Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Proteomics ; 15(11): 3373-3387, 2016 11.
Article in English | MEDLINE | ID: mdl-27624304

ABSTRACT

The innate immune response is highly conserved across all eukaryotes and has been studied in great detail in several model organisms. Hemocytes, the primary immune cell population in mosquitoes, are important components of the mosquito innate immune response, yet critical aspects of their biology have remained uncharacterized. Using a novel method of enrichment, we isolated phagocytic granulocytes and quantified their proteomes by mass spectrometry. The data demonstrate that phagocytosis, blood-feeding, and Plasmodium falciparum infection promote dramatic shifts in the proteomic profiles of An. gambiae granulocyte populations. Of interest, large numbers of immune proteins were induced in response to blood feeding alone, suggesting that granulocytes have an integral role in priming the mosquito immune system for pathogen challenge. In addition, we identify several granulocyte proteins with putative roles as membrane receptors, cell signaling, or immune components that when silenced, have either positive or negative effects on malaria parasite survival. Integrating existing hemocyte transcriptional profiles, we also compare differences in hemocyte transcript and protein expression to provide new insight into hemocyte gene regulation and discuss the potential that post-transcriptional regulation may be an important component of hemocyte gene expression. These data represent a significant advancement in mosquito hemocyte biology, providing the first comprehensive proteomic profiling of mosquito phagocytic granulocytes during homeostasis blood-feeding, and pathogen challenge. Together, these findings extend current knowledge to further illustrate the importance of hemocytes in shaping mosquito innate immunity and their principal role in defining malaria parasite survival in the mosquito host.


Subject(s)
Anopheles/immunology , Hemocytes/metabolism , Immunity, Innate , Insect Proteins/metabolism , Proteomics/methods , Animals , Anopheles/metabolism , Anopheles/parasitology , Female , Gene Expression Profiling , Gene Expression Regulation , Insect Proteins/genetics , Mass Spectrometry , Phagocytosis , Plasmodium falciparum/immunology , Stress, Physiological
2.
Malar J ; 13: 92, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24620841

ABSTRACT

BACKGROUND: An effective malaria vaccine remains elusive. The most effective experimental vaccines confer only limited and short-lived protection despite production of protective antibodies. However, immunization with irradiated sporozoites, or with live sporozoites under chloroquine cover, has resulted in long-term protection apparently due to the generation of protective CD8+ T cells. The nature and function of these protective CD8+ T cells has not been elucidated. In the current study, the phenotype of CD8+ T cells generated after immunization of C57BL/6 mice with live Plasmodium berghei sporozoites under chloroquine cover was investigated. METHODS: Female C57BL/6 mice, C57BL/6 mice B2 macroglobulin -/- [KO], or invariant chain-/- [Ic KO] [6-8 weeks old] were immunized with P. berghei sporozoites and treated daily with 800 µg/mouse of chloroquine for nine days. This procedure of immunization is referred to as "infection/cure". Mice were challenged by inoculating intravenously 1,000 infectious sporozoites. Appearance of parasitaemia was monitored by Giemsa-stained blood smears. RESULTS: By use of MHC I and MHC II deficient animals, results indicate that CD8+ T cells are necessary for full protection and that production of protective antibodies is either CD4+ T helper cells dependent and/or lymphokines produced by CD4 cells contribute to the protection directly or by helping CD8+ T cells. Further, the phenotype of infection/cure P. berghei responsive CD8+ T cells was determined to be KLRG1high CD27low CD44high and CD62Llow. CONCLUSION: The KLRG1high CD27low CD44high and CD62Llow phenotype of CD8+ T cells is associated with protection and should be investigated further as a candidate correlate of protection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chloroquine/administration & dosage , Immunization/methods , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Plasmodium berghei/immunology , Sporozoites/immunology , Animals , Antigens, CD/analysis , CD8-Positive T-Lymphocytes/chemistry , Female , Immunophenotyping , Lectins, C-Type , Mice , Mice, Inbred C57BL , Receptors, Immunologic/analysis
3.
Clin Vaccine Immunol ; 20(6): 803-10, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536694

ABSTRACT

Circumsporozoite protein (CSP) of Plasmodium falciparum is a protective human malaria vaccine candidate. There is an urgent need for models that can rapidly down-select novel CSP-based vaccine candidates. In the present study, the mouse-mosquito transmission cycle of a transgenic Plasmodium berghei malaria parasite stably expressing a functional full-length P. falciparum CSP was optimized to consistently produce infective sporozoites for protection studies. A minimal sporozoite challenge dose was established, and protection was defined as the absence of blood-stage parasites 14 days after intravenous challenge. The specificity of protection was confirmed by vaccinating mice with multiple CSP constructs of differing lengths and compositions. Constructs that induced high NANP repeat-specific antibody titers in enzyme-linked immunosorbent assays were protective, and the degree of protection was dependent on the antigen dose. There was a positive correlation between antibody avidity and protection. The antibodies in the protected mice recognized the native CSP on the parasites and showed sporozoite invasion inhibitory activity. Passive transfer of anti-CSP antibodies into naive mice also induced protection. Thus, we have demonstrated the utility of a mouse efficacy model to down-select human CSP-based vaccine formulations.


Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Parasitemia/prevention & control , Protozoan Proteins/immunology , Vaccination/methods , Animals , Antibodies, Protozoan/blood , Culicidae , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Malaria Vaccines/genetics , Malaria Vaccines/isolation & purification , Mice , Mice, Inbred C57BL , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
4.
PLoS One ; 7(10): e48304, 2012.
Article in English | MEDLINE | ID: mdl-23144750

ABSTRACT

BACKGROUND: The worldwide burden of malaria remains a major public health problem due, in part, to the lack of an effective vaccine against the Plasmodium falciparum parasite. An effective vaccine will most likely require the induction of antigen specific CD8(+) and CD4(+) T-cells as well as long-lasting antibody responses all working in concert to eliminate the infection. We report here the effective modification of a self-assembling protein nanoparticle (SAPN) vaccine previously proven effective in control of a P. berghei infection in a rodent model to now present B- and T-cell epitopes of the human malaria parasite P. falciparum in a platform capable of being used in human subjects. METHODOLOGY/PRINCIPAL FINDINGS: To establish the basis for a SAPN-based vaccine, B- and CD8(+) T-cell epitopes from the P. falciparum circumsporozoite protein (PfCSP) and the universal CD4 T-helper epitope PADRE were engineered into a versatile small protein (∼125 amino acids) that self-assembles into a spherical nanoparticle repetitively displaying the selected epitopes. P. falciparum epitope specific immune responses were evaluated in mice using a transgenic P. berghei malaria parasite of mice expressing the human malaria full-length P. falciparum circumsporozoite protein (Tg-Pb/PfCSP). We show that SAPN constructs, delivered in saline, can induce high-titer, long-lasting (1 year) protective antibody and poly-functional (IFNγ(+), IL-2(+)) long-lived central memory CD8(+) T-cells. Furthermore, we demonstrated that these Ab or CD8(+) T-cells can independently provide sterile protection against a lethal challenge of the transgenic parasites. CONCLUSION: The SAPN construct induces long-lasting antibody and cellular immune responses to epitope specific sequences of the P. falciparum circumsporozoite protein (PfCSP) and prevents infection in mice by a transgenic P. berghei parasite displaying the full length PfCSP.


Subject(s)
Antibodies, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Vaccines, DNA/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/metabolism , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Humans , Malaria/immunology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Models, Molecular , Molecular Sequence Data , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Multimerization , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccines, DNA/administration & dosage
5.
Immunopharmacol Immunotoxicol ; 32(4): 637-46, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20205507

ABSTRACT

BACKGROUND: Skin cancers are common, and there has recently been a dramatic increase in their incidence, particularly in the occurrence of melanoma. Furthermore, relapse after curative surgical treatment of melanoma remains a significant clinical challenge and accounts for most of the mortality of this disease. OBJECTIVE: The aim of this study was to determine whether IMMUNEPOTENT CRP affects B16F10 melanoma cells and tumors growth and vascular endothelial growth factor (VEGF) production in vivo and in vitro. METHODS: B16F10 cells and B16F10-inoculated mice were treated with different concentrations of IMMUNEPOTENT CRP. Outcomes were then evaluated using MTT, TUNEL, Caspase-3, senescence, ELISA and colorimetric assays. Parameters related to survival and tumor weight were also assessed. RESULTS: IMMUNEPOTENT CRP decreased the viability of B16F10 cells by increasing apoptosis of the treated cells, and VEGF production was decreased both in vitro and in vivo. Furthermore, treatment prevented metastasis, delayed the appearance of tumors, decreased tumor weight and improved the survival of tumor-bearing mice. DISCUSSION: These observations suggest that IMMUNEPOTENT CRP can be used to suppress growth and metastasis by using targeting proteins such as VEGF.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Melanoma, Experimental/prevention & control , Transfer Factor/pharmacology , Transfer Factor/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Caspase 3/metabolism , Cattle , Cell Line, Tumor , Cell Survival/drug effects , Cellular Senescence/drug effects , Dose-Response Relationship, Drug , Female , Melanoma, Experimental/metabolism , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/prevention & control , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism
6.
J Immunol ; 183(11): 7268-77, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19915055

ABSTRACT

We have designed and produced a prototypic malaria vaccine based on a highly versatile self-assembling polypeptide nanoparticle (SAPN) platform that can repetitively display antigenic epitopes. We used this platform to display a tandem repeat of the B cell immunodominant repeat epitope (DPPPPNPN)(2)D of the malaria parasite Plasmodium berghei circumsporozoite protein. Administered in saline, without the need for a heterologous adjuvant, the SAPN construct P4c-Mal conferred a long-lived, protective immune response to mice with a broad range of genetically distinct immune backgrounds including the H-2(b), H-2(d), and H-2(k) alleles. Immunized mice produced a CD4(+) T cell-dependent, high-titer, long-lasting, high-avidity Ab response against the B cell epitope. Mice were protected against an initial challenge of parasites up to 6 mo after the last immunization or for up to 15 mo against a second challenge after an initial challenge of parasites had successfully been cleared. Furthermore, we demonstrate that the SAPN platform not only functions to deliver an ordered repetitive array of B cell peptide epitopes but operates as a classical immunological carrier to provide cognate help to the P4c-Mal-specific B cells.


Subject(s)
Antigens, Protozoan/immunology , Epitopes, B-Lymphocyte/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Nanoparticles/therapeutic use , Peptides/therapeutic use , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antibody Affinity , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Malaria/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium berghei/immunology
7.
Vaccine ; 25(21): 4203-12, 2007 May 22.
Article in English | MEDLINE | ID: mdl-17442466

ABSTRACT

We report the first safety and immunogenicity trial of the Plasmodium falciparum vaccine candidate FMP2.1/AS02A, a recombinant E. coli-expressed protein based upon the apical membrane antigen-1 (AMA-1) of the 3D7 clone formulated with the AS02A adjuvant. We conducted an open-label, staggered-start, dose-escalating Phase I trial in 23 malaria-naïve volunteers who received 8, 20 or 40microg of FMP2.1 in a fixed volume of 0.5mL of AS02A on a 0, 1, and 2 month schedule. Nineteen of 23 volunteers received all three scheduled immunizations. The most frequent solicited local and systemic adverse events associated with immunization were injection site pain (68%) and headache (29%). There were no significant laboratory abnormalities or vaccine-related serious adverse events. All volunteers seroconverted after second immunization as determined by ELISA. Immune sera recognized sporozoites and merozoites by immunofluorescence assay (IFA), and exhibited both growth inhibition and processing inhibition activity against homologous (3D7) asexual stage parasites. Post-immunization, peripheral blood mononuculear cells exhibited FMP2.1-specific lymphoproliferation and IFN-gamma and IL-5 ELISPOT assay responses. This is the first PfAMA-1-based vaccine shown to elicit both potent humoral and cellular immunity in humans. Encouraged by the potential of FMP1/AS02A to target host immunity against PfAMA-1 that is known to be expressed by sporozoite, hepatic and erythrocytic stages, we have initiated field trials of FMP2.1/AS02A in an endemic population in the Republic of Mali.


Subject(s)
Antigens, Protozoan/immunology , Lipid A/analogs & derivatives , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Saponins/immunology , Adjuvants, Immunologic , Adolescent , Adult , Animals , Antibodies, Protozoan/blood , Cell Line , Cell Proliferation , Cells, Cultured , Cricetinae , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Female , Fluorescent Antibody Technique, Indirect , Headache , Humans , Immunization, Secondary , Interferon-gamma/biosynthesis , Interleukin-5/biosynthesis , Leukocytes, Mononuclear/immunology , Lipid A/immunology , Malaria Vaccines/administration & dosage , Male , Merozoites/immunology , Mesocricetus , Middle Aged , Pain , Plasmodium falciparum/growth & development , Sporozoites/immunology , Vaccines, Synthetic/immunology
8.
Infect Immun ; 75(2): 838-45, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17101665

ABSTRACT

Liver-stage antigen 1 (LSA1) is expressed by Plasmodium falciparum only during the intrahepatic cell stage of the parasite's development. Immunoepidemiological studies in regions where malaria is endemic suggested an association between the level of LSA1-specific humoral and cell-mediated immune responses and susceptibility to clinical malaria. A recombinant LSA1 protein, FMP011, has been manufactured as a preerythrocytic vaccine to induce an immune response that would have the effect of controlling parasitemia and disease in humans. To evaluate the immunogenicity of FMP011, we analyzed the immune response of three inbred strains of mice to antigen immunization using two different adjuvant formulations, AS01B and AS02A. We report here the ability of BALB/c and A/J mice, but not C57BL/6J mice, to mount FMP011-specific humoral (antibody titer) and cellular (gamma interferon [IFN-gamma] production) responses following immunization with FMP011 formulated in AS01B or AS02A. Immunization of BALB/c and A/J mice with FMP011/AS01B induced more antigen-specific IFN-gamma-producing splenocytes than immunization with FMP011/AS02A. A slightly higher titer of antibody was induced using AS02A than AS01B in both strains. C57BL/6J mice did not respond with any detectable FMP011-specific IFN-gamma splenocytes or antibody when immunized with FMP011 in AS01B or AS02A. Intracellular staining of cells isolated from FMP011/AS01B-immunized BALB/c mice indicated that CD4(+) cells, but not CD8(+) cells, were the main IFN-gamma-producing splenocyte. However, inclusion of blocking anti-CD4(+) antibody during the in vitro restimulation ELISpot analysis failed to completely abolish IFN-gamma production, indicating that while CD4(+) T cells were the major source of IFN-gamma, other cell types also were involved.


Subject(s)
Adjuvants, Immunologic , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Animals , Antibodies, Protozoan/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Interferon-gamma/biosynthesis , Lymphocyte Subsets/immunology , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
9.
J Leukoc Biol ; 78(2): 359-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15937142

ABSTRACT

The major histocompatibility complex nonrestricted cytotoxic leukemic T cell line T acute lymphoblastic leukemia (TALL)-104 is being pursued as a therapeutic agent for cancer. However, the receptors and effector mechanisms responsible for its broad tumoricidal function remain undefined. Here, we examined the roles played by natural cytotoxicity receptors (NCR), killer cell immunoglobulin-like receptors, cytolytic granule components, and tumor necrosis factor (TNF) family members in tumor recognition and lysis by TALL-104 cells. The perforin-granzyme pathway, TNF-related apoptosis-inducing ligand (TRAIL), and Fas were each involved in the lysis of particular tumor targets by TALL-104. Furthermore, phorbol 12-myristate 13-acetate/ionomycin treatment induced surface expression of Fas-L and TRAIL. In addition, supernatants from CD3-stimulated TALL-104 cultures exhibited antiproliferative activity, which was blocked 50-90% by anti-TNF-alpha monoclonal antibody (mAb). Although negative for the NCR natural killer (NK)p44, this cell line was found to express NKp46. An anti-NKp46 antibody strongly blocked TALL-104-mediated lysis of certain targets and directly induced cytokine production, granule release, and redirected lysis responses. Anti-NKG2D and anti-2B4 also stimulated redirected cytotoxicity by TALL-104. By contrast, anti-NKG2A mAb did not stain the cells or inhibit killing responses. Alternatively, KIR3DL2 was detected on TALL-104, and expression of its reported ligand, human leukocyte antigen (HLA)-A, on target cells provided protection from cytotoxicity. Thus, NKp46, NKG2D, and 2B4 are activating receptors, and KIR3DL2 is an inhibitory receptor on TALL-104. The data demonstrate the ability of TALL-104 cells to recognize a wide variety of tumors with NK cell receptors and kill them with a broad arsenal of cytolytic effector mechanisms, including cytolytic granules and TNF family ligands.


Subject(s)
Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Apoptosis Regulatory Proteins , CD3 Complex/immunology , Carcinogens/pharmacology , Cell Degranulation/immunology , Cytotoxicity, Immunologic/drug effects , Fas Ligand Protein , Histocompatibility Antigens Class I/immunology , Humans , Immediate-Early Proteins/immunology , Ionomycin/pharmacology , Ionophores/pharmacology , Jurkat Cells , Lymphocyte Activation/drug effects , Membrane Glycoproteins/immunology , Monomeric GTP-Binding Proteins/immunology , NK Cell Lectin-Like Receptor Subfamily C , NK Cell Lectin-Like Receptor Subfamily K , Natural Cytotoxicity Triggering Receptor 1 , Neoplasms/immunology , Neoplasms/therapy , Receptors, Immunologic/immunology , Receptors, KIR , Receptors, KIR3DL2 , Receptors, Natural Killer Cell , Signal Transduction/drug effects , Signaling Lymphocytic Activation Molecule Family , T-Lymphocytes, Cytotoxic/cytology , TNF-Related Apoptosis-Inducing Ligand , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factors/immunology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...