinária , Tricuríase/patologia , Tricuríase/veterinária , Doenças dos Animais/parasitologia , Doenças dos Animais/patologia , Animais , Doença Crônica , Coinfecção/parasitologia , Feminino , Granuloma/patologia , Humanos , Fígado/metabolismo , Fígado/parasitologia , Fígado/patologia , Hepatopatias Parasitárias/parasitologia , Masculino , Papio , Contagem de Ovos de Parasitas , Projetos Piloto , Primatas , Schistosoma mansoni , Esquistossomose/parasitologia , Transcriptoma , Tricuríase/parasitologia , Trichuris
15.
Front Immunol ; 11: 1246, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32636844

RESUMO

Recent advances in systems biology have shifted vaccine development from a largely trial-and-error approach to an approach that promote rational design through the search for immune signatures and predictive correlates of protection. These advances will doubtlessly accelerate the development of a vaccine for schistosomiasis, a neglected tropical disease that currently affects over 250 million people. For over 15 years and with contributions of over 120 people, we have endeavored to test and optimize Sm-p80-based vaccines in the non-human primate model of schistosomiasis. Using RNA-sequencing on eight different Sm-p80-based vaccine strategies, we sought to elucidate immune signatures correlated with experimental protective efficacy. Furthermore, we aimed to explore the role of antibodies through in vivo passive transfer of IgG obtained from immunized baboons and in vitro killing of schistosomula using Sm-p80-specific antibodies. We report that passive transfer of IgG from Sm-p80-immunized baboons led to significant worm burden reduction, egg reduction in liver, and reduced egg hatching percentages from tissues in mice compared to controls. In addition, we observed that sera from Sm-p80-immunized baboons were able to kill a significant percent of schistosomula and that this effect was complement-dependent. While we did not find a universal signature of immunity, the large datasets generated by this study will serve as a substantial resource for further efforts to develop vaccine or therapeutics for schistosomiasis.


Assuntos
Anticorpos Anti-Helmínticos/farmacologia , Antígenos de Helmintos/imunologia , Helmintíase Animal/prevenção & controle , Imunização Passiva , Vacinas/imunologia , Animais , Anticorpos Anti-Helmínticos/imunologia , Modelos Animais de Doenças , Helmintíase Animal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Papio , Schistosoma mansoni , Esquistossomose mansoni
16.
Parasitol Res ; 105(6): 1767-77, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19809833

RESUMO

Advent of an effective schistosome vaccine would contribute significantly toward reducing the disease spectrum and transmission of schistosomiasis. We have targeted a functionally important antigen, Sm-p80, as a vaccine candidate because of its consistent immunogenicity, protective and antifecundity potentials, and important role in the immune evasion process. In this study, we report that using two vaccination approaches (prime boost and recombinant protein), Sm-p80-based vaccine formulation(s) confer up to 70% reduction in worm burden in mice. Animals immunized with the vaccine exhibited a decrease in egg production by up to 75%. The vaccine elicited strong immune responses that included IgM, IgA, and IgG (IgG1, IgG2a, IgG2b, and IgG3) in vaccinated animals. Splenocytes proliferated in response to Sm-p80 produced Th1 and Th17 response enhancing cytokines. These results again emphasize the potential of Sm-p80 as a viable vaccine candidate for schistosomiasis.


Assuntos
Antígenos de Helmintos/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/prevenção & controle , Vacinas/imunologia , Animais , Anticorpos Anti-Helmínticos/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Imunização Secundária/métodos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contagem de Ovos de Parasitas , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Baço/imunologia , Vacinas/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
17.
Pathology ; 50(4): 442-449, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29739616

RESUMO

Schistosomiasis remains a serious chronic debilitating hepato-intestinal disease. Current control measures based on mass drug administration are inadequate due to sustained re-infection rates, low treatment coverage and emergence of drug resistance. Hence, there is an urgent need for a schistosomiasis vaccine for disease control. In this study, we assessed the anti-pathology efficacy of Schistosoma mansoni large subunit of calpain (Sm-p80)-based vaccine against schistosomiasis caused by infections with Schistosoma mansoni in baboons. We also evaluated the disease transmission-blocking potential of Sm-p80 vaccine. Immunisations with Sm-p80-based vaccine resulted in significant reduction of hepatic egg load in vaccinated baboons (67.7% reduction, p = 0.0032) when compared to the control animals, indicative of reduction in pathology. There was also a significant reduction in sizes of egg-induced granulomas in baboons immunised with Sm-p80 vaccine compared to their control counterparts. Egg hatching rate analysis revealed an overall 85.6% reduction (p = 0.0018) in vaccinated animals compared to the controls, highlighting the potential role of Sm-p80 vaccine in disease transmission. The findings on anti-pathology efficacy and transmission-blocking potential presented in this study have formed the basis for a large-scale double-blinded baboon experiment that is currently underway.


Assuntos
Fígado/imunologia , Fígado/patologia , Esquistossomose/imunologia , Vacinas/imunologia , Animais , Humanos , Imunoglobulina G/imunologia , Papio , Schistosoma mansoni/imunologia , Vacinação
18.
J Investig Med ; 66(8): 1124-1132, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29997146

RESUMO

Sm-p80, the large subunit of Schistosoma mansoni calpain, is a leading candidate for a schistosomiasis vaccine. The prophylactic and antifecundity efficacy of Sm-p80 has been tested in three animal models (mouse, hamster and baboon) using a multitude of vaccine formulations and approaches. In our continual effort to enhance the vaccine efficacy, in this study, we have utilized the adjuvant, synthetic hexa-acylated lipid A derivative, glucopyranosyl lipid A (GLA) formulated in aluminum (GLA-Alum) with recombinant Sm-p80. The rSm-p80+GLA-Alum immunization regimen provided 33.33%-53.13% reduction in worm burden in the mouse model and 38% worm burden reduction in vaccinated baboons. Robust Sm-p80-specific immunoglobulin (Ig)G, IgG1, IgG2a and IgM responses were observed in all immunized animals. The rSm-p80+GLA-Alum coadministration induced a mix of T-helper (Th) cells (Th1, Th2 and Th17) responses as determined via the release of interleukin (IL)-2, IL-4, IL-18, IL-21, IL-22 and interferon-γ.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alúmen/farmacologia , Antígenos de Helmintos/imunologia , Glucosídeos/imunologia , Lipídeo A/imunologia , Schistosoma mansoni/imunologia , Receptor 4 Toll-Like/agonistas , Vacinas/imunologia , Animais , Proliferação de Células , Citocinas/biossíntese , Citocinas/genética , Feminino , Imunidade Humoral , Camundongos Endogâmicos C57BL , Papio , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia , Linfócitos T Auxiliares-Indutores/imunologia , Vacinação
19.
Ann N Y Acad Sci ; 1425(1): 38-51, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30133707

RESUMO

Schistosomiasis is of public health importance to an estimated one billion people in 79 countries. A vaccine is urgently needed. Here, we report the results of four independent, double-blind studies of an Sm-p80-based vaccine in baboons. The vaccine exhibited potent prophylactic efficacy against transmission of Schistosoma mansoni infection and was associated with significantly less egg-induced pathology, compared with unvaccinated control animals. Specifically, the vaccine resulted in a 93.45% reduction of pathology-producing female worms and significantly resolved the major clinical manifestations of hepatic/intestinal schistosomiasis by reducing the tissue egg-load by 89.95%. A 35-fold decrease in fecal egg excretion in vaccinated animals, combined with an 81.51% reduction in hatching of eggs into the snail-infective stage (miracidia), demonstrates the parasite transmission-blocking potential of the vaccine. Substantially higher Sm-p80 expression in female worms and Sm-p80-specific antibodies in vaccinated baboons appear to play an important role in vaccine-mediated protection. Preliminary analyses of RNA sequencing revealed distinct molecular signatures of vaccine-induced effects in baboon immune effector cells. This study provides comprehensive evidence for the effectiveness of an Sm-p80-based vaccine for schistosomiasis.


Assuntos
Vacinas Protozoárias , Esquistossomose , Animais , Feminino , Masculino , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/imunologia , Método Duplo-Cego , Perfilação da Expressão Gênica , Papio , Contagem de Ovos de Parasitas , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/genética , Vacinas Protozoárias/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Schistosoma mansoni/imunologia , Esquistossomose/prevenção & controle , Esquistossomose/transmissão , Esquistossomose/veterinária , Transcrição Gênica
20.
Ann N Y Acad Sci ; 1425(1): 19-37, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29888790

RESUMO

Sm-p80-based vaccine efficacy for Schistosoma mansoni was evaluated in a baboon model of infection and disease. The study was designed to replicate a human vaccine implementation scenario for endemic regions in which vaccine would be administered following drug treatment of infected individuals. In our study, the Sm-p80-based vaccine reduced principal pathology producing hepatic egg burdens by 38.0% and egg load in small and large intestines by 72.2% and 49.4%, respectively, in baboons. Notably, hatching rates of eggs recovered from liver and small and large intestine of vaccinated animals were significantly reduced, by 60.4%, 48.6%, and 82.3%, respectively. Observed reduction in egg maturation/hatching rates was supported by immunofluorescence and confocal microscopy showing unique differences in Sm-p80 expression in worms of both sexes and matured eggs. Vaccinated baboons had a 64.5% reduction in urine schistosome circulating anodic antigen, a parameter that reflects worm numbers/health status in infected hosts. Preliminary analyses of RNA sequencing revealed unique genes and canonical pathways associated with establishment of chronic disease, praziquantel-mediated parasite killing, and Sm-p80-mediated protection in vaccinated baboons. Overall, our study demonstrated efficacy of the Sm-p80 vaccine and provides insight into some of the epistatic interactions associated with protection.


Assuntos
Praziquantel/uso terapêutico , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Vacinação/métodos , Vacinas/imunologia , Animais , Anti-Helmínticos/uso terapêutico , Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/imunologia , Doença Crônica , Feminino , Humanos , Masculino , Contagem de Ovos de Parasitas , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/fisiologia , Esquistossomose mansoni/parasitologia , Esquistossomose mansoni/terapia , Resultado do Tratamento , Vacinas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA