205 results on '"Nussenzweig RS"'
Search Results
2. Immunity to Plasmodium sporozoites: recent advances and applications to field research
- Author
-
Esposito, F, Lombardi, S, Modiano, David, Zavala, F, Reeme, J, Lamizana, L, Coluzzi, M, and Nussenzweig, Rs
- Published
- 1986
3. Interferon-gamma-independent CD8(+) T cell-mediated protective anti-malaria immunity elicited by recombinant adenovirus
- Author
-
Elaine Guadelupe Rodrigues, Claassen, J., Lee, S., Wilson, Jm, Nussenzweig, Rs, and Tsuji, M.
4. INFLUENZA AND VACCINIA VIRUSES EXPRESSING MALARIA CD8(+) T-CELL AND B-CELL EPITOPES - COMPARISON OF THEIR IMMUNOGENICITY AND CAPACITY TO INDUCE PROTECTIVE IMMUNITY
- Author
-
Rodrigues, M., Li, Sq, Murata, K., Rodriguez, D., Rodriguez, Jr, Bacik, I., Bennink, Jr, Yewdell, Jw, Garciasastre, A., Nussenzweig, Rs, Esteban, M., Palese, P., and Zavala, F.
5. Prime-boost vaccination with recombinant protein and adenovirus-vector expressing Plasmodium vivax circumsporozoite protein (CSP) partially protects mice against Pb/Pv sporozoite challenge.
- Author
-
de Camargo TM, de Freitas EO, Gimenez AM, Lima LC, de Almeida Caramico K, Françoso KS, Bruna-Romero O, Andolina C, Nosten F, Rénia L, Ertl HCJ, Nussenzweig RS, Nussenzweig V, Rodrigues MM, Reyes-Sandoval A, and Soares IS
- Subjects
- Adenoviridae genetics, Amino Acid Sequence, Animals, Antibodies, Protozoan immunology, Antibody Affinity immunology, Disease Models, Animal, Female, Genetic Vectors administration & dosage, Genetic Vectors chemistry, Immunization, Immunogenicity, Vaccine, Immunoglobulin G blood, Immunoglobulin G immunology, Malaria Vaccines genetics, Malaria, Vivax mortality, Mice, Plasmodium vivax genetics, Protozoan Proteins chemistry, Protozoan Proteins genetics, Malaria Vaccines immunology, Malaria, Vivax immunology, Malaria, Vivax prevention & control, Plasmodium vivax immunology, Protozoan Proteins immunology, Recombinant Proteins immunology
- Abstract
Vaccine development against Plasmodium vivax malaria lags behind that for Plasmodium falciparum. To narrow this gap, we administered recombinant antigens based on P. vivax circumsporozoite protein (CSP) to mice. We expressed in Pichia pastoris two chimeric proteins by merging the three central repeat regions of different CSP alleles (VK210, VK247, and P. vivax-like). The first construct (yPvCSP-All
FL ) contained the fused repeat regions flanked by N- and C-terminal regions. The second construct (yPvCSP-AllCT ) contained the fused repeat regions and the C-terminal domain, plus RI region. Mice were vaccinated with three doses of yPvCSP in adjuvants Poly (I:C) or Montanide ISA720. We also used replication-defective adenovirus vectors expressing CSP of human serotype 5 (AdHu5) and chimpanzee serotype 68 (AdC68) for priming mice which were subsequently boosted twice with yPvCSP proteins in Poly (I:C) adjuvant. Regardless of the regime used, immunized mice generated high IgG titres specific to all CSP alleles. After challenge with P. berghei ANKA transgenic parasites expressing Pb/PvVK210 or Pb/PvVK247 sporozoites, significant time delays for parasitemia were observed in all vaccinated mice. These vaccine formulations should be clinically tried for their potential as protective universal vaccine against P. vivax malaria.- Published
- 2018
- Full Text
- View/download PDF
6. Vaccine Containing the Three Allelic Variants of the Plasmodium vivax Circumsporozoite Antigen Induces Protection in Mice after Challenge with a Transgenic Rodent Malaria Parasite.
- Author
-
Gimenez AM, Lima LC, Françoso KS, Denapoli PMA, Panatieri R, Bargieri DY, Thiberge JM, Andolina C, Nosten F, Renia L, Nussenzweig RS, Nussenzweig V, Amino R, Rodrigues MM, and Soares IS
- Abstract
Plasmodium vivax is the most common species that cause malaria outside of the African continent. The development of an efficacious vaccine would contribute greatly to control malaria. Recently, using bacterial and adenoviral recombinant proteins based on the P. vivax circumsporozoite protein (CSP), we demonstrated the possibility of eliciting strong antibody-mediated immune responses to each of the three allelic forms of P. vivax CSP (PvCSP). In the present study, recombinant proteins representing the PvCSP alleles (VK210, VK247, and P. vivax -like), as well as a hybrid polypeptide, named PvCSP-All epitopes, were generated. This hybrid containing the conserved C-terminal of the PvCSP and the three variant repeat domains in tandem were successfully produced in the yeast Pichia pastoris . After purification and biochemical characterization, they were used for the experimental immunization of C57BL/6 mice in a vaccine formulation containing the adjuvant Poly(I:C). Immunization with a recombinant protein expressing all three different allelic forms in fusion elicited high IgG antibody titers reacting with all three different allelic variants of PvCSP. The antibodies targeted both the C-terminal and repeat domains of PvCSP and recognized the native protein on the surface of P. vivax sporozoites. More importantly, mice that received the vaccine formulation were protected after challenge with chimeric Plasmodium berghei sporozoites expressing CSP repeats of P. vivax sporozoites (Pb/PvVK210). Our results suggest that it is possible to elicit protective immunity against one of the most common PvCSP alleles using soluble recombinant proteins expressed by P. pastoris . These recombinant proteins are promising candidates for clinical trials aiming to develop a multiallele vaccine against P. vivax malaria.
- Published
- 2017
- Full Text
- View/download PDF
7. Human CD8+ T cells mediate protective immunity induced by a human malaria vaccine in human immune system mice.
- Author
-
Li X, Huang J, Zhang M, Funakoshi R, Sheetij D, Spaccapelo R, Crisanti A, Nussenzweig V, Nussenzweig RS, and Tsuji M
- Subjects
- Animals, Dependovirus, Genetic Vectors, HLA-A2 Antigen genetics, Humans, Mice, Mice, Inbred NOD, Mice, Transgenic, Plasmodium falciparum, Protozoan Proteins immunology, Sporozoites, CD8-Positive T-Lymphocytes immunology, Malaria Vaccines immunology, Malaria, Falciparum immunology, Malaria, Falciparum prevention & control
- Abstract
A number of studies have shown that CD8+ T cells mediate protective anti-malaria immunity in a mouse model. However, whether human CD8+ T cells play a role in protection against malaria remains unknown. We recently established human immune system (HIS) mice harboring functional human CD8+ T cells (HIS-CD8 mice) by transduction with HLA-A∗0201 and certain human cytokines using recombinant adeno-associated virus-based gene transfer technologies. These HIS-CD8 mice mount a potent, antigen-specific HLA-A∗0201-restricted human CD8+ T-cell response upon immunization with a recombinant adenovirus expressing a human malaria antigen, the Plasmodium falciparum circumsporozoite protein (PfCSP), termed AdPfCSP. In the present study, we challenged AdPfCSP-immunized HIS-CD8 mice with transgenic Plasmodium berghei sporozoites expressing full-length PfCSP and found that AdPfCSP-immunized (but not naïve) mice were protected against subsequent malaria challenge. The level of the HLA-A∗0201-restricted, PfCSP-specific human CD8+ T-cell response was closely correlated with the level of malaria protection. Furthermore, depletion of human CD8+ T cells from AdPfCSP-immunized HIS-CD8 mice almost completely abolished the anti-malaria immune response. Taken together, our data show that human CD8+ T cells mediate protective anti-malaria immunity in vivo., (Copyright © 2016 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
8. Human immune system mice immunized with Plasmodium falciparum circumsporozoite protein induce protective human humoral immunity against malaria.
- Author
-
Huang J, Li X, Coelho-dos-Reis JG, Zhang M, Mitchell R, Nogueira RT, Tsao T, Noe AR, Ayala R, Sahi V, Gutierrez GM, Nussenzweig V, Wilson JM, Nardin EH, Nussenzweig RS, and Tsuji M
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay, Heterografts, Histocompatibility Antigens Class II, Humans, Malaria Vaccines, Mice, Protozoan Proteins immunology, Disease Models, Animal, Immunity, Humoral immunology, Malaria, Falciparum immunology, Mice, Transgenic immunology
- Abstract
In this study, we developed human immune system (HIS) mice that possess functional human CD4+ T cells and B cells, named HIS-CD4/B mice. HIS-CD4/B mice were generated by first introducing HLA class II genes, including DR1 and DR4, along with genes encoding various human cytokines and human B cell activation factor (BAFF) to NSG mice by adeno-associated virus serotype 9 (AAV9) vectors, followed by engrafting human hematopoietic stem cells (HSCs). HIS-CD4/B mice, in which the reconstitution of human CD4+ T and B cells resembles to that of humans, produced a significant level of human IgG against Plasmodium falciparum circumsporozoite (PfCS) protein upon immunization. CD4+ T cells in HIS-CD4/B mice, which possess central and effector memory phenotypes like those in humans, are functional, since PfCS protein-specific human CD4+ T cells secreting IFN-γ and IL-2 were detected in immunized HIS-CD4/B mice. Lastly, PfCS protein-immunized HIS-CD4/B mice were protected from in vivo challenge with transgenic P. berghei sporozoites expressing the PfCS protein. The immune sera collected from protected HIS-CD4/B mice reacted against transgenic P. berghei sporozoites expressing the PfCS protein and also inhibited the parasite invasion into hepatocytes in vitro. Taken together, these studies show that our HIS-CD4/B mice could mount protective human anti-malaria immunity, consisting of human IgG and human CD4+ T cell responses both specific for a human malaria antigen., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
9. Immunogenicity of a prime-boost vaccine containing the circumsporozoite proteins of Plasmodium vivax in rodents.
- Author
-
Teixeira LH, Tararam CA, Lasaro MO, Camacho AG, Ersching J, Leal MT, Herrera S, Bruna-Romero O, Soares IS, Nussenzweig RS, Ertl HC, Nussenzweig V, and Rodrigues MM
- Subjects
- Adjuvants, Immunologic administration & dosage, Animals, Antibodies, Protozoan blood, Female, Immunoglobulin G blood, Malaria Vaccines administration & dosage, Malaria Vaccines genetics, Malaria, Vivax immunology, Mice, Mice, Inbred C57BL, Plasmodium vivax genetics, Poly I-C administration & dosage, Protozoan Proteins genetics, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Malaria Vaccines immunology, Malaria, Vivax prevention & control, Plasmodium vivax immunology, Protozoan Proteins immunology, Vaccination methods
- Abstract
Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.
- Published
- 2014
- Full Text
- View/download PDF
10. Antibodies to Plasmodium circumsporozoite protein (CSP) inhibit sporozoite's cell traversal activity.
- Author
-
Mishra S, Nussenzweig RS, and Nussenzweig V
- Subjects
- Animals, Cell Line, Dogs, Electric Impedance, Intracellular Signaling Peptides and Proteins immunology, Tight Junctions immunology, Antibodies, Monoclonal immunology, Antibodies, Protozoan immunology, Plasmodium berghei immunology, Protozoan Proteins immunology, Sporozoites immunology
- Abstract
Plasmodium sporozoites are deposited in the skin of the mammalian host by Anopheles mosquitoes. To continue the life cycle, the sporozoites have to invade the host's hepatocytes, where they transform into exoerythrocytic forms (EEFs) inside a parasitophorous vacuole. During their route from the skin to the liver, the parasites traverse the capillary epithelium in the dermis to enter the blood circulation, and cross the endothelium of liver sinusoids to enter the parenchyma. Cell traversal by sporozoites is usually measured by quantifying dyes that enter or are released from cells during incubation with salivary gland sporozoites. These methods do not distinguish cell traversal from cell wounding. Here we validate an assay that quantifies cell traversal of sporozoites through monolayers of MDCK cells that form tight junctions. We compared cell traversal of wt sporozoites and of parasites lacking the Type I membrane protein TLP (TRAP-like protein) previously implicated in cell traversal. We provide direct evidence that TLP ko sporozoites are defective in cell traversal and that they are retained inside the MDCK cytoplasm. We then used the MDCK assay to study the effect of a monoclonal antibody (3D11) to the circumsporozoite protein (CSP) on the parasite's cell traversal. We show that 3D11 inhibits cell traversal at nanomolar concentrations. We conclude that antibodies elicited by CSP-based vaccines are likely to inhibit the migration of sporozoites from the skin to the liver., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
11. Identification of non-CSP antigens bearing CD8 epitopes in mice immunized with irradiated sporozoites.
- Author
-
Mishra S, Rai U, Shiratsuchi T, Li X, Vanloubbeeck Y, Cohen J, Nussenzweig RS, Winzeler EA, Tsuji M, and Nussenzweig V
- Subjects
- Adenoviridae genetics, Adenoviridae immunology, Animals, CD8-Positive T-Lymphocytes immunology, Female, Immunization, Interferon-gamma, Malaria immunology, Malaria prevention & control, Mice, Mice, Inbred BALB C, Mice, Transgenic, Sporozoites radiation effects, Antigens, Protozoan immunology, Epitopes, T-Lymphocyte immunology, Malaria Vaccines immunology, Plasmodium yoelii immunology, Protozoan Proteins immunology, Sporozoites immunology
- Abstract
Immunization of BALB/c mice with irradiated sporozoites (IrSp) of Plasmodium yoelii can lead to sterile immunity. The circumsporozoite protein (CSP) plays a dominant role in protection. Nevertheless after hyper-immunization with IrSp, complete protection is obtained in CSP-transgenic BALB/c mice that are T-cell tolerant to the CSP and cannot produce antibodies [CSP-Tg/JhT(-/-)]. This protection is mediated exclusively by CD8(+) T cells [1]. To identify the non-CSP protective T cell antigens, we studied the properties of 34 P. yoelii sporozoite antigens that are predicted to be secreted and to contain strong Kd-restricted CD8(+) T cell epitopes. The synthetic peptides corresponding to the epitopes were used to screen for the presence of peptide-specific CD8(+) T cells secreting interferon-γ (IFN-γ) in splenocytes from CSP-Tg/JhT(-/-) BALB/c mice hyper immunized with IrSp. However, the numbers of IFN-γ-secreting splenocytes specific for the non-CSP antigen-derived peptides were 20-100 times lower than those specific for the CSP-specific peptide. When mice were immunized with recombinant adenoviruses expressing selected non-CSP antigens, the animals were not protected against challenge with P. yoelii sporozoites although large numbers of CD8(+) specific T cells were generated., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
12. TLR5-dependent immunogenicity of a recombinant fusion protein containing an immunodominant epitope of malarial circumsporozoite protein and the FliC flagellin of Salmonella Typhimurium.
- Author
-
Camacho AG, Teixeira LH, Bargieri DY, Boscardin SB, Soares IS, Nussenzweig RS, Nussenzweig V, and Rodrigues MM
- Subjects
- Animals, Antibodies, Protozoan immunology, Epitopes, B-Lymphocyte immunology, Epitopes, B-Lymphocyte metabolism, Escherichia coli Proteins immunology, Flagellin metabolism, Immunodominant Epitopes metabolism, Malaria Vaccines metabolism, Malaria, Vivax immunology, Mice, Mice, Inbred C57BL, Protozoan Proteins immunology, Protozoan Proteins metabolism, Recombinant Fusion Proteins metabolism, Salmonella typhimurium metabolism, Toll-Like Receptor 5 immunology, Flagellin immunology, Immunodominant Epitopes immunology, Malaria Vaccines immunology, Malaria, Vivax prevention & control, Plasmodium falciparum immunology, Recombinant Fusion Proteins immunology, Salmonella typhimurium immunology
- Abstract
Recently, we described the improved immunogenicity of new malaria vaccine candidates based on the expression of fusion proteins containing immunodominant epitopes of merozoites and Salmonella enterica serovar Typhimurium flagellin (FliC) protein as an innate immune agonist. Here, we tested whether a similar strategy, based on an immunodominant B-cell epitope from malaria sporozoites, could also generate immunogenic fusion polypeptides. A recombinant His6-tagged FliC protein containing the C-terminal repeat regions of the VK210 variant of Plasmodium vivax circumsporozoite (CS) protein was constructed. This recombinant protein was successfully expressed in Escherichia coli as soluble protein and was purified by affinity to Ni-agarose beads followed by ion exchange chromatography. A monoclonal antibody specific for the CS protein of P. vivax sporozoites (VK210) was able to recognise the purified protein. C57BL/6 mice subcutaneously immunised with the recombinant fusion protein in the absence of any conventional adjuvant developed protein-specific systemic antibody responses. However, in mice genetically deficient in expression of TLR5, this immune response was extremely low. These results extend our previous observations concerning the immunogenicity of these recombinant fusion proteins and provide evidence that the main mechanism responsible for this immune activation involves interactions with TLR5, which has not previously been demonstrated for any recombinant FliC fusion protein.
- Published
- 2011
- Full Text
- View/download PDF
13. Breakthroughs towards a malaria vaccine.
- Author
-
Nussenzweig RS and Krettli AU
- Abstract
Since the 1960s, the scientist Ruth Nussenzweig, C.V. Starr Professor at New York University School of Medicine, has been working to develop an antimalarial vaccine. In her testimony, she traces some of the stages through which her research has passed. At the beginning of her studies, most scientists held that it would be impossible to develop such a vaccine. However, a different opinion had been expressed in a paper on avian malaria written some forty years earlier by a British researcher and his collaborators from India. The immunization principle developed by this group was irradiation of sporozoites in order to deactivate the parasite that causes malaria. Ruth Nussenzweig revived and expanded upon this line of research, which now underpins her efforts to devise an antimalarial vaccine for human use.
- Published
- 2011
- Full Text
- View/download PDF
14. The Plasmodium eukaryotic initiation factor-2alpha kinase IK2 controls the latency of sporozoites in the mosquito salivary glands.
- Author
-
Zhang M, Fennell C, Ranford-Cartwright L, Sakthivel R, Gueirard P, Meister S, Caspi A, Doerig C, Nussenzweig RS, Tuteja R, Sullivan WJ Jr, Roos DS, Fontoura BM, Ménard R, Winzeler EA, and Nussenzweig V
- Subjects
- Animals, Cell Line, Cytoplasmic Granules metabolism, Gene Expression Regulation, Gene Targeting, Life Cycle Stages, Liver metabolism, Liver parasitology, Mice, Mice, Inbred C57BL, Phenotype, Phosphoprotein Phosphatases metabolism, Phosphorylation, Plasmodium berghei cytology, Plasmodium berghei pathogenicity, Plasmodium berghei ultrastructure, Protein Biosynthesis, Protozoan Proteins genetics, Protozoan Proteins metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Salivary Glands cytology, Salivary Glands ultrastructure, Sporozoites cytology, Sporozoites ultrastructure, Culicidae parasitology, Plasmodium berghei enzymology, Salivary Glands parasitology, Sporozoites enzymology, eIF-2 Kinase metabolism
- Abstract
Sporozoites, the invasive form of malaria parasites transmitted by mosquitoes, are quiescent while in the insect salivary glands. Sporozoites only differentiate inside of the hepatocytes of the mammalian host. We show that sporozoite latency is an active process controlled by a eukaryotic initiation factor-2alpha (eIF2alpha) kinase (IK2) and a phosphatase. IK2 activity is dominant in salivary gland sporozoites, leading to an inhibition of translation and accumulation of stalled mRNAs into granules. When sporozoites are injected into the mammalian host, an eIF2alpha phosphatase removes the PO4 from eIF2alpha-P, and the repression of translation is alleviated to permit their transformation into liver stages. In IK2 knockout sporozoites, eIF2alpha is not phosphorylated and the parasites transform prematurely into liver stages and lose their infectivity. Thus, to complete their life cycle, Plasmodium sporozoites exploit the mechanism that regulates stress responses in eukaryotic cells.
- Published
- 2010
- Full Text
- View/download PDF
15. Immunogenicity and protective efficacy of a recombinant yellow fever vaccine against the murine malarial parasite Plasmodium yoelii.
- Author
-
Stoyanov CT, Boscardin SB, Deroubaix S, Barba-Spaeth G, Franco D, Nussenzweig RS, Nussenzweig M, and Rice CM
- Subjects
- Adoptive Transfer, Animals, CD8-Positive T-Lymphocytes immunology, Female, Immunity, Cellular, Immunization, Secondary, Interferon-gamma immunology, Interleukin-2 immunology, Malaria immunology, Mice, Mice, Inbred BALB C, Mice, Transgenic, Plasmids, Plasmodium yoelii immunology, Vaccines, Attenuated immunology, Malaria prevention & control, Malaria Vaccines immunology, Protozoan Proteins immunology, Yellow Fever Vaccine immunology
- Abstract
The live-attenuated yellow fever vaccine (YF17D) is one of the safest and most effective vaccines available today. Here, YF17D was genetically altered to express the circumsporozoite protein (CSP) from the murine malarial parasite Plasmodium yoelii. Reconstituted recombinant virus was viable and exhibited robust CSP expression. Immunization of naïve mice resulted in extensive proliferation of adoptively transferred CSP-specific transgenic CD8(+) T-cells. A single immunization of naïve mice with recombinant YF17D resulted in robust production of IFN-gamma by CD8(+) T-cells and IFN-gamma and IL-2 by CD4(+) T-cells. A prime-boost regimen consisting of recombinant virus followed by a low-dose of irradiated sporozoites conferred protection against challenge with P. yoelii. Taken together, these results show that recombinant YF17D can efficiently express CSP in culture, and prime a protective immune response in vivo., ((c) 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
16. The circumsporozoite protein is an immunodominant protective antigen in irradiated sporozoites.
- Author
-
Kumar KA, Sano G, Boscardin S, Nussenzweig RS, Nussenzweig MC, Zavala F, and Nussenzweig V
- Subjects
- Animals, Malaria Vaccines immunology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Sporozoites chemistry, T-Lymphocytes, Cytotoxic immunology, Antigens, Protozoan immunology, Immunodominant Epitopes immunology, Protozoan Proteins immunology, Sporozoites immunology, Sporozoites radiation effects
- Abstract
Malaria infection starts when mosquitoes inject sporozoites into the skin. The parasites enter the blood stream and make their way to the liver where they develop into the exo-erythrocytic forms (EEFs). Immunization with irradiated sporozoites (IrSp) leads to robust protection against malaria infection in rodents, monkeys and humans by eliciting antibodies to circumsporozoite protein (CS) that inhibit sporozoite infectivity, and T cells that destroy the EEFs. To study the role of non-CS antigens in protection, we produced CS transgenic mice that were tolerant to CS T-cell epitopes. Here we show that in the absence of T-cell-dependent immune responses to CS, protection induced by immunization with two doses of IrSp was greatly reduced. Thus, although hundreds of other Plasmodium genes are expressed in sporozoites and EEFs, CS is a dominant protective antigen. Nevertheless, sterile immunity could be obtained by immunization of CS transgenics with three doses of IrSp.
- Published
- 2006
- Full Text
- View/download PDF
17. Antigen targeting to dendritic cells elicits long-lived T cell help for antibody responses.
- Author
-
Boscardin SB, Hafalla JC, Masilamani RF, Kamphorst AO, Zebroski HA, Rai U, Morrot A, Zavala F, Steinman RM, Nussenzweig RS, and Nussenzweig MC
- Subjects
- Adjuvants, Immunologic administration & dosage, Alum Compounds administration & dosage, Animals, Antibody Formation drug effects, Antigen Presentation drug effects, Chickens, Haptens immunology, Humans, Immunization methods, Immunoglobulin G immunology, Immunologic Memory drug effects, Immunologic Memory immunology, Mice, Mice, Inbred BALB C, Ovalbumin immunology, Plasmodium yoelii immunology, Protozoan Proteins immunology, Antibody Formation immunology, Antigen Presentation immunology, B-Lymphocytes immunology, Dendritic Cells immunology, T-Lymphocytes immunology
- Abstract
Resistance to several prevalent infectious diseases requires both cellular and humoral immune responses. T cell immunity is initiated by mature dendritic cells (DCs) in lymphoid organs, whereas humoral responses to most antigens require further collaboration between primed, antigen-specific helper T cells and naive or memory B cells. To determine whether antigens delivered to DCs in lymphoid organs induce T cell help for antibody responses, we targeted a carrier protein, ovalbumin (OVA), to DCs in the presence of a maturation stimulus and assayed for antibodies to a hapten, (4-hydroxy-3-nitrophenyl) acetyl (NP), after boosting with OVA-NP. A single DC-targeted immunization elicited long-lived T cell helper responses to the carrier protein, leading to large numbers of antibody-secreting cells and high titers of high-affinity antihapten immunoglobulin Gs. Small doses of DC-targeted OVA induced higher titers and a broader spectrum of anti-NP antibody isotypes than large doses of OVA in alum adjuvant. Similar results were obtained when the circumsporozoite protein of Plasmodium yoelii was delivered to DCs. We conclude that antigen targeting to DCs combined with a maturation stimulus produces broad-based and long-lived T cell help for humoral immune responses.
- Published
- 2006
- Full Text
- View/download PDF
18. Yellow fever 17D as a vaccine vector for microbial CTL epitopes: protection in a rodent malaria model.
- Author
-
Tao D, Barba-Spaeth G, Rai U, Nussenzweig V, Rice CM, and Nussenzweig RS
- Subjects
- Animals, Antigens, Protozoan immunology, CD8-Positive T-Lymphocytes immunology, Humans, Malaria immunology, Mice, Plasmodium yoelii immunology, Yellow Fever Vaccine genetics, Yellow fever virus genetics, Yellow fever virus immunology, Epitopes immunology, Malaria prevention & control, T-Lymphocytes, Cytotoxic immunology, Yellow Fever Vaccine immunology
- Abstract
The yellow fever vaccine 17D (17D) is safe, and after a single immunizing dose, elicits long-lasting, perhaps lifelong protective immunity. One of the major challenges facing delivery of human vaccines in underdeveloped countries is the need for multiple injections to achieve full efficacy. To examine 17D as a vector for microbial T cell epitopes, we inserted the H-2K(d)-restricted CTL epitope of the circumsporozoite protein (CS) of Plasmodium yoelii between 17D nonstructural proteins NS2B and NS3. The recombinant virus, 17D-Py, was replication competent and stable in vitro and in vivo. A single subcutaneous injection of 10(5) PFU diminished the parasite burden in the liver by approximately 70%. The high level of protection lasted between 4 and 8 wk after immunization, but a significant effect was documented even 24 wk afterwards. Thus, the immunogenicity of a foreign T cell epitope inserted into 17D mimics some of the remarkable properties of the human vaccine. Priming with 17D-Py followed by boosting with irradiated sporozoites conferred sterile immunity to 90% of the mice. This finding indicates that the immune response of vaccine-primed individuals living in endemic areas could be sustained and magnified by the bite of infected mosquitoes.
- Published
- 2005
- Full Text
- View/download PDF
19. Induction of protective immunity against malaria by priming-boosting immunization with recombinant cold-adapted influenza and modified vaccinia Ankara viruses expressing a CD8+-T-cell epitope derived from the circumsporozoite protein of Plasmodium yoelii.
- Author
-
González-Aseguinolaza G, Nakaya Y, Molano A, Dy E, Esteban M, Rodríguez D, Rodríguez JR, Palese P, García-Sastre A, and Nussenzweig RS
- Subjects
- Adaptation, Physiological, Animals, Antibodies, Protozoan blood, Cold Temperature, Disease Models, Animal, Epitopes, T-Lymphocyte genetics, Epitopes, T-Lymphocyte metabolism, Female, Genetic Vectors, Humans, Immunization, Immunization, Secondary, Malaria immunology, Malaria Vaccines administration & dosage, Malaria Vaccines genetics, Mice, Mice, Inbred BALB C, Orthomyxoviridae genetics, Plasmodium yoelii genetics, Protozoan Proteins genetics, Recombination, Genetic, Vaccinia virus genetics, CD8-Positive T-Lymphocytes immunology, Epitopes, T-Lymphocyte immunology, Malaria prevention & control, Malaria Vaccines immunology, Orthomyxoviridae immunology, Plasmodium yoelii immunology, Protozoan Proteins immunology, Vaccinia virus immunology
- Abstract
We immunized mice with an attenuated (cold-adapted) influenza virus followed by an attenuated vaccinia virus (modified vaccinia virus Ankara), both expressing a CD8(+)-T-cell epitope derived from malaria sporozoites. This vaccination regimen elicited high levels of protection against malaria. This is the first time that the vaccine efficacy of a recombinant cold-adapted influenza virus vector expressing a foreign antigen has been evaluated.
- Published
- 2003
- Full Text
- View/download PDF
20. Delayed-type hypersensitivity in volunteers immunized with a synthetic multi-antigen peptide vaccine (PfCS-MAP1NYU) against Plasmodium falciparum sporozoites.
- Author
-
Kublin JG, Lowitt MH, Hamilton RG, Oliveira GA, Nardin EH, Nussenzweig RS, Schmeckpeper BJ, Diggs CL, Bodison SA, and Edelman R
- Subjects
- Adjuvants, Immunologic administration & dosage, Adolescent, Adult, Animals, Antibodies, Protozoan biosynthesis, Antigens, Protozoan administration & dosage, Humans, Hypersensitivity, Delayed, Immunoglobulin G biosynthesis, Intradermal Tests, Malaria Vaccines administration & dosage, Malaria Vaccines immunology, Malaria, Falciparum immunology, Malaria, Falciparum prevention & control, Middle Aged, Plasmodium falciparum growth & development, T-Lymphocytes immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic immunology, Vaccines, Synthetic pharmacology, Malaria Vaccines pharmacology, Plasmodium falciparum immunology
- Abstract
During the testing of the safety and immunogenicity of an adjuvanted, synthetic Plasmodium falciparum CS multiple antigen peptide (MAP) vaccine, we investigated the potential for using cutaneous delayed-type hypersensitivity (DTH) reactions as a correlate of immune response. We evaluated 27 of our volunteers for DTH reactions to intradermal inoculation (0.02 ml) of several concentrations of the MAP vaccine and adjuvant control solutions. Induration was measured 2 days after skin tests were applied. Nine of 14 vaccinees (64%) with serum, high-titered anti-MAP antibody developed positive DTH (>or=5mm induration), that first appeared by 29 days after immunization and persisted for at least 3-6 months after 1-2 more immunizations. In contrast, DTH responses were negative in eight of eight vaccinees with no or low antibody titers, and in five of five non-immunized volunteers. Biopsies of positive DTH skin test sites were histologically compatible with a DTH reaction. We conclude that the presence of T cell functional activity reflected by a positive DTH skin test response to the MAP antigen serves as another marker for vaccine immunogenicity.
- Published
- 2002
- Full Text
- View/download PDF
21. Surface expression of an immunodominant malaria protein B cell epitope by yellow fever virus.
- Author
-
Bonaldo MC, Garratt RC, Caufour PS, Freire MS, Rodrigues MM, Nussenzweig RS, and Galler R
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal immunology, Antigens, Protozoan chemistry, Antigens, Protozoan genetics, Antigens, Protozoan immunology, Cell Line, Epitopes, B-Lymphocyte chemistry, Epitopes, B-Lymphocyte genetics, Gene Expression, Genome, Viral, Hydrogen-Ion Concentration, Immunodominant Epitopes chemistry, Immunodominant Epitopes genetics, Malaria parasitology, Malaria Vaccines administration & dosage, Malaria Vaccines adverse effects, Mice, Models, Molecular, Molecular Sequence Data, Neutralization Tests, Plasmodium falciparum chemistry, Plasmodium falciparum genetics, Protein Structure, Tertiary, Sequence Alignment, Serial Passage, Survival Rate, Vaccines, Attenuated adverse effects, Vaccines, Attenuated genetics, Vaccines, Attenuated immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic adverse effects, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Viral Envelope Proteins chemistry, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Yellow fever virus isolation & purification, Yellow fever virus pathogenicity, Yellow fever virus physiology, Epitopes, B-Lymphocyte immunology, Immunodominant Epitopes immunology, Malaria immunology, Malaria Vaccines genetics, Malaria Vaccines immunology, Plasmodium falciparum immunology, Yellow fever virus genetics
- Abstract
The yellow fever 17D virus (YF17D) has several characteristics that are desirable for the development of new, live attenuated vaccines. We approached its development as a vector for heterologous antigens by studying the expression of a humoral epitope at the surface of the E protein based on the results of modelling its three-dimensional structure. This model indicated that the most promising insertion site is between beta-strands f and g, a site that is exposed at the external surface of the virus. The large deletion of six residues from the fg loop of the E protein from yellow fever virus, compared to tick-born encephalitis virus, leaves space at the dimer interface for a large insertion without creating steric hindrance. We have tested this hypothesis by inserting a model humoral epitope from the circumsporozoite protein of Plasmodium falciparum consisting of triple NANP repeats. Recombinant virus (17D/8) expressing this insertion flanked by two glycine residues at each end, is specifically neutralized by a monoclonal antibody to the model epitope. Furthermore, mouse antibodies raised to the recombinant virus recognize the parasite protein in an ELISA assay. Serial passage analysis confirmed the genetic stability of the insertion made in the viral genome and the resulting 17D/8 virus is significantly more attenuated in mouse neurovirulence tests than the 17DD vaccine. The fg loop belongs to the dimerization domain of the E protein and lies at the interface between monomers. This domain undergoes a low pH transition, which is related to the fusion of the viral envelope to the endosome membrane. It is conceivable that a slower rate of fusion, resulting from the insertion close to the dimer interface, may delay the onset of virus production and thereby lead to a milder infection of the host. This would account for the more attenuated phenotype of the recombinant virus in the mouse model and lower extent of replication in cultured cells. The vectorial capacity of the yellow fever virus is being further explored for the expression and presentation of other epitopes, including those mediating T-cell responses., (Copyright 2002 Academic Press.)
- Published
- 2002
- Full Text
- View/download PDF
22. Complete, long-lasting protection against malaria of mice primed and boosted with two distinct viral vectors expressing the same plasmodial antigen.
- Author
-
Bruña-Romero O, González-Aseguinolaza G, Hafalla JC, Tsuji M, and Nussenzweig RS
- Subjects
- Animals, Antibody Formation, Antimalarials therapeutic use, BCG Vaccine therapeutic use, Disease Models, Animal, Fibrosis, Humans, Isoniazid therapeutic use, Macaca fascicularis, Macaca mulatta, Necrosis, T-Lymphocytes immunology, T-Lymphocytes pathology, Tuberculosis pathology, Tuberculosis prevention & control, Tuberculosis immunology
- Abstract
We report that complete protection against malaria and total inhibition of liver stage development and parasitemia was obtained in 100% of BALB/c mice primed with a replication-defective recombinant adenovirus expressing the circumsporozoite (CS) protein of Plasmodium yoelii (AdPyCS), followed by a booster with an attenuated recombinant vaccinia virus, expressing the same malaria antigen, VacPyCS. We found increased levels of activated CS-specific CD8(+) and CD4(+) T cells, higher anti-sporozoite antibody titers, and greater protection in these mice, when the time between priming and boosting with these two viral vectors was extended from 2 to 8 or more weeks. Most importantly, by using this immunization regimen, the protection of the immunized mice was found to be long-lasting, namely complete resistance to infection of all animals 3 1/2 months after priming. These results indicate that immunization with AdPyCS generates highly effective memory T and B cells that can be recalled long after priming by boosting with VacPyCS.
- Published
- 2001
- Full Text
- View/download PDF
23. Gene targeting in the rodent malaria parasite Plasmodium yoelii.
- Author
-
Mota MM, Thathy V, Nussenzweig RS, and Nussenzweig V
- Subjects
- Animals, Antimalarials pharmacology, Gene Deletion, Gene Targeting, Mice, Mice, Inbred BALB C, Multienzyme Complexes drug effects, Multienzyme Complexes genetics, Phenotype, Plasmids genetics, Plasmodium yoelii drug effects, Plasmodium yoelii growth & development, Plasmodium yoelii pathogenicity, Protozoan Proteins genetics, Pyrimethamine pharmacology, Tetrahydrofolate Dehydrogenase drug effects, Tetrahydrofolate Dehydrogenase genetics, Thymidylate Synthase drug effects, Thymidylate Synthase genetics, Malaria parasitology, Plasmodium yoelii genetics, Transfection, Transformation, Genetic
- Abstract
It is anticipated that the sequencing of Plasmodium falciparum genome will soon be completed. Rodent models of malaria infection and stable transformation systems provide powerful means of using this information to study gene function in vivo. To date, gene targeting has only been developed for one rodent malaria species, Plasmodium berghei. Another rodent species, Plasmodium yoelii, however, is favored to study the mechanisms of protective immunity to the pre-erythrocytic stages of infection and vaccine development. In addition, it offers the opportunity to investigate unique aspects of pathogenesis of blood stage infection. Here, we report on the stable transfection and gene targeting of P. yoelii. Purified late blood stage schizonts were used as targets for electroporation with a plasmid that contains a pyrimethamine-resistant form of the P. berghei dihydrofolate reductase-thymidylate synthase (Pbdhfr-ts) fused to green fluorescent protein (gfp) gene. After drug selection, fluorescent parasites contained intact, non-rearranged plasmids that remain stable under drug-pressure. In addition, we used another dhfr-ts/gfp based plasmid to disrupt the P. yoelii trap (thrombospondin-related anonymous protein) locus by site-specific integration. The phenotype of P. yoelii TRAP knockout was identical to that previously reported for the P. berghei TRAP knockout. In the absence of TRAP, the erythrocytic cycle, gametocyte and oocyst development of the mutant parasites were indistinguishable from wild type (WT). Although the sporozoites appeared morphologically normal, they failed to glide and to invade the salivary glands of mosquitoes.
- Published
- 2001
- Full Text
- View/download PDF
24. A striking property of recombinant poxviruses: efficient inducers of in vivo expansion of primed CD8(+) T cells.
- Author
-
Zavala F, Rodrigues M, Rodriguez D, Rodriguez JR, Nussenzweig RS, and Esteban M
- Subjects
- Animals, CD8-Positive T-Lymphocytes virology, Humans, Recombination, Genetic, Vaccines, Attenuated immunology, Virus Replication immunology, CD8-Positive T-Lymphocytes immunology, Genetic Vectors immunology, Poxviridae immunology, Viral Vaccines immunology
- Published
- 2001
- Full Text
- View/download PDF
25. Migration of Plasmodium sporozoites through cells before infection.
- Author
-
Mota MM, Pradel G, Vanderberg JP, Hafalla JC, Frevert U, Nussenzweig RS, Nussenzweig V, and Rodríguez A
- Subjects
- Animals, Cell Line, Cell Membrane parasitology, Cell Membrane physiology, Cell Membrane ultrastructure, Cell Movement, Cytosol metabolism, Cytosol parasitology, Dextrans metabolism, Endocytosis, Flow Cytometry, Fluorescein-5-isothiocyanate metabolism, Hepatocytes ultrastructure, Malaria parasitology, Mice, Mice, Inbred BALB C, Plasmodium physiology, Plasmodium yoelii growth & development, Propidium metabolism, Toxoplasma physiology, Tumor Cells, Cultured, Vacuoles parasitology, Vacuoles ultrastructure, Fluorescein-5-isothiocyanate analogs & derivatives, Hepatocytes parasitology, Plasmodium yoelii physiology
- Abstract
Intracellular bacteria and parasites typically invade host cells through the formation of an internalization vacuole around the invading pathogen. Plasmodium sporozoites, the infective stage of the malaria parasite transmitted by mosquitoes, have an alternative mechanism to enter cells. We observed breaching of the plasma membrane of the host cell followed by rapid repair. This mode of entry did not result in the formation of a vacuole around the sporozoite, and was followed by exit of the parasite from the host cell. Sporozoites traversed the cytosol of several cells before invading a hepatocyte by formation of a parasitophorous vacuole, in which they developed into the next infective stage. Sporozoite migration through several cells in the mammalian host appears to be essential for the completion of the life cycle.
- Published
- 2001
- Full Text
- View/download PDF
26. A totally synthetic polyoxime malaria vaccine containing Plasmodium falciparum B cell and universal T cell epitopes elicits immune responses in volunteers of diverse HLA types.
- Author
-
Nardin EH, Calvo-Calle JM, Oliveira GA, Nussenzweig RS, Schneider M, Tiercy JM, Loutan L, Hochstrasser D, and Rose K
- Subjects
- Adult, Animals, Antibodies, Protozoan biosynthesis, Antibody Specificity, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Line, Female, Humans, Immunoglobulin Isotypes biosynthesis, Interleukin-2 biosynthesis, Kinetics, Lymphocyte Activation, Malaria Vaccines adverse effects, Male, Oximes adverse effects, Vaccines, Synthetic adverse effects, Epitopes, B-Lymphocyte immunology, Epitopes, T-Lymphocyte immunology, HLA-DQ Antigens immunology, HLA-DR Antigens immunology, Malaria Vaccines immunology, Oximes immunology, Plasmodium falciparum immunology, Protozoan Proteins immunology, Vaccines, Synthetic immunology
- Abstract
This open-labeled phase I study provides the first demonstration of the immunogenicity of a precisely defined synthetic polyoxime malaria vaccine in volunteers of diverse HLA types. The polyoxime, designated (T1BT(*))(4)-P3C, was constructed by chemoselective ligation, via oxime bonds, of a tetrabranched core with a peptide module containing B cell epitopes and a universal T cell epitope of the Plasmodium falciparum circumsporozoite protein. The triepitope polyoxime malaria vaccine was immunogenic in the absence of any exogenous adjuvant, using instead a core modified with the lipopeptide P3C as an endogenous adjuvant. This totally synthetic vaccine formulation can be characterized by mass spectroscopy, thus enabling the reproducible production of precisely defined vaccines for human use. The majority of the polyoxime-immunized volunteers (7/10) developed high levels of anti-repeat Abs that reacted with the native circumsporozoite on P. falciparum sporozoites. In addition, these seven volunteers all developed T cells specific for the universal epitope, termed T(*), which was originally defined using CD4(+) T cells from protected volunteers immunized with irradiated P. falciparum sporozoites. The excellent correlation of T(*)-specific cellular responses with high anti-repeat Ab titers suggests that the T(*) epitope functioned as a universal Th cell epitope, as predicted by previous peptide/HLA binding assays and by immunogenicity studies in mice of diverse H-2 haplotypes. The current phase I trial suggests that polyoximes may prove useful for the development of highly immunogenic, multicomponent synthetic vaccines for malaria, as well as for other pathogens.
- Published
- 2001
- Full Text
- View/download PDF
27. Synthetic malaria peptide vaccine elicits high levels of antibodies in vaccinees of defined HLA genotypes.
- Author
-
Nardin EH, Oliveira GA, Calvo-Calle JM, Castro ZR, Nussenzweig RS, Schmeckpeper B, Hall BF, Diggs C, Bodison S, and Edelman R
- Subjects
- Adult, Animals, Cohort Studies, Female, Genotype, HLA-DQ beta-Chains, HLA-DRB1 Chains, Humans, Male, Saponins pharmacology, Antibodies, Protozoan biosynthesis, HLA-DQ Antigens genetics, HLA-DR Antigens genetics, Malaria Vaccines immunology, Plasmodium falciparum immunology, Vaccines, Synthetic immunology
- Abstract
A multiple antigen peptide (MAP) malaria vaccine containing minimal Plasmodium falciparum circumsporozoite protein repeat epitopes was assessed for safety and immunogenicity in volunteers of known class II genotypes. The MAP/alum/QS-21 vaccine formulation elicited high levels of parasite-specific antibodies in 10 of 12 volunteers expressing DQB1*0603, DRB1*0401, or DRB1*1101 class II molecules. In contrast, volunteers of other HLA genotypes were low responders or nonresponders. A second study of 7 volunteers confirmed the correlation of class II genotype and high responder phenotype. This is the first demonstration in humans that a peptide vaccine containing minimal T and B cell epitopes composed of only 5 amino acids (N, A, V, D, and P) can elicit antibody titers comparable to multiple exposures to irradiated P. falciparum-infected mosquitoes. Moreover, the high-responder phenotypes were predicted by analysis of peptide/HLA interactions in vitro, thus facilitating the rational design of epitope-based peptide vaccines for malaria, as well as for other pathogens.
- Published
- 2000
- Full Text
- View/download PDF
28. Immunogenicity of Ty-VLP bearing a CD8(+) T cell epitope of the CS protein of P. yoelii: enhanced memory response by boosting with recombinant vaccinia virus
- Author
-
Oliveira-Ferreira J, Myahira Y, Layton GT, Savage N, Esteban M, Rodriguez D, Rodriguez JR, Nussenzweig RS, and Zavala F
- Published
- 2000
- Full Text
- View/download PDF
29. Interferon-gamma-independent CD8+ T cell-mediated protective anti-malaria immunity elicited by recombinant adenovirus.
- Author
-
Rodrigues EG, Claassen J, Lee S, Wilson JM, Nussenzweig RS, and Tsuji M
- Subjects
- Adenoviridae immunology, Animals, Immunization, Liver parasitology, Mice, Mice, Inbred BALB C, Mice, Knockout, Plasmodium yoelii growth & development, Plasmodium yoelii immunology, Protozoan Proteins administration & dosage, Protozoan Proteins genetics, Recombinant Proteins immunology, Adenoviridae genetics, CD8-Positive T-Lymphocytes immunology, Interferon-gamma immunology, Malaria immunology, Protozoan Proteins immunology
- Abstract
Recombinant adenovirus, expressing the CS protein of Plasmodium yoelii, AdPyCS, was shown to induce a comparable degree of T cell-mediated protection against malaria as a single dose of irradiated P. yoelii sporozoites, causing inhibition of liver stage development. We now report that differently from sporozoite-induced immunity, interferon (IFN)-gamma does not mediate the protective immunity induced by AdPyCS, since a similar degree of protection was observed in AdPyCS immunized mice lacking IFN-gamma-/- and the IFN-gamma receptor (IFN-gammaR-/-) compared to that in wild-type mice. Depletion of CD8+ T cells from these immunized mice almost completely abolished the AdPyCS-induced immunity, indicating that the immunization with AdPyCS induces CD8+ T cell-mediated protective anti-malaria immunity, which is independent of IFN-gamma.
- Published
- 2000
- Full Text
- View/download PDF
30. Pre-erythrocytic malaria vaccine: mechanisms of protective immunity and human vaccine trials.
- Author
-
Nardin E, Zavala F, Nussenzweig V, and Nussenzweig RS
- Subjects
- Animals, Antibodies, Protozoan biosynthesis, CD8-Positive T-Lymphocytes immunology, Clinical Trials as Topic, Humans, Immunity, Cellular, Interferon-gamma metabolism, Mice, Malaria Vaccines, Plasmodium falciparum immunology, Plasmodium vivax immunology
- Abstract
In order to provide a rational basis for the development of a pre-erythrocytic malaria vaccine we have aimed at: (a) elucidating the mechanisms of protection, and (b) identifying vaccine formulations that best elicit protection in experimental animals and humans. Based on earlier successful immunization of experimental animals with irradiated sporozoites, human volunteers were exposed to the bites of large numbers of Plasmodium falciparum or P. vivax infected irradiated mosquitoes. The result of this vaccine trial demonstrated for the first time that a pre-erythrocytic vaccine, administered to humans, can result in their complete resistance to malaria infection. However, since infected irradiated mosquitoes are unavailable for large scale vaccination, the alternative is to develop subunit vaccines. The human trials using irradiated sporozoites provided valuable information on the human immune responses to pre-erythrocytic stages and studies on mice an excellent experimental model to characterize protective immune mechanisms. The circumsporozoite protein, the first pre-erythrocytic antigen identified, is present in all malaria species, displaying a similar structure, with a central region of repeats, and two conserved regions, essential for parasite development. Most pre-erythrocytic vaccine candidates are based on the CS protein, expressed in various cell lines, microorganisms, and recently the corresponding DNA. We and others have identified CS-specific B and T cell epitopes, recognized by the rodent and human immune systems, and used them for the development of synthetic vaccines. We used synthetic peptide vaccines, multiple antigen peptides and polyoximes, for immunization, first in experimental animals, and recently in two human safety and immunogenicity trials. We also report here on our work on T cell mediated immunity, particularly the protection of mice immunized with viral vectors expressing CS-specific cytotoxic CD8+ T cell epitopes, and the striking booster effect of recombinant vaccinia virus. To what degree CD8+ T cells, and/or other T cells specific for sporozoites and/or liver stage epitopes, contribute to pre-erythrocytic protective immunity in humans, remains to be determined.
- Published
- 1999
31. Preclinical evaluation of a synthetic Plasmodium falciparum MAP malaria vaccine in Aotus monkeys and mice.
- Author
-
Moreno CA, Rodriguez R, Oliveira GA, Ferreira V, Nussenzweig RS, Moya Castro ZR, Calvo-Calle JM, and Nardin E
- Subjects
- Alum Compounds administration & dosage, Animals, Aotus trivirgatus, Freund's Adjuvant administration & dosage, Immunization, Malaria Vaccines administration & dosage, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Saponins administration & dosage, Malaria Vaccines immunology, Plasmodium falciparum immunology, Protozoan Proteins immunology, Vaccines, Synthetic immunology
- Abstract
Multiple antigen peptides (MAPs) containing epitopes of the major surface protein of the malaria sporozoite, the circumsporozoite (CS) protein, have been shown in previous studies to elicit antibody-mediated protection against sporozoite challenge in experimental murine and simian hosts. For the preparation for a phase I trial of a P. falciparum (T1B)4 MAP, which contains T and B cell epitopes from the CS repeat region, pre-clinical immunogenicity and adjuvant formulation studies were carried out in mice and Aotus monkeys. The (T1B)4 MAP was found to be immunogenic in three different species of owl monkeys, Aotus nancymae, A. vociferans and A. nigriceps. Optimal antibody responses were obtained in A. nancymae immunized s.c. with (T1B)4 MAP emulsified in Freund's, in which peak titers of over 10(6) were obtained in individual monkeys. MAP immunized A. vociferans also developed high levels of anti-sporozoite antibodies, although the kinetics and the magnitude of the response differed from A. nancymae. (T1B)4 MAP adsorbed to alum (aluminum hydroxide), a formulation that is acceptable for human use, was less immunogenic in naive A. nancymae, as well as A. nigriceps. The injection of MAPs/alum, however, significantly enhanced antibody responses in sporozoite-primed monkeys, suggesting that the administration of the MAP vaccine may be an effective means to increase the low levels of antibody present in individuals living in malaria endemic areas. The addition of a co-adjuvant QS-21, a purified saponin, significantly increased the immunogenicity of the alum-adsorbed MAP in both mice and monkeys, providing a vaccine formulation suitable for phase I trials in human volunteers.
- Published
- 1999
- Full Text
- View/download PDF
32. Interferon-gamma responses are associated with resistance to reinfection with Plasmodium falciparum in young African children.
- Author
-
Luty AJ, Lell B, Schmidt-Ott R, Lehman LG, Luckner D, Greve B, Matousek P, Herbich K, Schmid D, Migot-Nabias F, Deloron P, Nussenzweig RS, and Kremsner PG
- Subjects
- Amino Acid Sequence, Child, Preschool, Female, Humans, Lymphocyte Activation, Male, Molecular Sequence Data, Recurrence, Time Factors, Interferon-gamma biosynthesis, Malaria, Falciparum immunology
- Abstract
The contribution of T cell-mediated responses was studied with regard to resistance to reinfection in groups of Gabonese children participating in a prospective study of severe and mild malaria due to infection with Plasmodium falciparum. In those admitted with mild malaria, but not in those with severe malaria, production of IFN-gamma by peripheral blood mononuclear cells (PBMC) in response to either liver-stage or merozoite antigen peptides was associated with significantly delayed first reinfections and with significantly lower rates of reinfection. Proliferative or tumor necrosis factor responses to the same peptides showed no such associations. Production of interferon-gamma by PBMC in response to sporozoite and merozoite antigen peptides was observed in a higher proportion of those presenting with mild malaria. Differences in the Th1/Th2 cytokine balance may be linked to the ability to control parasite multiplication in these young children, helping to explain the marked differences observed in both susceptibility to infection as well as in clinical presentation.
- Published
- 1999
- Full Text
- View/download PDF
33. Subtle mutagenesis by ends-in recombination in malaria parasites.
- Author
-
Nunes A, Thathy V, Bruderer T, Sultan AA, Nussenzweig RS, and Ménard R
- Subjects
- Animals, Models, Genetic, Plasmids genetics, Gene Targeting methods, Mutagenesis, Insertional, Plasmodium berghei genetics, Protozoan Proteins genetics, Recombination, Genetic
- Abstract
The recent advent of gene-targeting techniques in malaria (Plasmodium) parasites provides the means for introducing subtle mutations into their genome. Here, we used the TRAP gene of Plasmodium berghei as a target to test whether an ends-in strategy, i.e., targeting plasmids of the insertion type, may be suitable for subtle mutagenesis. We analyzed the recombinant loci generated by insertion of linear plasmids containing either base-pair substitutions, insertions, or deletions in their targeting sequence. We show that plasmid integration occurs via a double-strand gap repair mechanism. Although sequence heterologies located close (less than 450 bp) to the initial double-strand break (DSB) were often lost during plasmid integration, mutations located 600 bp and farther from the DSB were frequently maintained in the recombinant loci. The short lengths of gene conversion tracts associated with plasmid integration into TRAP suggests that an ends-in strategy may be widely applicable to modify plasmodial genes and perform structure-function analyses of their important products.
- Published
- 1999
- Full Text
- View/download PDF
34. Efficient induction of protective anti-malaria immunity by recombinant adenovirus.
- Author
-
Rodrigues EG, Zavala F, Nussenzweig RS, Wilson JM, and Tsuji M
- Subjects
- Adenoviridae genetics, Adenoviridae immunology, Adenoviridae metabolism, Amino Acid Sequence, Animals, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Female, Liver parasitology, Lymphocyte Activation immunology, Malaria Vaccines genetics, Malaria Vaccines immunology, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Plasmodium yoelii genetics, Protozoan Proteins biosynthesis, Protozoan Proteins genetics, Malaria immunology, Malaria prevention & control, Malaria Vaccines therapeutic use, Plasmodium yoelii immunology, Protozoan Proteins immunology
- Abstract
The immunogenicity of a previously constructed replication-defective recombinant adenovirus expressing the CS protein of Plasmodium yoelii was compared with that of irradiated sporozoites. We found that immunization of BALB/c mice with a single dose of this recombinant adenovirus induced a much greater CS-specific T-cell response compared with immunization with irradiated sporozoites. More importantly, we found that this recombinant adenovirus induces similar or higher levels of protective immunity than those induced by irradiated sporozoites, eliciting an appreciable resistance to malaria infection.
- Published
- 1998
- Full Text
- View/download PDF
35. Recombinant Sindbis viruses expressing a cytotoxic T-lymphocyte epitope of a malaria parasite or of influenza virus elicit protection against the corresponding pathogen in mice.
- Author
-
Tsuji M, Bergmann CC, Takita-Sonoda Y, Murata K, Rodrigues EG, Nussenzweig RS, and Zavala F
- Subjects
- Animals, Drug Administration Routes, Humans, Influenza A virus growth & development, Influenza Vaccines immunology, Influenza, Human immunology, Mice, Mice, Inbred BALB C, Nucleocapsid Proteins, Nucleoproteins genetics, Nucleoproteins immunology, Plasmodium yoelii genetics, Plasmodium yoelii growth & development, Protozoan Proteins genetics, Protozoan Proteins immunology, Protozoan Vaccines immunology, RNA, Protozoan, Vaccination, Vaccines, Synthetic immunology, Viral Core Proteins genetics, Viral Core Proteins immunology, Epitopes, T-Lymphocyte immunology, Genetic Vectors genetics, Influenza A virus immunology, Influenza, Human prevention & control, Malaria prevention & control, Plasmodium yoelii immunology, RNA-Binding Proteins, Sindbis Virus genetics, T-Lymphocytes, Cytotoxic immunology
- Abstract
Subcutaneous administration in mice of recombinant Sindbis viruses expressing a class I major histocompatibility complex-restricted 9-mer epitope of the Plasmodium yoelii circumsporozoite protein or the nucleoprotein of influenza virus induces a large epitope-specific CD8(+) T-cell response. This immunization also elicits a high degree of protection against infection with malaria or influenza A virus.
- Published
- 1998
- Full Text
- View/download PDF
36. Recombinant viruses expressing a human malaria antigen can elicit potentially protective immune CD8+ responses in mice.
- Author
-
Miyahira Y, García-Sastre A, Rodriguez D, Rodriguez JR, Murata K, Tsuji M, Palese P, Esteban M, Zavala F, and Nussenzweig RS
- Subjects
- Amino Acid Sequence, Animals, Antigens, Protozoan genetics, Gene Expression, Genes, Protozoan, Humans, Immunization, Malaria, Falciparum prevention & control, Mice, Molecular Sequence Data, Orthomyxoviridae genetics, Orthomyxoviridae immunology, Vaccinia virus genetics, Vaccinia virus immunology, Antigens, Protozoan immunology, CD8-Positive T-Lymphocytes immunology, Cytotoxicity, Immunologic, DNA, Recombinant immunology, Malaria, Falciparum immunology, Plasmodium falciparum immunology
- Abstract
Extensive studies on protective immunity to rodent malaria provided the basis for the current experiments in which mice were immunized with recombinant (re) influenza and vaccinia viruses expressing selected sequences of the circumsporozoite (CS) protein of the human malaria parasite Plasmodium falciparum. Mice of different H-2 haplotypes immunized with re influenza viruses expressing the immunodominant B cell epitope of this CS protein produced high titers of antibodies to the parasite. A cytotoxic T lymphocyte epitope of the CS protein of P. falciparum, PF3, recognized by CD8+ T cells of H-2(k) mice, was expressed in a re vaccinia virus (VacPf) and a re influenza virus (FluPf). Immunization of mice with either FluPf or VacPf elicited a modest CS-specific CD8+ T cell response detected by interferon gamma secretion of individual immune cells. Priming of mice with FluPf, followed by a booster with VacPf, resulted in a striking enhancement of this T cell response. The reverse protocol, i.e., priming with VacPf followed by a booster with FluPf, failed to enhance the primary response. VacPf also greatly enhanced the primary response of mice injected with P. falciparum sporozoites or with a lipopeptide containing PF3. A booster with FluPf also amplified the response of lipopeptide- or sporozoite-primed mice but less than a VacPf booster did. Although mice are not susceptible to infection by P. falciparum sporozoites, we demonstrated that administration of two distinct immunogens expressing PF3 elicited activated, extravasating CS-specific T cells that protected against an intracerebral VacPf challenge.
- Published
- 1998
- Full Text
- View/download PDF
37. TRAP is necessary for gliding motility and infectivity of plasmodium sporozoites.
- Author
-
Sultan AA, Thathy V, Frevert U, Robson KJ, Crisanti A, Nussenzweig V, Nussenzweig RS, and Ménard R
- Subjects
- Animals, Anopheles parasitology, Cloning, Molecular, Digestive System parasitology, Digestive System ultrastructure, Erythrocytes parasitology, Movement physiology, Plasmodium berghei ultrastructure, Polymerase Chain Reaction, Protozoan Proteins biosynthesis, Protozoan Proteins genetics, Rats, Recombinant Proteins biosynthesis, Spores, Genes, Protozoan, Liver parasitology, Plasmodium berghei pathogenicity, Plasmodium berghei physiology, Protozoan Proteins physiology, Salivary Glands parasitology
- Abstract
Many protozoans of the phylum Apicomplexa are invasive parasites that exhibit a substrate-dependent gliding motility. Plasmodium (malaria) sporozoites, the stage of the parasite that invades the salivary glands of the mosquito vector and the liver of the vertebrate host, express a surface protein called thrombospondin-related anonymous protein (TRAP) that has homologs in other Apicomplexa. By gene targeting in a rodent Plasmodium, we demonstrate that TRAP is critical for sporozoite infection of the mosquito salivary glands and the rat liver, and is essential for sporozoite gliding motility in vitro. This suggests that in Plasmodium sporozoites, and likely in other Apicomplexa, gliding locomotion and cell invasion have a common molecular basis.
- Published
- 1997
- Full Text
- View/download PDF
38. Development of novel influenza virus vaccines and vectors.
- Author
-
Palese P, Zavala F, Muster T, Nussenzweig RS, and García-Sastre A
- Subjects
- Animals, B-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Genetic Vectors, Humans, Immunity, Mucosal, Mice, Vaccines, Attenuated immunology, Influenza Vaccines immunology, Vaccines, Synthetic immunology
- Abstract
Approaches to improve the efficacy of the current (killed) influenza virus vaccines include the generation of cold-adapted and genetically engineered influenza viruses containing specific attenuating mutations. It is hoped that these genetically altered viruses, in which the hemagglutinin and neuraminidase genes from circulating strains have been incorporated by reassortment, can be used as safe live influenza virus vaccines to induce a long-lasting protective immune response in humans. In addition, genetically engineered influenza viruses may provide a means for expressing foreign antigens. Immunization of mice with recombinant influenza and vaccinia viruses expressing specific antigens of Plasmodium yoelii resulted in a dramatic protective immune response against malaria in this model. Mice immunized with recombinant influenza viruses expressing human immunodeficiency virus (HIV) epitopes generated long-lasting HIV-specific serum antibodies and secretory IgA in the secretory nasal, vaginal, and intestinal mucosa. These results suggest that genetically engineered influenza viruses may be developed for use as live virus vaccines against influenza as well as other diseases.
- Published
- 1997
- Full Text
- View/download PDF
39. Circumsporozoite protein is required for development of malaria sporozoites in mosquitoes.
- Author
-
Ménard R, Sultan AA, Cortes C, Altszuler R, van Dijk MR, Janse CJ, Waters AP, Nussenzweig RS, and Nussenzweig V
- Subjects
- Animals, Anopheles parasitology, Gene Targeting, Insecticide Resistance genetics, Multienzyme Complexes genetics, Plasmids, Plasmodium berghei genetics, Plasmodium berghei physiology, Protozoan Proteins genetics, Rats, Salivary Glands parasitology, Tetrahydrofolate Dehydrogenase genetics, Thymidylate Synthase genetics, Plasmodium berghei growth & development, Protozoan Proteins physiology
- Abstract
Malaria parasites undergo a sporogonic cycle in the mosquito vector. Sporozoites, the form of the parasite injected into the host during a bloodmeal, develop inside oocysts in the insect midgut, then migrate to and eventually invade the salivary glands. The circumsporozoite protein (CS), one of the major proteins synthesized by salivary gland sporozoites, is a surface-associated molecule which is important in sporozoite infectivity to the host. Here, by gene targeting, we created Plasmodium berghei lines in which the single-copy CS gene was disrupted. The CS(-) and wild-type parasites produced similar numbers of oocysts of comparable size in the mosquito midgut. In the CS(-) oocysts, however, sporozoite formation was profoundly inhibited. CS therefore appears to have a pleiotropic role and to be vital for malaria parasites in both the vector and the host: in mosquitoes, CS is essential for sporozoite development within oocysts, and in the vertebrate host it promotes sporozoite attachment to hepatocytes.
- Published
- 1997
- Full Text
- View/download PDF
40. A malaria vaccine based on a sporozoite antigen.
- Author
-
Nussenzweig RS and Zavala F
- Subjects
- Adjuvants, Immunologic, Animals, Antibodies, Protozoan blood, Humans, Plasmodium falciparum growth & development, Antigens, Protozoan immunology, Malaria Vaccines immunology, Malaria, Falciparum prevention & control, Protozoan Proteins immunology
- Published
- 1997
- Full Text
- View/download PDF
41. Plasmodium yoelii: peptide immunization induces protective CD4+ T cells against a previously unrecognized cryptic epitope of the circumsporozoite protein.
- Author
-
Takita-Sonoda Y, Tsuji M, Kamboj K, Nussenzweig RS, Clavijo P, and Zavala F
- Subjects
- Adoptive Transfer, Amino Acid Sequence, Animals, Female, Immunization, Immunodominant Epitopes, Interferon-gamma metabolism, Interleukin-5 metabolism, Malaria parasitology, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Plasmodium yoelii growth & development, Th1 Cells immunology, Antigens, Protozoan immunology, CD4-Positive T-Lymphocytes immunology, Malaria immunology, Peptides immunology, Plasmodium yoelii immunology, Protozoan Proteins immunology
- Abstract
In this study we characterized the CD4+ T cell response directed against two distinct epitopes located in the circumsporozoite (CS) protein of Plasmodium yoelii. The immunization of mice with P. yoelii sporozoites induced CD4+ T cells which were mostly directed against one of these peptides, Py-1, previously reported to contain a CD4+ epitope. The CD4+ T cells directed against this immunodominant epitope were mostly of the Th-1 type. Another newly identified peptide, AS44, induced a specific CD4+ T cell response, which was mainly detectable after immunization with the corresponding peptide. Several CD4+ T cell clones, recognizing this epitope, were generated and their lymphokine expression was characterized, as well as their surface markers and their anti-parasite activity in vivo. It was noteworthy that some of these CD4+ T cell clones, which recognize this cryptic epitope and were of different Th subtypes, were shown to have a strong inhibitory effect on the development of liver stages of malaria parasites.
- Published
- 1996
- Full Text
- View/download PDF
42. Characterization of in vivo primary and secondary CD8+ T cell responses induced by recombinant influenza and vaccinia viruses.
- Author
-
Murata K, García-Sastre A, Tsuji M, Rodrigues M, Rodriguez D, Rodriguez JR, Nussenzweig RS, Palese P, Esteban M, and Zavala F
- Subjects
- Animals, Antigens, Protozoan genetics, Epitopes immunology, Female, Genetic Vectors genetics, Genetic Vectors immunology, Humans, Influenza A virus genetics, Male, Mice, Mice, Inbred A, Mice, Inbred BALB C, Nucleocapsid Proteins, Plasmodium yoelii immunology, Protozoan Proteins genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Vaccinia virus genetics, Vaccinia virus immunology, Viral Core Proteins genetics, Antigens, Protozoan immunology, Antigens, Viral immunology, CD8-Positive T-Lymphocytes immunology, H-2 Antigens immunology, Influenza A virus immunology, Nucleoproteins, Protozoan Proteins immunology, Viral Core Proteins immunology
- Abstract
We characterized the in vivo primary and secondary murine CD8+ T cell responses induced by immunization with influenza and vaccinia viruses, which were engineered to express the same H-2K(k)- and H-2K(d)-restricted epitopes. Our results show that the induction and magnitude of the primary CD8+ T cell response closely depends on the viral dose used for immunization, while it is not affected by the route of immunization. The induction of secondary CD8+ T cell responses appears to be highly restricted, as suggested by the lack of in vivo expansion of antigen-specific CD8+ T cells after repeated immunization with the same virus. In contrast, a 20- to 30-fold increase in the frequency of antigen-specific CD8+ T cells could be induced after combined immunization with recombinant influenza and vaccinia viruses. These findings may provide the basis for the development of new prophylactic and therapeutic strategies to prevent or control intracellular infections and certain malignancies.
- Published
- 1996
- Full Text
- View/download PDF
43. Phenotypic and functional properties of murine gamma delta T cell clones derived from malaria immunized, alpha beta T cell-deficient mice.
- Author
-
Tsuji M, Eyster CL, O'Brien RL, Born WK, Bapna M, Reichel M, Nussenzweig RS, and Zavala F
- Subjects
- Animals, Antigens, Surface biosynthesis, Clone Cells, Cytokines genetics, Immunization, Immunotherapy, Adoptive, Lymphocyte Activation, Mice, Mice, Inbred BALB C, Mice, Inbred C3H, Mice, Inbred C57BL, Plasmodium yoelii immunology, RNA, Messenger biosynthesis, RNA, Messenger immunology, Receptors, Antigen, T-Cell, alpha-beta immunology, Receptors, Antigen, T-Cell, gamma-delta biosynthesis, Receptors, Antigen, T-Cell, gamma-delta isolation & purification, T-Lymphocytes metabolism, Malaria immunology, Receptors, Antigen, T-Cell, gamma-delta immunology, T-Lymphocytes immunology
- Abstract
Six murine T cell clones expressing gamma delta TCR were generated from malaria immunized, alpha beta T cell-deficient mice. Phenotypic characterization of these clones has revealed that, in contrast to conventional alpha beta T cells, there is a considerable degree of heterogeneity among these gamma delta clones with regard to their surface markers and their lymphokine profile. One clone was found to display significant anti-parasite activity in vivo upon adoptive transfer. We attempted to determine whether the protective clone differs in one or more key characteristics from the non-protective clones. Although no obvious pattern peculiar to the protective gamma delta clone was observed, it appears that more than one parameter may, in combination, define a distinct protective phenotype, and thus explain the functional difference between the protective and non-protective gamma delta clones.
- Published
- 1996
- Full Text
- View/download PDF
44. Detection of anti-Plasmodium falciparum antibodies directed against a repetitive peptide of the gametocyte antigen Pfs2400 in malaria patients in Brazil.
- Author
-
Marrelli MT, Nussenzweig RS, Collins WE, and Kloetzel JK
- Subjects
- Adolescent, Adult, Aged, Antigens, Protozoan blood, Brazil, Enzyme-Linked Immunosorbent Assay, Fluorescent Antibody Technique, Indirect, Humans, Middle Aged, Protozoan Proteins blood, Recurrence, Antibodies, Protozoan blood, Antigens, Protozoan immunology, Malaria, Falciparum immunology, Protozoan Proteins immunology
- Abstract
Sera collected from 164 individuals who had clinical Plasmodium falciparum malaria and came from several areas of Brazil where malaria is endemic were tested for the presence of anti-gametocyte antibodies. Antibodies directed against P. falciparum gametocytes were detected, by IFAT, in the sera of 67.1% of these patients. The prevalence of these antibodies was significantly higher in patients who had undergone multiple attacks of malaria than in those who were experiencing their first attack at the time of serum collection. Although circulating gametocytes were detected in 22% of the patients at this time, there was no difference in the percentages of IFAT positivity between apparent gametocyte 'carriers' and 'non-carriers'. All sera were also tested by ELISA, using a dimer of the nonamer peptide [PEE(L/V)VEEV(I/V)]2, which represents a tandem consensus repeat of the P. falciparum gametocyte antigen, Pfs2400, a target of transmission-blocking antibodies. ELISA demonstrated that 32.9% of the patients had antibodies that reacted with this peptide. Positive ELISA reactions were significantly more frequent amongst the sera of patients who had had multiple malaria attacks than in those undergoing their first malaria episode; positivity was lower in the gametocyte 'carriers' than in their 'non-carriers'. These results demonstrate that anti-gametocyte antibodies, which have already been shown to have potential transmission-blocking activity, are naturally elicited in Brazilian patients, the highest rates of seropositivity occurring after multiple malaria attacks.
- Published
- 1995
- Full Text
- View/download PDF
45. Development of antimalaria immunity in mice lacking IFN-gamma receptor.
- Author
-
Tsuji M, Miyahira Y, Nussenzweig RS, Aguet M, Reichel M, and Zavala F
- Subjects
- Amino Acid Oxidoreductases biosynthesis, Animals, Fluorescent Antibody Technique, Mice, Mice, Mutant Strains, Mutation genetics, Nitric Oxide Synthase, Polymerase Chain Reaction, RNA, Messenger biosynthesis, RNA, Protozoan analysis, RNA, Ribosomal analysis, Interferon gamma Receptor, Malaria immunology, Plasmodium chabaudi immunology, Plasmodium yoelii immunology, Receptors, Interferon genetics, Receptors, Interferon immunology
- Abstract
IFN-gamma receptor deficient (IFN-gamma R-/-) mice, immunized with different developmental stages of malaria parasites, were used to define the mechanisms of protection against the various stages of this infection. IFN-gamma R-/- mice failed to develop protective immunity against Plasmodium yoelii sporozoites or liver stages, upon immunization with a single dose of irradiated sporozoites, whereas in immunized wild-type mice, parasite development was strongly inhibited. Immunized wild-type mice expressed high levels of inducible nitric oxide synthase (iNOS) mRNA in their liver, upon challenge with viable sporozoites, whereas only background levels of iNOS were detected in immunized IFN-gamma R-/- mice. In contrast, after immunization with multiple doses of irradiated sporozoites, both IFN-gamma R-/- and wild-type mice mounted an immune response, which strongly inhibited the development of liver stage parasites. In both types of mice, protection occurred in the absence of appreciable expression of liver iNOS mRNA. As for the course of the erythrocytic phase of infection by nonlethal malaria species, P. yoelii yoelii and P. chabaudi adami, we observed only a moderately prolonged parasitemia in IFN-gamma R-/- mice compared with wild-type mice, indicating that IFN-gamma may only play a modest role in immunity against erythrocytic stages. These results indicate that IFN-gamma is the main mediator of the protective mechanism that develops first upon immunization with sporozoites. However, the nature of the anti-parasite mechanism(s) changes in the course of immunization, so that multiple immunizing doses elicit additional protective mechanisms, which are independent of IFN-gamma and its receptor.
- Published
- 1995
46. The use of multiple antigen peptides in the analysis and induction of protective immune responses against infectious diseases.
- Author
-
Nardin EH, Oliveira GA, Calvo-Calle JM, and Nussenzweig RS
- Subjects
- Amino Acid Sequence, Animals, Antigens, Helminth, Bacterial Infections prevention & control, Epitopes immunology, Epitopes, B-Lymphocyte immunology, Epitopes, T-Lymphocyte immunology, Helminthiasis prevention & control, Humans, Immunodominant Epitopes immunology, Malaria Vaccines, Mice, Molecular Sequence Data, Protozoan Infections prevention & control, Virus Diseases prevention & control, Antigens immunology, Peptides immunology, Vaccines, Synthetic immunology
- Published
- 1995
- Full Text
- View/download PDF
47. Influenza and vaccinia viruses expressing malaria CD8+ T and B cell epitopes. Comparison of their immunogenicity and capacity to induce protective immunity.
- Author
-
Rodrigues M, Li S, Murata K, Rodriguez D, Rodriguez JR, Bacik I, Bennink JR, Yewdell JW, Garcia-Sastre A, and Nussenzweig RS
- Subjects
- Amino Acid Sequence, Animals, Antigens, Protozoan genetics, CD4-Positive T-Lymphocytes immunology, Cytotoxicity, Immunologic, Enzyme-Linked Immunosorbent Assay methods, Epitopes immunology, Female, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Protozoan Proteins immunology, Recombinant Proteins immunology, Antigens, Protozoan immunology, B-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Orthomyxoviridae genetics, Plasmodium yoelii immunology, Vaccinia virus genetics
- Abstract
We compared the effectiveness of several recombinant influenza and vaccinia viruses to induce a malaria-specific immune response. The CD8+ T cell epitope of the circumsporozoite (CS) protein of Plasmodium yoelii, a rodent malaria parasite, was expressed in two distinct influenza virus proteins, the hemagglutinin and the neuraminidase. These recombinant viruses were found to be equally efficient at inducing CS-specific CD8+ T cells in mice. A third recombinant virus, which expresses a B cell epitope of the CS protein, induced neutralizing anti-sporozoite Abs. Expression in the same recombinant virus of the CD8+ T cell epitope and of the B cell epitope did not impair the capacity of this recombinant virus to induce malaria-specific CD8+ T cells and neutralizing Abs. The immunogenicity of a vaccinia virus, expressing the entire CS protein, was compared with that of a highly attenuated vaccinia strain expressing the same protein and with that of another vaccinia virus expressing only the CD8+ T cell epitope. All three vaccinia virus recombinants elicited CS-specific CD8+ cells and a potent inhibitory response against pre-erythrocytic stages of malaria parasites. Optimal levels of anti-sporozoite Abs, inhibition of liver stage development, and protection against malaria infection resulted from repeatedly immunizing the animals with recombinant influenza viruses followed by boosters with a recombinant vaccinia virus. These findings support the concept that live viral vectors expressing the appropriate proteins and/or epitopes can be used as promising vaccine candidates.
- Published
- 1994
48. Malaria vaccines: multiple targets.
- Author
-
Nussenzweig RS and Long CA
- Subjects
- Animals, Anopheles parasitology, Antigens, Protozoan immunology, Humans, Insect Vectors parasitology, Malaria transmission, Malaria, Falciparum prevention & control, Malaria, Falciparum transmission, Plasmodium growth & development, Plasmodium immunology, Plasmodium falciparum growth & development, Plasmodium falciparum immunology, T-Lymphocytes, Cytotoxic immunology, Malaria prevention & control, Malaria Vaccines immunology
- Published
- 1994
- Full Text
- View/download PDF
49. Immunogenicity of an alum-adsorbed synthetic multiple-antigen peptide based on B- and T-cell epitopes of the Plasmodium falciparum CS protein: possible vaccine application.
- Author
-
de Oliveira GA, Clavijo P, Nussenzweig RS, and Nardin EH
- Subjects
- Adsorption, Amino Acid Sequence, Animals, Antigens, Protozoan chemistry, B-Lymphocytes immunology, Enzyme-Linked Immunosorbent Assay, Epitopes immunology, Female, Fluorescent Antibody Technique, Malaria Vaccines chemistry, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Protozoan Proteins chemistry, T-Lymphocytes, Helper-Inducer immunology, Adjuvants, Immunologic chemistry, Alum Compounds chemistry, Antigens, Protozoan immunology, Malaria Vaccines immunology, Plasmodium falciparum immunology, Protozoan Proteins immunology
- Abstract
Multiple-antigen peptides (MAPs), containing B- and T-cell epitopes of the Plasmodium falciparum circumsporozoite (CS) protein, have been designed to overcome the limitations of first-generation peptide vaccines caused by low epitope density, carrier toxicity and the lack of parasite-derived T-cell epitopes. The immunogenicity of a P. falciparum MAP construct (T1B4), containing four copies of the 5' repeat cell T epitope (T1) combined with the 3' repeat epitope (NANP)3, has been examined using different adjuvant formulations. Mice immunized intraperitoneally or subcutaneously with (T1B)4 in alum, a formulation suitable for human vaccines, developed high anti-peptide and anti-sporozoite antibody titres, comparable with those obtained with Freund's adjuvant. The MAP/alum formulation also elicited a strong anamnestic antibody response in sporozoite-primed mice, raising the possibility of using a MAP/alum vaccine to increase the low anti-sporozoite antibody levels of people living in malaria-endemic areas.
- Published
- 1994
- Full Text
- View/download PDF
50. Gamma delta T cells contribute to immunity against the liver stages of malaria in alpha beta T-cell-deficient mice.
- Author
-
Tsuji M, Mombaerts P, Lefrancois L, Nussenzweig RS, Zavala F, and Tonegawa S
- Subjects
- Animals, Immunization, Passive, Liver parasitology, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, gamma-delta genetics, Malaria immunology, T-Lymphocyte Subsets immunology
- Abstract
The functional role of gamma delta T cells (expressing the gamma delta heterodimeric T-cell receptor for antigen) in infectious diseases remains largely unknown. We have therefore attempted to define the possible role of these T cells in the immune response against the various developmental stages of malaria parasites. For this purpose, we monitored the immune response and the development of liver and blood stages of Plasmodium yoelii, a rodent malaria parasite, in immunized and nonimmunized alpha beta T-cell-deficient and gamma delta T-cell-deficient mice. Immunization of alpha beta T-cell-deficient mice with irradiated sporozoites induced an immune response that significantly inhibited the development of the parasite's liver stages. This inhibitory immune response was abolished by an antibody-mediated transient in vivo depletion of gamma delta T cells. Two gamma delta T-cell clones were derived from malaria-immunized alpha beta T-cell-deficient mice. The adoptive transfer of one of these gamma delta T-cell clones to normal mice inhibited the development of liver stages, following sporozoite inoculation. These results provide evidence for gamma delta T-cell-mediated protective immunity against parasites, in the absence of alpha beta T cells. As for the blood phase of the infection, both normal mice and gamma delta T-cell-deficient mice cleared the blood stages of the nonlethal strain of P. yoelii, while alpha beta T-cell-deficient mice failed to control the parasitemia.
- Published
- 1994
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.