Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Malar J ; 23(1): 35, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38281044

ABSTRACT

BACKGROUND: Sri Lanka after eliminating malaria in 2012, is in the prevention of re-establishment (POR) phase. Being a tropical country with high malariogenic potential, maintaining vigilance is important. All malaria cases are investigated epidemiologically and followed up by integrated drug efficacy surveillance (iDES). Occasionally, that alone is not adequate to differentiate Plasmodium falciparum reinfections from recrudescences. This study evaluated the World Health Organization and Medicines for Malaria Venture (MMV) recommended genotyping protocol for the merozoite surface proteins (msp1, msp2) and the glutamate-rich protein (glurp) to discriminate P. falciparum recrudescence from reinfection in POR phase. METHODS: All P. falciparum patients detected from April 2014 to December 2019 were included in this study. Patients were treated and followed up by iDES up to 28 days and were advised to get tested if they develop fever at any time over the following year. Basic socio-demographic information including history of travel was obtained. Details of the malariogenic potential and reactive entomological and parasitological surveillance carried out by the Anti Malaria Campaign to exclude the possibility of local transmission were also collected. The msp1, msp2, and glurp genotyping was performed for initial and any recurrent infections. Classification of recurrent infections as recrudescence or reinfection was done based on epidemiological findings and was compared with the genotyping outcome. RESULTS: Among 106 P. falciparum patients, six had recurrent infections. All the initial infections were imported, with a history of travel to malaria endemic countries. In all instances, the reactive entomological and parasitological surveillance had no evidence for local transmission. Five recurrences occurred within 28 days of follow-up and were classified as recrudescence. They have not travelled to malaria endemic countries between the initial and recurrent infections. The other had a recurrent infection after 105 days. It was assumed a reinfection, as he had travelled to the same malaria endemic country in between the two malaria attacks. Genotyping confirmed the recrudescence and the reinfection. CONCLUSIONS: The msp1, msp2 and glurp genotyping method accurately differentiated reinfections from recrudescence. Since reinfection without a history of travel to a malaria endemic country would mean local transmission, combining genotyping outcome with epidemiological findings will assist classifying malaria cases without any ambiguity.


Subject(s)
Frontotemporal Dementia , Malaria, Falciparum , Merozoite Surface Protein 1 , Muscular Dystrophies, Limb-Girdle , Myositis, Inclusion Body , Osteitis Deformans , Male , Humans , Merozoite Surface Protein 1/genetics , Plasmodium falciparum/genetics , Reinfection , Protozoan Proteins/genetics , Protozoan Proteins/therapeutic use , Antigens, Protozoan/genetics , Antigens, Protozoan/therapeutic use , Genotype , Glutamic Acid , Sri Lanka/epidemiology , Genetic Variation , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/drug therapy , Recurrence
2.
Front Immunol ; 12: 634508, 2021.
Article in English | MEDLINE | ID: mdl-33717176

ABSTRACT

In areas where Plasmodium falciparum transmission is endemic, clinical immunity against malaria is progressively acquired during childhood and adults are usually protected against the severe clinical consequences of the disease. Nevertheless, pregnant women, notably during their first pregnancies, are susceptible to placental malaria and the associated serious clinical outcomes. Placental malaria is characterized by the massive accumulation of P. falciparum infected erythrocytes and monocytes in the placental intervillous spaces leading to maternal anaemia, hypertension, stillbirth and low birth weight due to premature delivery, and foetal growth retardation. Remarkably, the prevalence of placental malaria sharply decreases with successive pregnancies. This protection is associated with the development of antibodies directed towards the surface of P. falciparum-infected erythrocytes from placental origin. Placental sequestration is mediated by the interaction between VAR2CSA, a member of the P. falciparum erythrocyte membrane protein 1 family expressed on the infected erythrocytes surface, and the placental receptor chondroitin sulfate A. VAR2CSA stands today as the leading candidate for a placental malaria vaccine. We recently reported the safety and immunogenicity of two VAR2CSA-derived placental malaria vaccines (PRIMVAC and PAMVAC), spanning the chondroitin sulfate A-binding region of VAR2CSA, in both malaria-naïve and P. falciparum-exposed non-pregnant women in two distinct Phase I clinical trials (ClinicalTrials.gov, NCT02658253 and NCT02647489). This review discusses recent advances in placental malaria vaccine development, with a focus on the recent clinical data, and discusses the next clinical steps to undertake in order to better comprehend vaccine-induced immunity and accelerate vaccine development.


Subject(s)
Antigens, Protozoan/therapeutic use , Drug Development , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Placenta/parasitology , Pregnancy Complications, Parasitic/prevention & control , Animals , Antigens, Protozoan/adverse effects , Antigens, Protozoan/immunology , Female , Host-Parasite Interactions , Humans , Immunization , Immunogenicity, Vaccine , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Placenta/immunology , Pregnancy , Pregnancy Complications, Parasitic/immunology , Pregnancy Complications, Parasitic/parasitology , Treatment Outcome
3.
Rev. bras. ciênc. vet ; 28(1): 37-41, jan./mar. 2021.
Article in Portuguese | LILACS, VETINDEX | ID: biblio-1368572

ABSTRACT

O presente trabalho tem como objetivo relatar um caso de um Pastor Belga, do município de Ponta Porã, Mato Grosso do Sul, positivo para Leishmaniose Visceral, atendido em 2017 em uma clínica veterinária localizada em Pedro Juan Caballero, Paraguai. O diagnóstico foi confirmado através dos sinais clínicos característicos, e dos exames ELISA e PCR positivos. O animal foi submetido ao tratamento clínico para melhora dos sintomas, cujo tratamento antiparasitário inicial foi realizado com a associação de estibogluconato de sódio 75 mg/kg e alopurinol 100 mg seguido de aloputinol 100mg de uso contínuo e uso da coleira antileishmaniose. Tratamento esse considerado eficiente, com melhora clínica do animal. Após 24 meses o animal foi diagnosticado com tumor de mama e lesão da bolsa escrotal, sendo submetido a tratamento clínico e cirúrgico. Com 30 e 36 meses do diagnóstico inicial repetiu-se os exames ELISA (positivo) e PCR (negativo), e então o animal foi considerado curado clinicamente devido à ausência de sinais clínicos. Tendo em vista a complexidade dos fatores no ciclo de transmissão, conclui-se que as medidas em saúde ainda são insuficientes para o controle efetivo da doença. É importante o papel do Médico Veterinário na saúde pública, devido a obrigatoriedade de notificação de casos de Leishmaniose Visceral Canina, sendo necessários esforços nas diferentes áreas da saúde animal, humana e do meio ambiente, visando medidas de vigilância e controle da doença no país.


The present work aims to report a case of a Belgian Shepherd, from the municipality of Ponta Porã, Mato Grosso do Sul, positive for Visceral Leishmaniasis, treated in 2017 in a veterinary clinic located in Pedro Juan Caballero, Paraguay. The diagnosis was confirmed through the characteristic clinical signs, and the positive ELISA and PCR tests. The animal was submitted to clinical treatment for improvement of symptoms, whose initial antiparasitic treatment was performed with the association of sodium stibogluconate 75 mg/kg and allopurinol 100 mg followed by alloputinol 100mg of continuous use and use of the antileishmaniasis collar. This treatment was considered efficient, with clinical improvement of the animal. After 24 months the animal was diagnosed with a breast tumor and scrotum injury, and was submitted to clinical and surgical treatment. At 30 and 36 months from the initial diagnosis, the ELISA tests (positive) and PCR (negative) were repeated, and then the animal was considered clinically cured due to the absence of clinical signs. Considering the complexity of the factors in the transmission cycle, it is concluded that the health measures are still insufficient for the effective control of the disease. The role of the veterinarian in public health is important, due to the obligatory notification of cases of Canine Visceral Leishmaniasis, being necessary efforts in the different areas of animal health, human and environment, aiming at measures of surveillance and control of the disease in the country.


Subject(s)
Animals , Dogs , Dogs/parasitology , Leishmaniasis, Visceral/veterinary , Zoonoses/parasitology , Continuity of Patient Care , Veterinary Public Health , Antigens, Protozoan/therapeutic use
4.
Malar J ; 18(1): 394, 2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31796032

ABSTRACT

BACKGROUND: Despite the extensive endeavours, developing an effective malaria vaccine remains as a great challenge. Apical membrane antigen 1 (AMA-1) located on the merozoite surface of parasites belonging to the genus Plasmodium is involved in red blood cell invasion. METHODS: Influenza virus-like particle (VLP) vaccines containing codon-optimized or native (non-codon optimized) AMA-1 from Plasmodium berghei were generated. VLP-induced protective immunity was evaluated in a mouse model. RESULTS: Mice immunized with VLP vaccine containing the codon-optimized AMA-1 elicited higher levels of P. berghei-specific IgG and IgG2a antibody responses compared to VLPs containing non-codon optimized AMA-1 before and after challenge infection. Codon-optimized AMA-1 VLP vaccination induced higher levels of CD4+ T cells, CD8+ T cells, B cells, and germinal centre cell responses compared to non-codon optimized AMA-1 VLPs. Importantly, the codon-optimized AMA-1 VLP vaccination showed lower body weight loss, longer survival and a significant decrease in parasitaemia compared to non-codon optimized VLP vaccination. CONCLUSION: Overall, VLP vaccine expressing codon-optimized AMA-1 induced better protective efficacy than VLPs expressing the non-codon optimized AMA-1. Current findings highlight the importance of codon-optimization for vaccine use and its potential involvement in future malaria vaccine design strategies.


Subject(s)
Antigens, Protozoan/therapeutic use , Malaria Vaccines/pharmacology , Malaria/prevention & control , Membrane Proteins/therapeutic use , Plasmodium berghei/immunology , Protozoan Proteins/therapeutic use , Vaccines, Virus-Like Particle/pharmacology , Animals , Codon/immunology , Female , Mice , Mice, Inbred BALB C
5.
Front Immunol ; 10: 2480, 2019.
Article in English | MEDLINE | ID: mdl-31695697

ABSTRACT

Background: Malaria elimination remains a priority research agenda with the need for interventions that reduce and/or block malaria transmission from humans to mosquitoes. Transmission-blocking vaccines (TBVs) are in development, most of which target the transmission stage (i.e., gametocyte) antigens Pfs230 and Pfs48/45. For these interventions to be implemented, there is a need to understand the naturally acquired immunity to gametocytes. Several studies have measured the prevalence of immune responses to Pfs230 and Pfs48/45 in populations in malaria-endemic areas. Methods: We conducted a systematic review of studies carried out in African populations that measured the prevalence of immune responses to the gametocyte antigens Pfs230 and Pfs48/45. We assessed seroprevalence of antibody responses to the two antigens and investigated the effects of covariates such as age, transmission intensity/endemicity, season, and parasite prevalence on the prevalence of these antibody responses by meta-regression. Results: We identified 12 studies covering 23 sites for inclusion in the analysis. We found that the range of reported seroprevalence to Pfs230 and Pfs48/45 varied widely across studies, from 0 to 64% for Pfs48/45 and from 6 to 72% for Pfs230. We also found a modest association between increased age and increased seroprevalence to Pfs230: adults were associated with higher seroprevalence estimates in comparison to children (ß coefficient 0.21, 95% CI: 0.05-0.38, p = 0.042). Methodological factors were the most significant contributors to heterogeneity between studies which prevented calculation of pooled prevalence estimates. Conclusions: Naturally acquired sexual stage immunity, as detected by antibodies to Pfs230 and Pfs48/45, was present in most studies analyzed. Significant between-study heterogeneity was seen, and methodological factors were a major contributor to this, and prevented further analysis of epidemiological and biological factors. This demonstrates a need for standardized protocols for conducting and reporting seroepidemiological analyses.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Life Cycle Stages/immunology , Malaria Vaccines/immunology , Malaria, Falciparum , Plasmodium falciparum/immunology , Africa , Antigens, Protozoan/therapeutic use , Female , Humans , Malaria Vaccines/therapeutic use , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Male
6.
Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi ; 31(4): 400-403, 2019 Sep 23.
Article in Chinese | MEDLINE | ID: mdl-31612675

ABSTRACT

OBJECTIVE: To investigate the effect of Toxoplasma gondii excretory-secretory antigens (ESA) on CD4+ CD25+ Foxp3+ T (Treg) cells in mice carrying Lewis lung carcinoma, and examine the inhibitory effect of T. gondii ESA on tumor growth. METHODS: C57BL/6 mice were randomly assigned into the PBS group (n = 14) and the Lewis group (n = 34). Mice in the Lewis group were subcutaneously injected with 2 × 105 Lewis lung carcinoma cells in the right axilla, while animals in the PBS group were injected with the same volume of sterile PBS. On day 7 post-injection (D7), mice in the PBS group were further divided into the PBS2 group and the PBS2 + ESA group, of 7 mice in each group, and mice in the Lewis group were further divided into the Lewis2 group and the Lewis2 + ESA group, of 17 mice in each group. Then, mice in the PBS2 + ESA group and the Lewis2 + ESA group were intraperitoneally injected with 100 µL of ESA. The mouse spleen coefficient was calculated in each group 7 days post-injection with ESA, and the changes of Treg cell counts and the long-term tumor growth were measured in tumor-bearing mice. RESULTS: The spleen coefficient was significantly greater in the PBS2 + ESA group and the Lewis2 + ESA group than in the PBS2 (0.66% ± 0.09% vs. 0.30% ± 0.02%, P < 0.05) and Lewis2 groups (0.69% ± 0.07% vs. 0.33% ± 0.03%, P < 0.05) 7 days post-treatment with ESA, respectively, and the percentage of splenic Treg cells in splenocytes was significantly lower in the PBS2 + ESA group and the Lewis2 + ESA group than in the PBS2 (1.28% ± 0.14% vs. 2.06% ± 0.07%, P < 0.05) and Lewis2 groups (1.58% ± 0.14% vs. 2.44% ± 0.23%, P < 0.05), respectively. T. gondii ESA treatment caused a delay in tumor growth, and the tumor size was significantly smaller in the Lewis2 + ESA group than in the Lewis2 group (P < 0.05). CONCLUSIONS: T. gondii ESA may reduce the proportion of splenic Treg cells in splenocytes and inhibit tumor growth in mice carrying Lewis lung carcinoma.


Subject(s)
Antigens, Protozoan , Carcinoma, Lewis Lung , Toxoplasma , Animals , Antigens, Protozoan/pharmacology , Antigens, Protozoan/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Cell Count , Cell Proliferation/drug effects , Mice , Mice, Inbred C57BL , Random Allocation , Spleen/drug effects , T-Lymphocytes, Regulatory/cytology , Toxoplasma/chemistry , Treatment Outcome
7.
Infect Genet Evol ; 70: 27-35, 2019 06.
Article in English | MEDLINE | ID: mdl-30738195

ABSTRACT

There is a need for new, effective, and less expensive and toxic treatment for Leishmaniasis. It seems that the use of a suitable adjuvant and a delivery system is effective in inducing immune reactions for protection. Liposomes can be applied as immunoadjuvants to trigger immune reactions to different antigens. The adjuvant effects of imiquimod using DSPC liposomes containing SLA (soluble Leishmania antigens) were studied on the type and intensity of the produced immune reaction to the challenge of Leishmania major in BALB/c mice. Liposomes were produced by the lipid film procedure. BALB/C mice were immunized subcutaneously, three times at 2-week intervals and with various formulations. Lesion development and the parasite burden in the spleens and feet after the challenge with Leishmania major, Th1 cytokine (IFN-γ), and the IgG isotype titration were assessed to evaluate the induced immune reaction and the protection level. The group of mice immunized with Liposome DSPC +Imiquimod +SLA revealed less severe footpad swelling, being significantly different (P < .05) from other groups. A higher level of IgG2a and IFN-γ secretion was observed in the mice immunized with Liposome DSPC +Imiquimod +SLA than the control group. These observations imply that the DSPC liposome containing imiquimod induces the Th1 immune response that is protective against the challenge of Leishmania major.


Subject(s)
Antigens, Protozoan/therapeutic use , Leishmania major/immunology , Leishmaniasis, Cutaneous/therapy , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Protozoan/immunology , Female , Imiquimod/therapeutic use , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/pathology , Liposomes/immunology , Liposomes/therapeutic use , Mice , Mice, Inbred BALB C
8.
Article in English | MEDLINE | ID: mdl-31921703

ABSTRACT

Visceral leishmaniasis (VL), caused by digenetic protozoa of the genus Leishmania, is the most severe form of leishmaniasis. Leishmania infantum is one of the species responsible for VL and the disease caused is considered a zoonosis whose main reservoir is the dog. Canine visceral leishmaniasis (CVL) can lead to the death of the animal if left untreated. Furthermore, the available pharmocologial treatment for CVL presents numerous disadvantages, such as relapses, toxicity, drug resistance, and the fact treated animals continue to be reservoirs when treatment fails to achieve parasitological cure. Moreover, the available VL control methods have not been adequate when it comes to controlling parasite transmission. Advances in immune response knowledge in recent years have led to a better understanding of VL pathogenesis, allowing new treatments to be developed based on immune system activation, often referred to as immunotherapy. In fact, well-defined protocols have been described, ranging from the use of immunomodulators to the use of vaccines. This treatment, which can also be associated with chemotherapy, has been shown to be effective in restoring or inducing an adequate immune response to reduce parasitic burden, leading to clinical improvement. This review focuses on immunotherapy directed at dogs infected by L. infantum, including a literature review of what has already been done in dogs. We also introduce a promising strategy to improve the efficacy of immunotherapy.


Subject(s)
Antigens, Protozoan/therapeutic use , Dog Diseases/therapy , Immunotherapy/methods , Leishmaniasis, Visceral/therapy , Leishmaniasis, Visceral/veterinary , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/immunology , Biomarkers , Dog Diseases/immunology , Dog Diseases/parasitology , Dogs , Humans , Immunologic Factors/therapeutic use , Leishmania infantum/immunology , Leishmaniasis, Visceral/immunology , Protozoan Vaccines/therapeutic use , Treatment Outcome
9.
Parasitol Int ; 68(1): 63-72, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30339837

ABSTRACT

Clioquinol (5-chloro-7-iodoquinolin-8-ol or ICHQ) was recently showed to presents an in vitro effective antileishmanial action, causing changes in membrane permeability, mitochondrial functionality, and parasite morphology. In the present study, ICHQ was incorporated into a Poloxamer 407-based polymeric micelles system (ICHQ/M), and its antileishmanial activity was in vivo evaluated in L. amazonensis-infected BALB/c mice. Amphotericin B (AmpB) and its liposomal formulation (Ambisome®) were used as controls. Parasitological and immunological evaluations were performed 30 days after the treatment. Results indicated more significant reductions in the average lesion diameter and parasite burden in ICHQ or ICHQ/M-treated mice, which were associated with the development of a polarized Th1 immune response, based on production of high levels of IFN-γ, IL-12, TNF-α, GM-CSF, and antileishmanial IgG2a antibody. Control groups´ mice produced high levels of IL-4, IL-10, and IgG1 isotype antibody. No organic toxicity was found by using ICHQ or ICHQ/M to treat the animals, although those receiving AmpB and Ambisome® have presented higher levels of renal and hepatic damage markers. In conclusion, results suggested that the ICHQ/M composition can be considered as an antileishmanial candidate to be tested against human leishmaniasis.


Subject(s)
Antiprotozoal Agents/immunology , Antiprotozoal Agents/therapeutic use , Clioquinol/immunology , Clioquinol/therapeutic use , Leishmania mexicana/drug effects , Leishmaniasis, Visceral/drug therapy , Poloxamer/administration & dosage , Amphotericin B/administration & dosage , Amphotericin B/therapeutic use , Amphotericin B/toxicity , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/immunology , Antigens, Protozoan/therapeutic use , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/toxicity , Clioquinol/administration & dosage , Cytokines/biosynthesis , Cytokines/immunology , Drug Delivery Systems/methods , Humans , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Leishmania mexicana/growth & development , Leishmaniasis, Visceral/immunology , Mice , Mice, Inbred BALB C , Micelles , Parasite Load , Poloxamer/chemistry , Th1 Cells
10.
Cell Mol Biol (Noisy-le-grand) ; 64(12): 1-5, 2018 Sep 30.
Article in English | MEDLINE | ID: mdl-30301493

ABSTRACT

Cancer is the main cause of death in the developed countries. There are some scientific evidences indicating that parasitic infections induce antitumor activity against certain types of cancers. Hydatid cyst is the larval stage of Echinococcus granulosus, which causes hydatidosis in human and livestock. We have already shown that vaccination of mice with hydatid cyst crude antigens and subsequently challenge them with cancer cells, causes inhibition of melanoma cancer growth. In this study, therapeutic effects of hydatid cyst antigens on C57/black mice that had already been challenged with melanoma tumor were investigated. In this experimental study, 6 groups of C57 black mice were subcutaneously inoculated with melanoma cancer cells (line B16F10) in PBS inside their chest site. After 2 weeks case groups were injected with hydatid cyst fluid, a fraction of cyst fluid, live protoscolices or BCG.  control groups were injected with alum alone and other control group was left intact without any intervention. The size of each tumor was measured in all mice. Blood samples were also taken to estimate Interleukin-2 (IL-2), Tumor necrosis factor alpha (TNF-α), Interferon gamma (IFN-γ) and Interleukin-4 (IL-4) levels. Treatment of mice bearing melanoma cancer with hydatid cyst antigens resulted in inhibition of tumor growth and the difference between mean size of tumor in case and control groups was statistically significant. Also, according to our results mean level of measured cytokines between case and control groups was statistically different. Hydatid cyst antigens have anti-melanoma activities and this effect may be related to immune response to parasite antigens.


Subject(s)
Antigens, Protozoan/therapeutic use , Echinococcosis/immunology , Echinococcus granulosus/immunology , Melanoma/blood , Melanoma/drug therapy , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Interferon-gamma/blood , Interleukin-2/blood , Interleukin-4/blood , Melanoma/immunology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/blood
11.
Drug Des Devel Ther ; 12: 25-40, 2018.
Article in English | MEDLINE | ID: mdl-29317800

ABSTRACT

Leishmania is an obligate intracellular pathogen that invades phagocytic host cells. Approximately 30 different species of Phlebotomine sand flies can transmit this parasite either anthroponotically or zoonotically through their bites. Leishmaniasis affects poor people living around the Mediterranean Basin, East Africa, the Americas, and Southeast Asia. Affected regions are often remote and unstable, with limited resources for treating this disease. Leishmaniasis has been reported as one of the most dangerous neglected tropical diseases, second only to malaria in parasitic causes of death. People can carry some species of Leishmania for long periods without becoming ill, and symptoms depend on the form of the disease. There are many drugs and candidate vaccines available to treat leishmaniasis. For instance, antiparasitic drugs, such as amphotericin B (AmBisome), are a treatment of choice for leishmaniasis depending on the type of the disease. Despite the availability of different treatment approaches to treat leishmaniasis, therapeutic tools are not adequate to eradicate this infection. In the meantime, drug therapy has been limited because of adverse side effects and unsuccessful vaccine preparation. However, it can immediately make infections inactive. According to other studies, vaccination cannot eradicate leishmaniasis. There is no perfect vaccine or suitable drug to eradicate leishmaniasis completely. So far, no vaccine or drug has been provided to induce long-term protection and ensure effective immunity against leishmaniasis. Therefore, it is necessary that intensive research should be performed in drug and vaccine fields to achieve certain results.


Subject(s)
Antigens, Protozoan/therapeutic use , Leishmania/drug effects , Leishmaniasis Vaccines/therapeutic use , Leishmaniasis/drug therapy , Trypanocidal Agents/therapeutic use , Animals , Antigens, Protozoan/adverse effects , Antigens, Protozoan/immunology , Drug Resistance , Drug Therapy, Combination , Humans , Leishmania/immunology , Leishmaniasis/diagnosis , Leishmaniasis/immunology , Leishmaniasis/parasitology , Leishmaniasis Vaccines/adverse effects , Leishmaniasis Vaccines/immunology , Treatment Outcome , Trypanocidal Agents/adverse effects
12.
Proc Natl Acad Sci U S A ; 114(48): E10438-E10445, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29138320

ABSTRACT

Acquired resistance against antimalarial drugs has further increased the need for an effective malaria vaccine. The current leading candidate, RTS,S, is a recombinant circumsporozoite protein (CSP)-based vaccine against Plasmodium falciparum that contains 19 NANP repeats followed by a thrombospondin repeat domain. Although RTS,S has undergone extensive clinical testing and has progressed through phase III clinical trials, continued efforts are underway to enhance its efficacy and duration of protection. Here, we determined that two monoclonal antibodies (mAbs 311 and 317), isolated from a recent controlled human malaria infection trial exploring a delayed fractional dose, inhibit parasite development in vivo by at least 97%. Crystal structures of antibody fragments (Fabs) 311 and 317 with an (NPNA)3 peptide illustrate their different binding modes. Notwithstanding, one and three of the three NPNA repeats adopt similar well-defined type I ß-turns with Fab311 and Fab317, respectively. Furthermore, to explore antibody binding in the context of P. falciparum CSP, we used negative-stain electron microscopy on a recombinant shortened CSP (rsCSP) construct saturated with Fabs. Both complexes display a compact rsCSP with multiple Fabs bound, with the rsCSP-Fab311 complex forming a highly organized helical structure. Together, these structural insights may aid in the design of a next-generation malaria vaccine.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/therapy , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Antigens, Protozoan/isolation & purification , Antigens, Protozoan/therapeutic use , Clinical Trials, Phase II as Topic , Crystallography, X-Ray , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Humans , Malaria Vaccines/chemistry , Malaria Vaccines/therapeutic use , Malaria, Falciparum/immunology , Mice , Mice, Inbred C57BL , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/isolation & purification , Protozoan Proteins/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Repetitive Sequences, Amino Acid/immunology , Structure-Activity Relationship
13.
J Nanobiotechnology ; 15(1): 62, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28877692

ABSTRACT

BACKGROUND: The parasitic disease malaria remains a major global public health concern and no truly effective vaccine exists. One approach to the development of a malaria vaccine is to target the asexual blood stage that results in clinical symptoms. Most attempts have failed. New antigens such as P27A and P27 have emerged as potential new vaccine candidates. Multiple studies have demonstrated that antigens are more immunogenic and are better correlated with protection when presented on particulate delivery systems. One such particulate delivery system is the self-assembling protein nanoparticle (SAPN) that relies on coiled-coil domains of proteins to form stable nanoparticles. In the past we have used de novo designed amino acid domains to drive the formation of the coiled-coil scaffolds which present the antigenic epitopes on the particle surface. RESULTS: Here we use naturally occurring domains found in the tex1 protein to form the coiled-coil scaffolding of the nanoparticle. Thus, by engineering P27A and a new extended form of the coiled-coil domain P27 onto the N and C terminus of the SAPN protein monomer we have developed a particulate delivery system that effectively displays both antigens on a single particle that uses malaria tex1 sequences to form the nanoparticle scaffold. These particles are immunogenic in a murine model and induce immune responses similar to the ones observed in seropositive individuals in malaria endemic regions. CONCLUSIONS: We demonstrate that our P27/P27A-SAPNs induce an immune response akin to the one in seropositive individuals in Burkina Faso. Since P27 is highly conserved among different Plasmodium species, these novel SAPNs may even provide cross-protection between Plasmodium falciparum and Plasmodium vivax the two major human malaria pathogens. As the SAPNs are also easy to manufacture and store they can be delivered to the population in need without complication thus providing a low cost malaria vaccine.


Subject(s)
Antigens, Protozoan/therapeutic use , Malaria Vaccines/therapeutic use , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Nanoparticles/therapeutic use , Plasmodium falciparum/immunology , Proliferating Cell Nuclear Antigen/therapeutic use , Protozoan Proteins/therapeutic use , Amino Acid Sequence , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Humans , Immunization , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Models, Molecular , Nanoparticles/chemistry , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/immunology , Protein Domains , Protein Engineering , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology
14.
Biomed Res Int ; 2017: 1295038, 2017.
Article in English | MEDLINE | ID: mdl-28875149

ABSTRACT

Toxoplasma gondii can infect all warm-blooded animals including human beings. T. gondii dense granule protein 16 (TgGRA16) as a crucial virulence factor could modulate the host gene expression. Here, a DNA vaccine expressing TgGRA16 was constructed to explore the protective efficacy against T. gondii infection in Kunming mice. The immune responses induced by pVAX-GRA16 were also evaluated. Mice immunized with pVAX-GRA16 could elicit higher levels of specific IgG antibody and strong cellular response compared to those in controls. The DNA vaccination significantly increased the levels of cytokines (IFN-γ, IL-2, IL-4, and IL-10) and the percentages of CD4+ and CD8+ T cells in mice. After lethal challenge, mice immunized with pVAX-GRA16 (8.4 ± 0.78 days) did not show a significant longer survival time than that in controls (7.1 ± 0.30 days) (p > 0.05). However, in chronic toxoplasmosis model (administration of 10 brain cysts of PRU strain orally), numbers of tissue cysts in mice immunized with pVAX-GRA16 were significantly reduced compared to those in controls (p < 0.05) and the rate of reduction could reach 43.89%. The results indicated that the TgGRA16 would be a promising vaccine candidate for further development of effective epitope-based vaccines against chronic T. gondii infection in mice.


Subject(s)
Antigens, Protozoan/genetics , Drug Resistance/drug effects , Protozoan Proteins/genetics , Toxoplasmosis, Animal/prevention & control , Vaccines, DNA/administration & dosage , Animals , Antigens, Protozoan/immunology , Antigens, Protozoan/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Drug Resistance/genetics , Drug Resistance/immunology , Host-Parasite Interactions/genetics , Humans , Mice , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/adverse effects , Protozoan Vaccines/immunology , Toxoplasma/genetics , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/genetics , Toxoplasmosis, Animal/parasitology , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology
15.
Pharm Res ; 34(9): 1970-1983, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28646324

ABSTRACT

PURPOSE: Production and characterization of a chimeric fusion protein (GMZ2'.10C) which combines epitopes of key malaria parasite antigens: glutamate-rich protein (GLURP), merozoite surface protein 3 (MSP3), and the highly disulphide bonded Pfs48/45 (10C). GMZ2'.10C is a potential candidate for a multi-stage malaria vaccine that targets both transmission and asexual life-cycle stages of the parasite. METHODS: GMZ2'.10C was produced in Lactococcus lactis and purified using either an immunoaffinity purification (IP) or a conventional purification (CP) method. Protein purity and stability was analysed by RP-HPLC, SEC-HPLC, 2-site ELISA, gel-electrophoresis and Western blotting. Structural characterization (mass analysis, peptide mapping and cysteine connectivity mapping) was performed by LC-MS/MS. RESULTS: CP-GMZ2'.10C resulted in similar purity, yield, structure and stability as compared to IP-GMZ2'.10C. CP-GMZ2'.10C and IP-GMZ2'.10C both elicited a high titer of transmission blocking (TB) antibodies in rodents. The intricate disulphide-bond connectivity of C-terminus Pfs48/45 was analysed by tandem mass spectrometry and was established for GMZ2'.10C and two reference fusion proteins encompassing similar parts of Pfs48/45. CONCLUSION: GMZ2'.10C, combining GMZ2' and correctly-folded Pfs48/45 can be produced by the Lactoccus lactis P170 based expression system in purity and quality for pharmaceutical development and elicit high level of TB antibodies. The cysteine connectivity for the 10C region of Pfs48/45 was revealed experimentally, providing an important guideline for employing the Pfs48/45 antigen in vaccine design.


Subject(s)
Antigens, Protozoan/therapeutic use , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/therapeutic use , Amino Acid Sequence , Animals , Antibody Formation , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Cloning, Molecular , Humans , Lactococcus lactis/genetics , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protein Stability , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use
16.
Expert Rev Vaccines ; 16(7): 1-13, 2017 07.
Article in English | MEDLINE | ID: mdl-28525963

ABSTRACT

INTRODUCTION: Transgenic malaria parasites expressing foreign genes, for example fluorescent and luminescent proteins, are used extensively to interrogate parasite biology and host-parasite interactions associated with malaria pathology. Increasingly transgenic parasites are also exploited to advance malaria vaccine development. Areas covered: We review how transgenic malaria parasites are used, in vitro and in vivo, to determine protective efficacy of different antigens and vaccination strategies and to determine immunological correlates of protection. We describe how chimeric rodent parasites expressing P. falciparum or P. vivax antigens are being used to directly evaluate and rank order human malaria vaccines before their advancement to clinical testing. In addition, we describe how transgenic human and rodent parasites are used to develop and evaluate live (genetically) attenuated vaccines. Expert commentary: Transgenic rodent and human malaria parasites are being used to both identify vaccine candidate antigens and to evaluate both sub-unit and whole organism vaccines before they are advanced into clinical testing. Transgenic parasites combined with in vivo pre-clinical testing models (e.g. mice) are used to evaluate vaccine safety, potency and the durability of protection as well as to uncover critical protective immune responses and to refine vaccination strategies.


Subject(s)
Antigens, Protozoan/therapeutic use , Drug Discovery/methods , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Malaria, Vivax/prevention & control , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Protozoan Proteins/therapeutic use , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Gene Expression Regulation , Humans , Immunogenicity, Vaccine , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Malaria, Vivax/transmission , Plasmodium falciparum/genetics , Plasmodium vivax/genetics , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use
17.
BMC Immunol ; 17(1): 37, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27716047

ABSTRACT

BACKGROUND: Toxoplasma gondii is an opportunistic protozoan closely associated with AIDS and vertical transmission. T. gondii actin depolymerizing factor (TgADF) plays an important role in actin cytoskeleton remodeling, and it is required to invade host cells. TgADF was a promising vaccine candidate. To observe the immunological changes and protective efficacy of recombinant TgADF protein (rTgADF) against T. gondii infection, we optimized the intranasal immunization dose of rTgADF and analyzed the survival rate and tachyzoite loads in mouse tissues after oral challenge with T. gondii tachyzoites. RESULTS: rTgADF was prepared, purified, and combined with mouse anti-His antibody and rabbit anti-T. gondii serum. After intranasal immunization with 10 µg, 20 µg, 30 µg, or 40 µg of rTgADF, the 30-µg group elicited high levels of secretory IgA (sIgA) in nasal, intestinal, and vesical washes, raised IgG titres in the sera, strong proliferation of splenocytes, and increased secretion of IL-2 and IFN-γ when compared with the control group. When the mice were orally challenged with T. gondii, an increase in the survival rate (36.36 %) and a decrease in the tachyzoite loads in the liver (67.77 %) and brain (51.01 %) were observed. CONCLUSIONS: Our findings demonstrate that intranasal immunization with rTgADF can simultaneously trigger mucosal and systemic immune responses and protect the mice against T. gondii infection.


Subject(s)
Antigens, Protozoan/therapeutic use , Destrin/therapeutic use , Immunity, Mucosal , Lymphocytes/immunology , Recombinant Proteins/therapeutic use , Toxoplasma/immunology , Toxoplasmosis/therapy , Administration, Intranasal , Animals , Antigens, Protozoan/immunology , Cell Proliferation , Cells, Cultured , Destrin/immunology , Female , Humans , Immune Sera/administration & dosage , Immunoglobulin A/blood , Lymphocytes/parasitology , Mice , Mice, Inbred BALB C , Rabbits , Toxoplasmosis/immunology
18.
PLoS One ; 10(9): e0137683, 2015.
Article in English | MEDLINE | ID: mdl-26367128

ABSTRACT

In the present study, two Leishmania infantum hypothetical proteins present in the amastigote stage, LiHyp1 and LiHyp6, were combined with a promastigote protein, IgE-dependent histamine-releasing factor (HRF); to compose a polyproteins vaccine to be evaluated against L. infantum infection. Also, the antigenicity of the three proteins was analyzed, and their use for the serodiagnosis of canine visceral leishmaniasis (CVL) was evaluated. The LiHyp1, LiHyp6, and HRF DNA coding sequences were cloned in prokaryotic expression vectors and the recombinant proteins were purified. When employed in ELISA assays, all proteins were recognized by sera from visceral leishmaniasis (VL) dogs, and presented no cross-reactivity with either sera from dogs vaccinated with a Brazilian commercial vaccine, or sera of Trypanosoma cruzi-infected or Ehrlichia canis-infected animals. In addition, the antigens were not recognized by antibodies from non-infected animals living in endemic or non-endemic areas for leishmaniasis. The immunogenicity and protective efficacy of the three proteins administered in the presence of saponin, individually or in combination (composing a polyproteins vaccine), were evaluated in a VL murine model: BALB/c mice infected with L. infantum. Spleen cells from mice inoculated with the individual proteins or with the polyproteins vaccine plus saponin showed a protein-specific production of IFN-γ, IL-12, and GM-CSF after an in vitro stimulation, which was maintained after infection. These animals presented significant reductions in the parasite burden in different evaluated organs, when compared to mice inoculated with saline or saponin. The decrease in parasite burden was associated with an IL-12-dependent production of IFN-γ against parasite total extracts (produced mainly by CD4+ T cells), correlated to the induction of parasite proteins-driven NO production. Mice inoculated with the recombinant protein-based vaccines showed also high levels of parasite-specific IgG2a antibodies. The polyproteins vaccine administration induced a more pronounced Th1 response before and after challenge infection than individual vaccines, which was correlated to a higher control of parasite dissemination to internal organs.


Subject(s)
Antigens, Protozoan/therapeutic use , Leishmania infantum/immunology , Leishmaniasis, Visceral/prevention & control , Protozoan Proteins/immunology , Protozoan Vaccines/therapeutic use , Animals , Cytokines/metabolism , Dogs , Female , Immunity, Humoral , Leishmania infantum/growth & development , Mice, Inbred BALB C , Nitrites/metabolism , Parasite Load
19.
Immunobiology ; 220(3): 428-36, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25454810

ABSTRACT

Trypanosoma cruzi is a real challenge to the host's immune system, because it requires strong humoral and cellular immune response to remove circulating trypomastigote forms, and to prevent the replication of amastigote forms in tissues, involving many regulator and effector components. This protozoan is responsible for Chagas disease, a major public health problem in Latinamerica. We have developed a model of vaccination with Trypanosoma rangeli, a parasite closely related to T. cruzi, but nonpathogenic to humans, which reduces the infectiousness in three different species of animals, mice, dogs and guinea pigs, against challenge with T. cruzi. In a previous work, we demonstrated that mice vaccinated with T. rangeli showed important soluble mediators that stimulate phagocytic activity versus only infected groups. The aim of this work was to study the innate immune response in mice vaccinated or not with T. rangeli. Different population cells and some soluble mediators (cytokines) in peritoneal fluid and plasma in mice vaccinated-infected and only infected with T. cruzi were studied. In the first hours of challenge vaccinated mice showed an increase of macrophages, NK, granulocytes, and regulation of IL6, IFNγ, TNFα and IL10, with an increase of IL12, with respect to only infected mice. Furthermore an increase was observed of Li T, Li B responsible for adaptative response. Finally the findings showed that the innate immune response plays an important role in vaccinated mice for the early elimination of the parasites, complementary with the adaptative immune response, suggesting that vaccination with T. rangeli modulates the innate response, which develops some kind of immunological memory, recognizing shared antigens with T. cruzi. These results could contribute to the knowledge of new mechanisms which would have an important role in the immune response to Chagas disease.


Subject(s)
Chagas Disease/immunology , Immunologic Memory/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Trypanosoma rangeli/immunology , Adaptive Immunity/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/therapeutic use , Chagas Disease/prevention & control , Cross Protection/immunology , Female , Granulocytes/immunology , Immunity, Innate/immunology , Interferon-gamma/blood , Interleukin-10/blood , Interleukin-12/blood , Interleukin-6/blood , Killer Cells, Natural/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Parasitemia/immunology , Parasitemia/prevention & control , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factor-alpha/blood , Vaccination
20.
PLoS One ; 8(2): e56061, 2013.
Article in English | MEDLINE | ID: mdl-23457498

ABSTRACT

A recent clinical trial in African children demonstrated the potential utility of merozoite surface protein (MSP)-3 as a vaccine against Plasmodium falciparum malaria. The present study evaluated the use of Plasmodium vivax MSP-3 (PvMSP-3) as a target antigen in vaccine formulations against malaria caused by P. vivax. Recombinant proteins representing MSP-3α and MSP-3ß of P. vivax were expressed as soluble histidine-tagged bacterial fusions. Antigenicity during natural infection was evaluated by detecting specific antibodies using sera from individuals living in endemic areas of Brazil. A large proportion of infected individuals presented IgG antibodies to PvMSP-3α (68.2%) and at least 1 recombinant protein representing PvMSP-3ß (79.1%). In spite of the large responder frequency, reactivity to both antigens was significantly lower than was observed for the immunodominant epitope present on the 19-kDa C-terminal region of PvMSP-1. Immunogenicity of the recombinant proteins was studied in mice in the absence or presence of different adjuvant formulations. PvMSP-3ß, but not PvMSP-3α, induced a TLR4-independent humoral immune response in the absence of any adjuvant formulation. The immunogenicity of the recombinant antigens were also tested in formulations containing different adjuvants (Alum, Salmonella enterica flagellin, CpG, Quil A,TiterMax® and incomplete Freunds adjuvant) and combinations of two adjuvants (Alum plus flagellin, and CpG plus flagellin). Recombinant PvMSP-3α and PvMSP-3ß elicited higher antibody titers capable of recognizing P. vivax-infected erythrocytes harvested from malaria patients. Our results confirm that P. vivax MSP-3 antigens are immunogenic during natural infection, and the corresponding recombinant proteins may be useful in elucidating their vaccine potential.


Subject(s)
Antigens, Protozoan/therapeutic use , Malaria Vaccines/therapeutic use , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/therapeutic use , Animals , Antibody Formation , Antigens, Protozoan/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Malaria Vaccines/immunology , Malaria, Vivax/blood , Malaria, Vivax/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protozoan Proteins/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...