Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
J Biol Chem ; 286(30): 26396-405, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21652718

ABSTRACT

Invasion of hepatocytes by Plasmodium sporozoites deposited by Anopheles mosquitoes, and their subsequent transformation into infective merozoites is an obligatory step in the initiation of malaria. Interactions between the sporozoites and hepatocytes lead to a distinct, complex and coordinated cellular and systemic host response. Little is known about host liver cell response to sporozoite invasion, or whether it is primarily adaptive for the parasite, for the host, or for both. Our present study used gene expression profiling of human HepG2-A16 liver cells infected with Plasmodium falciparum sporozoites to understand the host early cellular events and factors influencing parasite infectivity and sporozoite development. Our results show that as early as 30 min following wild-type, non-irradiated sporozoite exposure, the expressions of at least 742 genes was selectively altered. These genes regulate diverse biological functions, such as immune processes, cell adhesion and communications, metabolism pathways, cell cycle regulation, and signal transduction. These functions reflect cellular events consistent with initial host cell defense responses, as well as alterations in host cells to sustain sporozoites growth and survival. Irradiated sporozoites gave very similar gene expression pattern changes, but direct comparative analysis between liver gene expression profiles caused by irradiated and non-irradiated sporozoites identified 29 genes, including glypican-3, that were specifically up-regulated only in irradiated sporozoites. Elucidating the role of this subset of genes may help identify the molecular basis for the irradiated sporozoites inability to develop intrahepatically, and their usefulness as an immunogen for developing protective immunity against pre-erythrocytic stage malaria.


Subject(s)
Gene Expression Regulation , Hepatocytes/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum , Sporozoites , Transcription, Genetic , Gene Expression Profiling , Hep G2 Cells , Hepatocytes/parasitology , Humans
3.
PLoS One ; 6(4): e18393, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21533224

ABSTRACT

BACKGROUND: The malaria parasite Plasmodium falciparum EBA-175 binds its receptor sialic acids on glycophorin A when invading erythrocytes. The receptor-binding region (RII) contains two cysteine-rich domains with similar cysteine motifs (F1 and F2). Functional relationships between F1 and F2 domains and characterization of EBA-175 were studied using specific monoclonal antibodies (mAbs) against these domains. METHODS AND FINDINGS: Five mAbs specific for F1 or F2 were generated. Three mAbs specific for F2 potently blocked binding of EBA-175 to erythrocytes, and merozoite invasion of erythrocytes (IC(50) 10 to 100 µg/ml IgG in growth inhibition assays). A mAb specific for F1 blocked EBA-175 binding and merozoite invasion less effectively. The difference observed between the IC(50) of F1 and F2 mAbs was not due to differing association and disassociation rates as determined by surface plasmon resonance. Four of the mAbs recognized conformation-dependent epitopes within F1 or F2. Used in combination, F1 and F2 mAbs blocked the binding of native EBA-175 to erythrocytes and inhibited parasite invasion synergistically in vitro. MAb R217, the most potent, did not recognize sporozoites, 3-day hepatocyte stage parasites, nor rings, trophozoites, gametocytes, retorts, ookinetes, and oocysts but recognized 6-day hepatocyte stage parasites, and schizonts. Even though efficient at blocking binding to erythrocytes and inhibiting invasion into erythrocytes, MAb R217 did not inhibit sporozoite invasion and development in hepatocytes in vitro. CONCLUSIONS: The role of the F1 and F2 domains in erythrocyte invasion and binding was elucidated with mAbs. These mAbs interfere with native EBA-175 binding to erythrocyte in a synergistic fashion. The stage specific expression of EBA-175 showed that the primary focus of activity was the merozoite stage. A recombinant RII protein vaccine consisting of both F1 and F2 domains that could induce synergistic activity should be optimal for induction of antibody responses that interfere with merozoite invasion of erythrocytes.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Protozoan/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Antigens, Protozoan/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/physiology , Protozoan Proteins/immunology
4.
Transfusion ; 51(3): 630-5, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20849405

ABSTRACT

BACKGROUND: Transfusion-transmitted malaria remains a serious concern for blood safety. Viable Plasmodium parasites must be present in human blood to transmit malaria, but their survival in blood over time stored under refrigeration has never been carefully investigated. STUDY DESIGN AND METHODS: We spiked leukoreduced normal human blood with Plasmodium falciparum (3D7 strain) asexual ring-stage parasites and stored it at 4 °C for 28 days, taking samples at different days intervals. We evaluated the samples for parasitemia by blood film microscopy and by culturing red blood cells (RBCs) to allow further development of parasites. RESULTS: We observed a significant reduction in parasitemia (0.5% vs. 0.12%) after only 1 day in storage at 4 °C. Thereafter, reduction in parasitemia was relatively gradual. Microscopically detectable parasites were present even after 28 days of storage. However, after storing for more than 14 days at 4 °C, parasites no longer replicated when cultured in vitro. CONCLUSION: Although the storage of asexual blood-stage P. falciparum parasites at 4 °C is detrimental to their survival (a 7.1-fold reduction in parasitemia after 14 days in storage), parasites remained microscopically detectable for 28 days, the end time point of our study. Further in vitro and in vivo studies will be needed to confirm loss of viability of P. falciparum after 14 days in storage, but our initial efforts repeatedly failed to show maturation and development of the parasites in cultured RBCs after that time.


Subject(s)
Parasitemia/parasitology , Plasmodium falciparum/physiology , Blood Preservation , Humans , Refrigeration
5.
PLoS One ; 5(12): e14275, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-21151554

ABSTRACT

Plasmodium vivax (Pv) is the second most important human malaria parasite. Recent data indicate that the impact of Pv malaria on the health and economies of the developing world has been dramatically underestimated. Pv has a unique feature in its life cycle. Uninucleate sporozoites (spz), after invasion of human hepatocytes, either proceed to develop into tens of thousands of merozoites within the infected hepatocytes or remain as dormant forms called hypnozoites, which cause relapses of malaria months to several years after the primary infection. Elimination of malaria caused by Pv will be facilitated by developing a safe, highly effective drug that eliminates Pv liver stages, including hypnozoites. Identification and development of such a drug would be facilitated by the development of a medium to high throughput assay for screening drugs against Pv liver stages. We undertook the present pilot study to (1) assess the feasibility of producing large quantities of purified, vialed, cryopreserved Pv sporozoites and (2) establish a system for culturing the liver stages of Pv in order to assess the effects of drugs on the liver stages of Pv. We used primaquine (PQ) to establish this assay model, because PQ is the only licensed drug known to clear all Pv hepatocyte stages, including hypnozoites, and the effect of PQ on Pv hepatocyte stage development in vitro has not previously been reported. We report that we have established the capacity to reproducibly infect hepatoma cells with purified, cyropreserved Pv spz from the same lot, quantitate the primary outcome variable of infected hepatoma cells and demonstrate the inhibitory activity of primaquine on the infected hepatoma cells. We have also identified small parasite forms that may be hypnozoites. These data provide the foundation for finalizing a medium throughput, high content assay to identify new drugs for the elimination of all Pv liver stages.


Subject(s)
Liver/drug effects , Liver/metabolism , Malaria, Vivax/metabolism , Animals , Anopheles/parasitology , Antimalarials/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/parasitology , Cryopreservation , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/parasitology , Humans , In Vitro Techniques , Microscopy, Fluorescence/methods , Pan troglodytes , Plasmodium vivax/metabolism , Primaquine/pharmacology , Salivary Glands/metabolism , Sporozoites/chemistry
6.
Infect Immun ; 78(11): 4613-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20823210

ABSTRACT

The multiple antigen peptide (MAP) approach is an effective method to chemically synthesize and deliver multiple T-cell and B-cell epitopes as the constituents of a single immunogen. Here we report on the design, chemical synthesis, and immunogenicity of three Plasmodium falciparum MAP vaccines that incorporated antigenic epitopes from the sporozoite, liver, and blood stages of the life cycle. Antibody and cellular responses were determined in three inbred (C57BL/6, BALB/c, and A/J) strains, one congenic (HLA-A2 on the C57BL/6 background) strain, and one outbred strain (CD1) of mice. All three MAPs were immunogenic and induced both antibody and cellular responses, albeit in a somewhat genetically restricted manner. Antibodies against MAP-1, MAP-2, and MAP-3 had an antiparasite effect that was also dependent on the mouse major histocompatibility complex background. Anti-MAP-1 (CSP-based) antibodies blocked the invasion of HepG2 liver cells by P. falciparum sporozoites (highest, 95.16% in HLA-A2 C57BL/6; lowest, 11.21% in BALB/c). Furthermore, antibodies generated following immunizations with the MAP-2 (PfCSP, PfLSA-1, PfMSP-1(42), and PfMSP-3b) and MAP-3 (PfRAP-1, PfRAP-2, PfSERA, and PfMSP-1(42)) vaccines were able to reduce the growth of blood stage parasites in erythrocyte cultures to various degrees. Thus, MAP-based vaccines remain a viable option to induce effective antibody and cellular responses. These results warrant further development and preclinical and clinical testing of the next generation of candidate MAP vaccines that are based on the conserved protective epitopes from Plasmodium antigens that are widely recognized by populations of divergent HLA types from around the world.


Subject(s)
Antigens, Protozoan , Malaria Vaccines , Malaria, Falciparum/prevention & control , Vaccines, Subunit , Vaccines, Synthetic , Amino Acid Sequence , Animals , Animals, Outbred Strains , Antibodies, Protozoan/blood , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Cell Line , Cell Line, Tumor , Cells, Cultured , Drug Design , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Erythrocytes/parasitology , Female , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Hepatocytes/parasitology , Humans , Immunization , Malaria Vaccines/administration & dosage , Malaria Vaccines/chemical synthesis , Malaria Vaccines/chemistry , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Mice , Mice, Congenic , Mice, Inbred Strains , Molecular Sequence Data , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , T-Lymphocytes/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemical synthesis , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
7.
PLoS Negl Trop Dis ; 4(4): e653, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20386602

ABSTRACT

BACKGROUND: Up to 40% of the world's population is at risk for Plasmodium vivax malaria, a disease that imposes a major public health and economic burden on endemic countries. Because P. vivax produces latent liver forms, eradication of P. vivax malaria is more challenging than it is for P. falciparum. Genetic analysis of P. vivax is exceptionally difficult due to limitations of in vitro culture. To overcome the barriers to traditional molecular biology in P. vivax, we examined parasite transcriptional changes in samples from infected patients and mosquitoes in order to characterize gene function, define regulatory sequences and reveal new potential vaccine candidate genes. PRINCIPAL FINDINGS: We observed dramatic changes in transcript levels for various genes at different lifecycle stages, indicating that development is partially regulated through modulation of mRNA levels. Our data show that genes involved in common biological processes or molecular machinery are co-expressed. We identified DNA sequence motifs upstream of co-expressed genes that are conserved across Plasmodium species that are likely binding sites of proteins that regulate stage-specific transcription. Despite their capacity to form hypnozoites we found that P. vivax sporozoites show stage-specific expression of the same genes needed for hepatocyte invasion and liver stage development in other Plasmodium species. We show that many of the predicted exported proteins and members of multigene families show highly coordinated transcription as well. CONCLUSIONS: We conclude that high-quality gene expression data can be readily obtained directly from patient samples and that many of the same uncharacterized genes that are upregulated in different P. vivax lifecycle stages are also upregulated in similar stages in other Plasmodium species. We also provide numerous examples of how systems biology is a powerful method for determining the likely function of genes in pathogens that are neglected due to experimental intractability.


Subject(s)
Culicidae/parasitology , Gene Expression Profiling , Life Cycle Stages , Malaria, Vivax/parasitology , Plasmodium vivax/genetics , Plasmodium vivax/pathogenicity , Animals , Humans
8.
PLoS One ; 5(1): e8947, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-20126610

ABSTRACT

It was recently reported that when mosquitoes infected with P. berghei sporozoites feed on mice, they deposit approximately 100-300 sporozoites in the dermis. When we inoculate P. yoelii (Py) sporozoites intravenously (IV) into BALB/c mice, the 50% infectious dose (ID(50)) is often less than 3 sporozoites, indicating that essentially all Py sporozoites in salivary glands are infectious. Thus, it should only take the bite of one infected mosquito to infect 100% of mice. In human subjects, it takes the bite of at least 5 P. falciparum-infected mosquitoes to achieve 100% blood stage infection. Exposure to 1-2 infected mosquitoes only leads to blood stage infection in approximately 50% of subjects. If mosquitoes carrying Py sporozoites inoculate 100-300 sporozoites per bite, and 1 to 2 mosquito bites achieve 50% blood stage infection rates, then this would suggest that the majority of sporozoites inoculated by mosquitoes into the dermis are not responsible for a productive infection, or that a significant number of sporozoite-infected mosquitoes do not inoculate any sporozoites. The objective of this study was to determine if this is the case. We therefore studied the infectivity to mice of the bites of 1, 2, 4, or 5-8 Py-infected mosquitoes. The bite of one Py sporozoite-infected mosquito caused blood stage infection in 41.4% (12/29) of mice, two bites infected 66.7% (22/33), four bites infected 75% (18/24), and five to eight bites infected 100% (21/21). These findings demonstrate that inoculation of sporozoites by mosquito bite is much less efficient than IV inoculation of Py sporozoites by needle and syringe. Such data may have implications for determining the best route and dose of administration to humans of our attenuated P. falciparum sporozoite vaccine, the scientific basis of which is immunity by bites from irradiated infected mosquitoes, and suggest that the challenge is to develop a method of administration that approximates IV inoculation, not one that mimics mosquito bite.


Subject(s)
Anopheles/parasitology , Malaria/transmission , Plasmodium yoelii/physiology , Animals , Malaria/parasitology , Mice , Mice, Inbred BALB C
9.
Hum Vaccin ; 6(1): 97-106, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19946222

ABSTRACT

Immunization of volunteers by the bite of mosquitoes carrying radiation-attenuated Plasmodium falciparum sporozoites protects greater than 90% of such volunteers against malaria, if adequate numbers of immunizing biting sessions and sporozoite-infected mosquitoes are used. Nonetheless, until recently it was considered impossible to develop, license and commercialize a live, whole parasite P. falciparum sporozoite (PfSPZ) vaccine. In 2003 Sanaria scientists reappraised the potential impact of a metabolically active, non-replicating PfSPZ vaccine, and outlined the challenges to producing such a vaccine. Six years later, significant progress has been made in overcoming these challenges. This progress has enabled the manufacture and release of multiple clinical lots of a 1(st) generation metabolically active, non-replicating PfSPZ vaccine, the Sanaria PfSPZ Vaccine, submission of a successful Investigational New Drug application to the US Food and Drug Administration, and initiation of safety, immunogenicity and protective efficacy studies in volunteers in MD, US. Efforts are now focused on how best to achieve submission of a successful Biologics License Application and introduce the vaccine to the primary target population of African children in the shortest possible period of time. This will require implementation of a systematic, efficient clinical development plan. Short term challenges include optimizing the (1) efficiency and scale up of the manufacturing process and quality control assays, (2) dosage regimen and method of administration, (3) potency of the vaccine, and (4) logistics of delivering the vaccine to those who need it most, and finalizing the methods for vaccine stabilization and attenuation. A medium term goal is to design and build a facility for manufacturing highly potent and stable vaccine for pivotal Phase 3 studies and commercial launch.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Sporozoites/immunology , Clinical Trials as Topic , Drug Approval , Humans , United States , Vaccines, Attenuated/immunology
10.
Indian J Exp Biol ; 47(7): 527-36, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19761036

ABSTRACT

Development of an effective malaria vaccine remains one of the biggest challenges faced by modern science. Although in the last decade tremendous advances have taken place in the design, construction and testing of malaria vaccines, many questions still remained unanswered. This review highlights exclusively some of the exciting and most recent progress in the development and clinical testing of candidate malaria vaccines and points out some of the outstanding scientific issues and technological challenges that must be met to develop a successful vaccine.


Subject(s)
Malaria Vaccines/isolation & purification , Malaria Vaccines/pharmacology , Animals , Humans , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/economics , Plasmodium/growth & development , Plasmodium/immunology , Research/economics , Research/trends
11.
Vaccine ; 27(27): 3675-80, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19071177

ABSTRACT

We are developing a radiation attenuated Plasmodium falciparum sporozoite (PfSPZ) malaria vaccine. An important step was to determine the minimum dose of irradiation required to adequately attenuate each sporozoite. This was studied in the Plasmodium yoelii rodent model system. Exposure to 100 Gy completely attenuated P. yoelii sporozoites (PySPZ). Next we demonstrated that immunization of mice intravenously with 3 doses of 750 PySPZ that had received 200 Gy, double the radiation dose required for attenuation, resulted in 100% protection. These results support the contention that a radiation attenuated sporozoite vaccine for malaria will be safe and effective at a range of radiation doses.


Subject(s)
Malaria Vaccines/radiation effects , Plasmodium yoelii/immunology , Sporozoites/radiation effects , Animals , Dose-Response Relationship, Radiation , Female , Immunization , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Safety , Sporozoites/immunology , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Vaccines, Attenuated/radiation effects
12.
Exp Parasitol ; 118(2): 247-52, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17935717

ABSTRACT

Humans immunized by the bites of irradiated Plasmodium falciparum (Pf) sporozoite-infected mosquitoes are protected against malaria. Radiation attenuates the sporozoites preventing them from fully developing and replicating in hepatocytes, but the effects of radiation on gene expression in sporozoites are unknown. We used RT-PCR (35 cycles of PCR followed by densitometry) to assess the expression of ten genes in Pf sporozoites, and in sporozoites irradiated with 15,000cGy. Irradiation reduced expression substantially (>60%) of two DNA repair genes; moderately (30-60%) of PfUIS3, the Pf orthologue of PbUIS3, a gene up-regulated in Plasmodium berghei sporozoites and of a third DNA repair gene; and minimally (<30%) of the Pf18S ribosomal RNA, PfCSP, PfSSP2/TRAP, and PfCELTOS genes. Irradiation increased expression of PfSPATR minimally. PfLSA1 RNA was not detectable in sporozoites. These results establish that radiation of sporozoites affects gene expression levels and provide the foundation for studies to identify specific genes involved in attenuation and protective immunity.


Subject(s)
Gene Expression/radiation effects , Plasmodium falciparum/radiation effects , Animals , DNA Repair/genetics , DNA, Complementary/analysis , DNA, Complementary/biosynthesis , Malaria Vaccines , Membrane Proteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , RNA, Ribosomal, 18S/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sporozoites/radiation effects , Vaccines, Attenuated
13.
Expert Opin Drug Saf ; 6(5): 505-21, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17877439

ABSTRACT

Antimalarial drugs remain the major intervention tool for the global malaria control efforts that save millions of lives. Nonetheless, emergence and spread of Plasmodium parasites resistant against chloroquine and other major antimalarial drugs has brought the urgency to develop a new generation of safe and effective drugs against malaria. In this article, the safety data for major antimalarial drugs is reviewed. Although an ample amount of clinical data regarding the safety and tolerability of several of these drugs in older children and adults is available, more critical safety and tolerability studies in pregnant women and young children is desirable. To offset the partial loss in efficacy due to drug resistance in malaria parasites acquired against specific drugs, treatment regimens often rely upon the combination of two or more drugs. However, combination therapy requires additional safety, toxicity and tolerability studies in all population groups where these drugs are administered. A uniform standard in assessing the safety and tolerability of antimalarial drugs will be useful in the formulation and implementation of malaria treatment policies that are based on the drug effectiveness, safety and tolerability.


Subject(s)
Antimalarials/adverse effects , Malaria/drug therapy , Animals , Antimalarials/pharmacokinetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/physiology , Humans , Malaria/immunology , Malaria/metabolism , Malaria/prevention & control , Malaria Vaccines/adverse effects , Malaria Vaccines/pharmacokinetics
14.
Infect Immun ; 73(9): 5402-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16113256

ABSTRACT

Proteins present on the surface of malaria parasites that participate in the process of invasion and adhesion to host cells are considered attractive vaccine targets. Aided by the availability of the partially completed genome sequence of the simian malaria parasite Plasmodium knowlesi, we have identified a 786-bp DNA sequence that encodes a 262-amino-acid-long protein, containing an altered version of the thrombospondin type I repeat domain (SPATR). Thrombospondin type 1 repeat domains participate in biologically diverse functions, such as cell attachment, mobility, proliferation, and extracellular protease activities. The SPATR from P. knowlesi (PkSPATR) shares 61% and 58% sequence identity with its Plasmodium falciparum and Plasmodium yoelii orthologs, respectively. By immunofluorescence analysis, we determined that PkSPATR is a multistage antigen that is expressed on the surface of P. knowlesi sporozoite and erythrocytic stage parasites. Recombinant PkSPATR produced in Escherichia coli binds to a human hepatoma cell line, HepG2, suggesting that PkSPATR is a parasite ligand that could be involved in sporozoite invasion of liver cells. Furthermore, recombinant PkSPATR reacted with pooled sera from P. knowlesi-infected rhesus monkeys, indicating that native PkSPATR is immunogenic during infection. Further efficacy evaluation studies in the P. knowlesi-rhesus monkey sporozoite challenge model will help to decide whether the SPATR molecule should be developed as a vaccine against human malarias.


Subject(s)
Antigens, Protozoan/genetics , Membrane Proteins/genetics , Plasmodium knowlesi/genetics , Protozoan Proteins/genetics , Thrombospondins/chemistry , Amino Acid Sequence , Animals , Antigens, Protozoan/metabolism , Base Sequence , Cloning, Molecular , Fluorescent Antibody Technique , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Sequence Data , Plasmodium knowlesi/chemistry , Protein Sorting Signals , Protein Structure, Tertiary , Protozoan Proteins/biosynthesis , Protozoan Proteins/chemistry , Repetitive Sequences, Amino Acid , Sequence Alignment , Thrombospondins/genetics
15.
J Biol Chem ; 280(21): 20524-9, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15781464

ABSTRACT

Circumsporozoite, a predominant surface protein, is involved in invasion of liver cells by Plasmodium sporozoites, which leads to malaria. We have previously reported that the amino terminus region (amino acids 27-117) of P. falciparum circumsporozoite protein plays a critical role in the invasion of liver cells by the parasite. Here we show that invasion-blocking antibodies are induced by a polypeptide encoding these 91 amino acids, only when it is presented in the absence of the rest of the protein. This suggests that when present in the whole protein, the amino terminus remains immunologically cryptic. A single reactive epitope was identified and mapped to a stretch of 21 amino acids from position 93 to 113. The epitope is configurational in nature, since its recognition was affected by deleting as little as 3 amino acids from either end of the 21-residue peptide. Lysine 104, the only known polymorphic position in the epitope, affected its recognition by the antibodies, and its conversion to leucine in the protein led to a substantial loss of binding activity of the protein to the hepatocytes. This indicated that in the protein, the epitope serves as a binding ligand and facilitates the interaction between sporozoite and hepatic cells. When considered along with the observation that in its native state this motif is immunologically unresponsive, we suggest that hiding functional moieties of the protein from the immune system is an evasion strategy to preserve liver cell binding function and may be of importance in designing anti-sporozoite vaccines.


Subject(s)
Antibodies/immunology , Antigens, Protozoan/immunology , Liver/immunology , Liver/parasitology , Plasmodium falciparum , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Carcinoma, Hepatocellular , Fluorescent Antibody Technique , Humans , Liver Neoplasms , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmodium falciparum/chemistry , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Polymerase Chain Reaction , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sporozoites/growth & development , Sporozoites/pathogenicity , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Am J Trop Med Hyg ; 71(4): 457-65, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15516643

ABSTRACT

We describe the epidemiology of malaria in San Dulakudar, a village in Sundargarh District in the state of Orissa in eastern India. Malaria transmission is perennial with Plasmodium falciparum, accounting for greater than 80% of malaria cases. Transmission intensity varies with season with high transmission after the monsoon rains in autumn and winter, low transmission in summer, and intermediate transmission in spring. The anthropophagic mosquito Anopheles fluviatilis was identified as the main vector for malaria transmission. Based on observations of spleen rates and supported by data on malaria parasite prevalence and malaria incidence, San Dulakudar can be classified as a hyperendemic area for P. falciparum malaria. Parasite prevalence and malaria incidence rates decrease with age, suggesting that residents of San Dulakudar develop immunity to malaria. The study demonstrates the presence of regions in the Indian subcontinent such as Sundargarh District where P. falciparum is the primary cause of malaria and where malaria transmission rates are comparable to those found in many parts of Africa.


Subject(s)
Endemic Diseases , Immunity, Innate , Malaria, Falciparum , Plasmodium falciparum/classification , Plasmodium falciparum/isolation & purification , Adolescent , Adult , Animals , Anopheles/parasitology , Child , Child, Preschool , Humans , Incidence , India/epidemiology , Infant , Insect Vectors/parasitology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Prevalence , Rural Population
17.
J Immunol ; 172(9): 5561-9, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15100299

ABSTRACT

Vaccine-induced protection against diseases like malaria, AIDS, and cancer may require induction of Ag-specific CD8(+) and CD4(+) T cell and Ab responses in the same individual. In humans, a recombinant Plasmodium falciparum circumsporozoite protein (PfCSP) candidate vaccine, RTS,S/adjuvant system number 2A (AS02A), induces T cells and Abs, but no measurable CD8(+) T cells by CTL or short-term (ex vivo) IFN-gamma ELISPOT assays, and partial short-term protection. P. falciparum DNA vaccines elicit CD8(+) T cells by these assays, but no protection. We report that sequential immunization with a PfCSP DNA vaccine and RTS,S/AS02A induced PfCSP-specific Abs and Th1 CD4(+) T cells, and CD8(+) cytotoxic and Tc1 T cells. Depending upon the immunization regime, CD4(+) T cells were involved in both the induction and production phases of PfCSP-specific IFN-gamma responses, whereas, CD8(+) T cells were involved only in the production phase. IFN-gamma mRNA up-regulation was detected in both CD45RA(-) (CD45RO(+)) and CD45RA(+)CD4(+) and CD8(+) T cell populations after stimulation with PfCSP peptides. This finding suggests CD45RA(+) cells function as effector T cells. The induction in humans of the three primary Ag-specific adaptive immune responses establishes a strategy for developing immunization regimens against diseases in desperate need of vaccines.


Subject(s)
Antibodies, Protozoan/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunization Schedule , Malaria Vaccines/administration & dosage , Plasmodium falciparum/immunology , Protozoan Proteins/administration & dosage , Vaccines, DNA/administration & dosage , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Hepatitis B Antibodies/biosynthesis , Hepatitis B Surface Antigens/immunology , Humans , Immunization, Secondary/methods , Interferon-gamma/biosynthesis , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Molecular Sequence Data , Protozoan Proteins/genetics , Protozoan Proteins/immunology , T-Lymphocyte Subsets , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Vaccines, DNA/genetics , Vaccines, DNA/immunology
18.
Infect Immun ; 72(3): 1594-602, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14977966

ABSTRACT

We describe a novel approach for identifying target antigens for preerythrocytic malaria vaccines. Our strategy is to rapidly test hundreds of DNA vaccines encoding exons from the Plasmodium yoelii yoelii genomic sequence. In this antigen identification method, we measure reduction in parasite burden in the liver after sporozoite challenge in mice. Orthologs of protective P. y. yoelii genes can then be identified in the genomic databases of Plasmodium falciparum and Plasmodium vivax and investigated as candidate antigens for a human vaccine. A pilot study to develop the antigen identification method approach used 192 P. y. yoelii exons from genes expressed during the sporozoite stage of the life cycle. A total of 182 (94%) exons were successfully cloned into a DNA immunization vector with the Gateway cloning technology. To assess immunization strategies, mice were vaccinated with 19 of the new DNA plasmids in addition to the well-characterized protective plasmid encoding P. y. yoelii circumsporozoite protein. Single plasmid immunization by gene gun identified a novel vaccine target antigen which decreased liver parasite burden by 95% and which has orthologs in P. vivax and P. knowlesi but not P. falciparum. Intramuscular injection of DNA plasmids produced a different pattern of protective responses from those seen with gene gun immunization. Intramuscular immunization with plasmid pools could reduce liver parasite burden in mice despite the fact that none of the plasmids was protective when given individually. We conclude that high-throughput cloning of exons into DNA vaccines and their screening is feasible and can rapidly identify new malaria vaccine candidate antigens.


Subject(s)
Antigens, Protozoan/genetics , Malaria Vaccines/genetics , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Animals , Antibodies, Protozoan/blood , Base Sequence , Biolistics , Cloning, Molecular , DNA Primers , DNA, Protozoan/genetics , Exons , Female , Fluorescent Antibody Technique, Indirect , Genome, Protozoan , Humans , Injections, Intramuscular , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Plasmids/genetics , Plasmodium yoelii/growth & development , Polymerase Chain Reaction , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/pharmacology
19.
Mol Biochem Parasitol ; 133(2): 255-65, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14698437

ABSTRACT

The ability of Plasmodium falciparum-infected erythrocytes to adhere to endothelial receptors and sequester in diverse host organs is an important pathogenic mechanism. Cytoadherence is mediated by variant surface antigens, which are referred to as PfEMP-1 and are encoded by var genes. The extracellular regions of PfEMP-1 contain multiple conserved cysteine-rich domains that are referred to as Duffy-binding-like (DBL) domains. Here, we analyze the adhesive phenotype of an Indian P. falciparum field isolate, JDP8, which binds ICAM-1 but does not bind CD36. This is a unique cytoadherence phenotype because P. falciparum strains that bind ICAM-1 described thus far usually also bind CD36. Moreover, binding to both receptors is thought to be important for static adhesion under flow. The ICAM-1 binding population of P. falciparum JDP8 adheres to endothelial cells under flow despite poor binding to CD36. We have also identified an expressed var gene, JDP8Icvar, which mediates the ICAM-1 binding phenotype of JDP8. Expression of different regions of JDP8Icvar on the surface of COS-7 cells followed by binding assays demonstrates that the ICAM-1 binding domain maps to the DBL2betaC2 domain of JDP8Icvar. Sequence comparison with two previously identified ICAM-1 binding domains of PfEMP-1, which also map to DBLbetaC2 domains, suggests that diverse P. falciparum isolates use a structurally conserved domain to bind ICAM-1. It thus appears that functional constraints may place limits on the extent of sequence diversity in receptor-binding domains of PfEMP-1.


Subject(s)
Cell Adhesion , Intercellular Adhesion Molecule-1/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , CD36 Antigens/metabolism , CHO Cells , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , DNA, Protozoan/chemistry , DNA, Protozoan/isolation & purification , Endothelial Cells/parasitology , Gene Expression , Genes, Protozoan , Humans , Molecular Sequence Data , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protein Binding , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Alignment , Sequence Analysis, DNA
20.
J Biol Chem ; 278(28): 25977-81, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12716913

ABSTRACT

The annotated sequence of chromosome 2 of Plasmodium falciparum was examined for genes encoding proteins that may be of interest for vaccine development. We describe here the characterization of a protein with an altered thrombospondin Type I repeat domain (PfSPATR) that is expressed in the sporozoite, asexual, and sexual erythrocytic stages of the parasite life cycle. Immunoelectron microscopy indicated that this protein was expressed on the surface of the sporozoites and around the rhoptries in the asexual erythrocytic stage. An Escherichia coli-produced recombinant form of the protein bound to HepG2 cells in a dose-dependent manner and antibodies raised against this protein blocked the invasion of sporozoites into a transformed hepatoma cell line. Sera from Ghanaian adults and from a volunteer who had been immunized with radiation-attenuated P. falciparum sporozoites specifically recognized the expression of this protein on transfected COS-7 cells. These data support the evaluation of this protein as a vaccine candidate.


Subject(s)
Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Thrombospondins/chemistry , Amino Acid Sequence , Animals , COS Cells , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Line, Transformed , Cloning, Molecular , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Humans , Malaria/blood , Mice , Microscopy, Fluorescence , Microscopy, Immunoelectron , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protozoan Proteins/biosynthesis , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...