Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 62(1): 149-59, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22865267

ABSTRACT

We have previously observed that in vivo exposure to growing melanoma tumors fundamentally alters activated T cell homeostasis by suppressing the ability of naïve T cells to undergo antigen-driven proliferative expansion. We hypothesized that exposure of T cells in later stages of differentiation to melanoma would have similar suppressive consequences. C57BL/6 mice were inoculated with media or syngeneic B16F10 melanoma tumors 8 or 60 days after infection with lymphocytic choriomeningitis virus (LCMV), and splenic populations of LCMV-specific T cells were quantified using flow cytometry 18 days after tumor inoculation. Inoculation with melanoma on post-infection day 8 potentiated the contraction of previously activated T cells. This enhanced contraction was associated with increased apoptotic susceptibility among T cells from tumor-bearing mice. In contrast, inoculation with melanoma on post-infection day 60 did not affect the ability of previously established memory T cells to maintain themselves in stable numbers. In addition, the ability of previously established memory T cells to respond to LCMV challenge was unaffected by melanoma. Following adoptive transfer into melanoma-bearing mice, tumor-specific memory T cells were significantly more effective at controlling melanoma growth than equivalent numbers of tumor-specific effector T cells. These observations suggest that memory T cells are uniquely resistant to suppressive influences exerted by melanoma on activated T cell homeostasis; these findings may have implications for T cell-based cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Animals , Apoptosis/immunology , Disease Models, Animal , Female , Flow Cytometry , Immunotherapy, Adoptive , Male , Mice , Mice, Inbred C57BL
2.
Cell Immunol ; 271(1): 104-9, 2011.
Article in English | MEDLINE | ID: mdl-21741629

ABSTRACT

We have observed that in vivo interaction between melanoma and resting T cells promotes suppression of antigen-driven proliferative T cell expansion. We hypothesized that this suppression would affect tumor antigen-specific T cell populations more potently than tumor-unrelated T cell populations. A B16F10 cell line was stably transfected to express low levels of the lymphocytic choriomeningitis virus (LCMV) glycoprotein GP33 (B16GP33). Mice bearing B16F10 or B16GP33 tumors were infected with LCMV, and proliferative expansion of LCMV epitope-specific T cell populations was quantified. In vitro and in vivo assays confirmed low levels of antigenic GP33 expression by B16GP33 tumors. Suppressed expansion of GP33-specific T cells was equivalent between mice bearing B16F10 and B16GP33 tumors. These observations suggest that the ability of growing melanoma tumors to impair antigen-driven proliferative expansion of activated T cells is global and not antigen-specific, and provide further insight into the influence of cancer on activated T cell homeostasis.


Subject(s)
Antigens, Neoplasm/immunology , Cell Proliferation , Melanoma, Experimental/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line, Tumor , Female , Flow Cytometry , Glycoproteins/genetics , Glycoproteins/immunology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Peptide Fragments/genetics , Peptide Fragments/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Tumor Burden/immunology , Viral Proteins/genetics , Viral Proteins/immunology
3.
Expert Rev Vaccines ; 9(7): 731-45, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20624047

ABSTRACT

Prostate cancer is a significant public health problem, and the most commonly diagnosed cancer in the USA. The long natural history of prostate cancer, the presence of a serum biomarker that can be used to detect very early recurrences, and the previous identification of multiple potential tissue-specific target antigens are all features that make this disease suitable for the development of anti-tumor vaccines. To date, many anti-tumor vaccines have entered clinical testing for patients with prostate cancer, and some have demonstrated clinical benefit. DNA vaccines represent one vaccine approach that has been evaluated in multiple preclinical models and clinical trials. The safety, specificity for the target antigen, ease of manufacturing and ease of incorporating other immune-modulating approaches make DNA vaccines particularly relevant for future development. This article focuses on DNA vaccines specifically in the context of prostate cancer treatment, focusing on antigens targeted in preclinical models, recent clinical trials and efforts to improve the potency of these vaccines.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy/methods , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Vaccines, DNA/immunology , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Male
4.
Vaccine ; 25(50): 8410-9, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17980467

ABSTRACT

The anthrax exotoxin, which is a key mediator of anthrax related pathogenesis, is composed of two separate toxins formed by pairwise combinations of three proteins that are encoded on the pXO1 plasmid of Bacillus anthracis. Lethal toxin is composed of protective antigen (PA) combined with lethal factor (LF) while edema toxin is composed of PA and edema factor (EF). The present study found that the catalytic mutants of LF (LFE687A) and EF (EFH351A) competitively inhibited lethal toxin and edema toxin-mediated activity in vitro and lethality in vivo and were non-toxic to sensitive cell lines when combined with PA. While PA combined with EFH351A was non-lethal in mice, PA combined with LFE687A was of reduced virulence. Full protection of mice against a lethal toxin challenge required injection of mice with PA combined with both LFE687A and EFH351A. The potential use of these full-length, biologically inactive mutant proteins combined with PA as prophylactics or therapeutics is discussed.


Subject(s)
Anthrax Vaccines , Anthrax/prevention & control , Antibodies, Bacterial/blood , Antigens, Bacterial , Bacillus anthracis/genetics , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins , Animals , Anthrax/immunology , Anthrax Vaccines/genetics , Anthrax Vaccines/immunology , Anthrax Vaccines/therapeutic use , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/therapeutic use , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/therapeutic use , CHO Cells , Catalysis , Cell Line , Cricetinae , Cricetulus , Female , Macrophages , Mice , Mutation
5.
Arch Biochem Biophys ; 446(1): 28-34, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16386701

ABSTRACT

Edema factor is a calmodulin dependent adenylyl cyclase secreted as one of the primary exotoxins by Bacillus anthracis. A histidine residue at position 351 located in its active site has been implicated in catalysis but direct evidence of its functional role is still lacking. In the present study, we introduced mutations in full-length edema factor (EF) to generate alanine (H351A), asparagine (H351N), and phenylalanine (H351F) variants. Spectral analysis of these variants displayed no gross structural deformities. Kinetic characterization showed that the adenylyl cyclase activity of H351N and H351F mutants decreased 34- and 40-fold, respectively, whereas H351A mutant completely lost activity. K(m) and K(i) values for ATP, pH activity profiles, and calmodulin activation curves of asparagine and phenylalanine mutants were not altered markedly. This kinetic data corroborated our ligand binding studies. Apparent K(d) values for calmodulin and ATP binding were found to be similar for wild-type EF and these active site variants. The effective substitution of H351 by asparagine and phenylalanine, albeit at a greatly reduced K(cat), without perturbing the ATP binding highlights the importance of this residue in transition-state stabilization. This was also evident from the positive free energy difference calculated for these mutants. However, equilibrium dialysis experiments revealed noticeable increase in ATP binding constant of H351A mutant, suggesting an additional role of H351 in precise substrate binding in the catalytic pocket. This is the first comprehensive study that describes the kinetic and ligand binding properties of H351 mutants and validates the importance of this residue in EF catalysis.


Subject(s)
Adenylyl Cyclases/metabolism , Bacillus anthracis/enzymology , Histidine/genetics , Adenosine Triphosphate/metabolism , Adenylyl Cyclases/chemistry , Adenylyl Cyclases/genetics , Alanine/genetics , Animals , Asparagine/genetics , Base Sequence , Binding Sites , Calmodulin/metabolism , Catalysis , Hydrogen-Ion Concentration , Kinetics , Mutation , Phenylalanine/genetics , Viper Venoms
6.
Biochem Biophys Res Commun ; 339(1): 107-14, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16293226

ABSTRACT

Edema toxin is a key virulence determinant in anthrax pathogenesis that causes augmentation of cAMP inside host cells. This exotoxin has been implicated in facilitating bacterial invasion by impairing host defenses. Here, we report for the first time that edema toxin plays an important role in suppression of platelet aggregation; an effect that could be of vital significance in anthrax afflicted subjects. It was found that edema toxin induces a dose dependent and time dependent increase in cAMP inside rabbit platelets. Elevation of cAMP led to suppression of platelet aggregation as demonstrated by in vitro aggregation assays. A 95% suppression of platelet aggregation in response to thrombin and a complete suppression in response to ADP, at toxin concentrations of 7 and 2.2 nM, respectively, were observed. Antibody neutralized wild type edema factor and non-toxic mutants of this binary toxin failed to show any alteration in the normal aggregation pattern. Edema toxin caused the activation of cAMP dependent protein kinase A inside platelets, a phenomenon that could be speculated to initiate the cascade of events responsible for suppressing platelet aggregation. Furthermore, in vivo bleeding time registered a sharp increase in response to edema toxin. These findings can explicate the systemic occurrence of hemorrhage, which is a prominent symptom of anthrax. This study exemplifies how Bacillus anthracis has evolved the ability to use host's physiological processes by mimicking the eukaryotic signal transduction machinery, thus inflicting persistent infection.


Subject(s)
Antigens, Bacterial/physiology , Bacillus anthracis/physiology , Platelet Aggregation , Adenosine Diphosphate/pharmacology , Animals , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Bleeding Time , Blood Platelets/metabolism , Cyclic AMP/biosynthesis , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Mice , Platelet Aggregation/drug effects , Rabbits , Thrombin/pharmacology
7.
Biochem Biophys Res Commun ; 307(3): 446-50, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12893241

ABSTRACT

The anthrax toxin complex is primarily responsible for most of the symptoms of anthrax. This complex is composed of three proteins, anthrax protective antigen, anthrax edema factor, and anthrax lethal factor. The three proteins act in binary combination of protective antigen plus edema factor (edema toxin) and protective antigen plus lethal factor (lethal toxin) that paralyze the host defenses and eventually kill the host. Both edema factor and lethal factor are intracellularly acting proteins that require protective antigen for their delivery into the host cell. In this study, we show that deletion of certain residues of protective antigen results in variants of protective antigen that inhibit the action of anthrax toxin both in vitro and in vivo. These mutants protected mice against both lethal toxin and edema toxin challenge, even when injected at a 1:8 ratio relative to the wild-type protein. Thus, these mutant proteins are promising candidates that may be used to neutralize the action of anthrax toxin.


Subject(s)
Anthrax/prevention & control , Antigens, Bacterial , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , CHO Cells , Cell Line , Cricetinae , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...