Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 9(4): 415-429, 2021 04.
Article in English | MEDLINE | ID: mdl-33500272

ABSTRACT

Metabolic dysregulation is a hallmark of cancer. Many tumors exhibit auxotrophy for various amino acids, such as arginine, because they are unable to meet the demand for these amino acids through endogenous production. This vulnerability can be exploited by employing therapeutic strategies that deplete systemic arginine in order to limit the growth and survival of arginine auxotrophic tumors. Pegzilarginase, a human arginase-1 enzyme engineered to have superior stability and enzymatic activity relative to the native human arginase-1 enzyme, depletes systemic arginine by converting it to ornithine and urea. Therapeutic administration of pegzilarginase in the setting of arginine auxotrophic tumors exerts direct antitumor activity by starving the tumor of exogenous arginine. We hypothesized that in addition to this direct effect, pegzilarginase treatment indirectly augments antitumor immunity through increased antigen presentation, thus making pegzilarginase a prime candidate for combination therapy with immuno-oncology (I-O) agents. Tumor-bearing mice (CT26, MC38, and MCA-205) receiving pegzilarginase in combination with anti-PD-L1 or agonist anti-OX40 experienced significantly increased survival relative to animals receiving I-O monotherapy. Combination pegzilarginase/immunotherapy induced robust antitumor immunity characterized by increased intratumoral effector CD8+ T cells and M1 polarization of tumor-associated macrophages. Our data suggest potential mechanisms of synergy between pegzilarginase and I-O agents that include increased intratumoral MHC expression on both antigen-presenting cells and tumor cells, and increased presence of M1-like antitumor macrophages. These data support the clinical evaluation of I-O agents in conjunction with pegzilarginase for the treatment of patients with cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Arginase/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/pharmacology , Receptors, OX40/antagonists & inhibitors , Adoptive Transfer , Animals , Arginase/analysis , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Receptors, OX40/metabolism
2.
Mol Cancer Ther ; 13(10): 2352-60, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25053820

ABSTRACT

miR34a is a tumor-suppressor miRNA that functions within the p53 pathway to regulate cell-cycle progression and apoptosis. With apparent roles in metastasis and cancer stem cell development, miR34a provides an interesting opportunity for therapeutic development. A mimic of miR34a was complexed with an amphoteric liposomal formulation and tested in two different orthotopic models of liver cancer. Systemic dosing of the formulated miR34a mimic increased the levels of miR34a in tumors by approximately 1,000-fold and caused statistically significant decreases in the mRNA levels of several miR34a targets. The administration of the formulated miR34a mimic caused significant tumor growth inhibition in both models of liver cancer, and tumor regression was observed in more than one third of the animals. The antitumor activity was observed in the absence of any immunostimulatory effects or dose-limiting toxicities. Accumulation of the formulated miR34a mimic was also noted in the spleen, lung, and kidney, suggesting the potential for therapeutic use in other cancers.


Subject(s)
Biomimetic Materials/administration & dosage , Liver Neoplasms/drug therapy , MicroRNAs/genetics , Animals , Biomimetic Materials/adverse effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Liver Neoplasms/genetics , Mice, Inbred NOD , Mice, SCID , RNA, Messenger/genetics , Xenograft Model Antitumor Assays
3.
Mol Ther ; 19(6): 1116-22, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21427705

ABSTRACT

MicroRNAs (miRNAs) are emerging as potential cancer therapeutics, but effective delivery mechanisms to tumor sites are a roadblock to utility. Here we show that systemically delivered, synthetic miRNA mimics in complex with a novel neutral lipid emulsion are preferentially targeted to lung tumors and show therapeutic benefit in mouse models of lung cancer. Therapeutic delivery was demonstrated using mimics of the tumor suppressors, microRNA-34a (miR-34a) and let-7, both of which are often down regulated or lost in lung cancer. Systemic treatment of a Kras-activated autochthonous mouse model of non-small cell lung cancer (NSCLC) led to a significant decrease in tumor burden. Specifically, mice treated with miR-34a displayed a 60% reduction in tumor area compared to mice treated with a miRNA control. Similar results were obtained with the let-7 mimic. These findings provide direct evidence that synthetic miRNA mimics can be systemically delivered to the mammalian lung and support the promise of miRNAs as a future targeted therapy for lung cancer.


Subject(s)
Emulsions/chemistry , Genetic Vectors/chemistry , Lipids/chemistry , Lung Neoplasms/therapy , MicroRNAs/physiology , Animals , Cell Line, Tumor , Humans , Mice , MicroRNAs/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
Nat Med ; 17(2): 211-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21240262

ABSTRACT

Cancer stem cells (CSCs), or tumor-initiating cells, are involved in tumor progression and metastasis. MicroRNAs (miRNAs) regulate both normal stem cells and CSCs, and dysregulation of miRNAs has been implicated in tumorigenesis. CSCs in many tumors--including cancers of the breast, pancreas, head and neck, colon, small intestine, liver, stomach, bladder and ovary--have been identified using the adhesion molecule CD44, either individually or in combination with other marker(s). Prostate CSCs with enhanced clonogenic and tumor-initiating and metastatic capacities are enriched in the CD44(+) cell population, but whether miRNAs regulate CD44(+) prostate cancer cells and prostate cancer metastasis remains unclear. Here we show, through expression analysis, that miR-34a, a p53 target, was underexpressed in CD44(+) prostate cancer cells purified from xenograft and primary tumors. Enforced expression of miR-34a in bulk or purified CD44(+) prostate cancer cells inhibited clonogenic expansion, tumor regeneration, and metastasis. In contrast, expression of miR-34a antagomirs in CD44(-) prostate cancer cells promoted tumor development and metastasis. Systemically delivered miR-34a inhibited prostate cancer metastasis and extended survival of tumor-bearing mice. We identified and validated CD44 as a direct and functional target of miR-34a and found that CD44 knockdown phenocopied miR-34a overexpression in inhibiting prostate cancer regeneration and metastasis. Our study shows that miR-34a is a key negative regulator of CD44(+) prostate cancer cells and establishes a strong rationale for developing miR-34a as a novel therapeutic agent against prostate CSCs.


Subject(s)
Hyaluronan Receptors/drug effects , MicroRNAs/therapeutic use , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , Animals , Gene Expression Regulation, Neoplastic/drug effects , Male , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Reverse Transcriptase Polymerase Chain Reaction
5.
Cancer Res ; 70(14): 5923-30, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20570894

ABSTRACT

Tumor suppressor microRNAs (miRNA) provide a new opportunity to treat cancer. This approach, "miRNA replacement therapy," is based on the concept that the reintroduction of miRNAs depleted in cancer cells reactivates cellular pathways that drive a therapeutic response. Here, we describe the development of a therapeutic formulation using chemically synthesized miR-34a and a lipid-based delivery vehicle that blocks tumor growth in mouse models of non-small-cell lung cancer. This formulation is effective when administered locally or systemically. The antioncogenic effects are accompanied by an accumulation of miR-34a in the tumor tissue and downregulation of direct miR-34a targets. Intravenous delivery of formulated miR-34a does not induce an elevation of cytokines or liver and kidney enzymes in serum, suggesting that the formulation is well tolerated and does not induce an immune response. The data provide proof of concept for the systemic delivery of a synthetic tumor suppressor mimic, obviating obstacles associated with viral-based miRNA delivery and facilitating a rapid route for miRNA replacement therapy into the clinic.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/therapy , MicroRNAs/administration & dosage , MicroRNAs/genetics , Animals , Biomimetic Materials/administration & dosage , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , Oligonucleotides/administration & dosage , Oligonucleotides/genetics , Transcription, Genetic , Transfection , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
6.
Cell Cycle ; 7(6): 759-64, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18344688

ABSTRACT

MicroRNAs have been increasingly implicated in human cancer and interest has grown about the potential to use microRNAs to combat cancer. Lung cancer is the most prevalent form of cancer worldwide and lacks effective therapies. Here we have used both in vitro and in vivo approaches to show that the let-7 microRNA directly represses cancer growth in the lung. We find that let-7 inhibits the growth of multiple human lung cancer cell lines in culture, as well as the growth of lung cancer cell xenografts in immunodeficient mice. Using an established orthotopic mouse lung cancer model, we show that intranasal let-7 administration reduces tumor formation in vivo in the lungs of animals expressing a G12D activating mutation for the K-ras oncogene. These findings provide direct evidence that let-7 acts as a tumor suppressor gene in the lung and indicate that this miRNA may be useful as a novel therapeutic agent in lung cancer.


Subject(s)
Genes, Tumor Suppressor , Genetic Therapy/methods , Lung Neoplasms/therapy , MicroRNAs/therapeutic use , Adenoviridae , Administration, Intranasal , Animals , Cell Line, Tumor , Cell Proliferation , Genetic Vectors , Humans , Lung Neoplasms/genetics , Mice , MicroRNAs/administration & dosage
7.
Infect Immun ; 75(11): 5175-84, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17724066

ABSTRACT

Bacillus anthracis protective antigen (PA) is an 83-kDa (PA83) protein that is cleaved to the 63-kDa protein (PA63) as an essential step in binding and internalizing lethal factor (LF). To assess in vivo receptor saturating PA concentrations, we injected mice with PA variants and measured the PA remaining in the blood at various times using PA83- and PA63-specific enzyme-linked immunosorbent assays. We found that both wild-type PA (WT-PA) and a receptor-binding-defective mutant (Ub-PA) were cleaved to PA63 independent of their ability to bind cells. This suggested a PA-acting protease activity in the blood. The protease cleaved PA at the furin cleavage sequence because furin site-modified PA mutants were not cleaved. Cleavage measured in vitro was leupeptin sensitive and dependent on calcium. Cell surface cleavage was important for toxin clearance, however, as Ub-PA and uncleavable PA mutants were cleared at slower rates than WT-PA. The cell binding-independent cleavage of PA was also verified by using Ub-PA (which is still cleaved) to rescue mice from toxin challenge by competitively binding circulating LF. This mutant was able to rescue mice even when given 12 h before toxin challenge. Its therapeutic ability was comparable to that of dominant-negative PA, which binds cells but does not allow LF translocation, and to the protection afforded through receptor clearance by WT-PA and uncleavable receptor binding-competent mutants. The PA cleavage and clearance observed in mice did not appear to have a role in the differential mouse susceptibility as it occurred similarly in lethal toxin (LT)-resistant DBA/2J and LT-sensitive BALB/cJ mice. Interestingly, PA63 was not found in LT-resistant or -sensitive rats and PA83 clearance was slower in rats than in mice. Finally, to determine the minimum amount of PA required in circulation for LT toxicity in mice, we administered time-separated injections of PA and LF and showed that lethality of LF for mice after PA was no longer measurable in circulation, suggesting active PA sequestration at tissue surfaces.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacokinetics , Blood/metabolism , Animals , Anthrax/prevention & control , Antidotes/pharmacology , Antigens, Bacterial/toxicity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/toxicity , Calcium/pharmacology , Coenzymes/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Leupeptins/pharmacology , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mutation , Peptide Hydrolases/metabolism , Protein Binding/genetics , Rats , Rats, Inbred F344 , Survival Analysis
8.
Infect Immun ; 75(5): 2120-5, 2007 May.
Article in English | MEDLINE | ID: mdl-17339348

ABSTRACT

Anthrax toxin is made up of three separate protein components: the receptor-binding protective antigen (PA), the adenylyl cyclase edema factor (EF), and the metalloproteinase lethal factor (LF). EF and PA constitute edema toxin (ET), which causes edema when injected subcutaneously. At higher doses, ET causes severe pathologies and death in BALB/cJ mice (A. M. Firoved et al., Am. J. Pathol. 167:1309-1320, 2005). A striking effect of ET at lethal doses is adrenal necrosis. Here we show that low doses of ET (10 microg) that produce no overt signs of illness in mice still cause substantial adrenal lesions. These lesions are not associated with reduced corticosterone production; instead, ET-treated mice have increased corticosterone production. Because the resistance of mice to the other component of anthrax toxin, lethal toxin (LT; LF plus PA), has been shown to be overcome by the perturbation of the endocrine system, we hypothesized that sublethal doses of ET might sensitize LT-resistant DBA/2J mice to LT-mediated lethality. We report that a low dose of ET (5 microg) is sufficient to sensitize DBA/2J mice when given concurrently with LT. Higher doses of ET (e.g., 15 microg) given to male and female DBA/2J mice 18 h prior to LT challenge also sensitize them to LT. This study using highly purified ET and LT demonstrates how the components of anthrax toxin can work together to increase lethality.


Subject(s)
Adenylyl Cyclases/toxicity , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Adrenal Glands/pathology , Animals , Antigens, Bacterial/administration & dosage , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/administration & dosage , Corticosterone/blood , Edema/etiology , Female , Male , Metalloproteases/chemistry , Metalloproteases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred DBA
9.
Antimicrob Agents Chemother ; 50(8): 2658-65, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16870755

ABSTRACT

Bacillus anthracis lethal toxin (LT) produces symptoms of anthrax in mice and induces rapid lysis of macrophages derived from certain inbred strains. LT is comprised of a receptor binding component, protective antigen (PA), which delivers the enzymatic component, lethal factor (LF), into cells. We found that mouse macrophages were protected from toxin by the antitumor drug cis-diammineplatinum (II) dichloride (cisplatin). Cisplatin was shown to inhibit LT-mediated cleavage of cellular mitogen-activated protein kinases (MEKs) without inhibiting LF's in vitro proteolytic activity. Cisplatin-treated PA lost 100% of its ability to function in toxicity assays when paired with untreated LF, despite maintaining the ability to bind to cells. Cisplatin-treated PA was unable to form heptameric oligomers required for LF binding and translocation. The drug was shown to modify PA in a reversible noncovalent manner. Not surprisingly, cisplatin also blocked the actions of anthrax edema toxin and of LF-Pseudomonas aeruginosa exotoxin A fusion peptide (FP59), both of which require PA for translocation. Treatment of BALB/cJ mice or Fischer F344 rats with cisplatin at biologically relevant concentrations completely protected the animals from a coadministered lethal dose of LT. However, treatment with cisplatin 2 hours before or after animals received a lethal bolus of toxin did not protect them.


Subject(s)
Antigens, Bacterial/toxicity , Antineoplastic Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/toxicity , Cisplatin/pharmacology , Exotoxins/antagonists & inhibitors , Exotoxins/toxicity , Animals , Anthrax/drug therapy , Antineoplastic Agents/administration & dosage , Cell Line , Cell Survival/drug effects , Cisplatin/administration & dosage , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Injections, Intravenous , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Rats , Rats, Inbred F344 , Time Factors
10.
Infect Immun ; 74(7): 3707-14, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16790743

ABSTRACT

Bacillus anthracis lethal toxin (LT) induces rapid lysis (<90 min) of murine macrophages from certain inbred strains. The mechanism for LT-induced cytolysis is currently unknown. We hypothesized that the ATP-activated macrophage P2X7 receptors implicated in nucleotide-mediated macrophage lysis could play a role in LT-mediated cytolysis and discovered that a potent P2X7 antagonist, oxidized ATP (o-ATP), protects macrophages against LT. Other P2X7 receptor antagonists, however, had no effect on LT function, while oxidized nucleotides, o-ADP, o-GTP, and o-ITP, which did not act as receptor ligands, provided protection. Cleavage of the LT substrates, the mitogen-activated protein kinases, was inhibited by o-ATP in RAW274.6 macrophages and CHO cells. We investigated the various steps in the intoxication pathway and found that binding of the protective-antigen (PA) component of LT to cells and the enzymatic proteolytic ability of the lethal factor (LF) component of LT were unaffected by o-ATP. Instead, the drug inhibited formation of the sodium dodecyl sulfate-resistant PA oligomer, which occurs in acidified endosomes, but did not prevent cell surface PA oligomerization, as evidenced by binding and translocation of LF to a protease-resistant intracellular location. We found that o-ATP also protected cells from anthrax edema toxin and diphtheria toxin, which also require an acidic environment for escape from endosomes. Confocal microscopy using pH-sensitive fluorescent dyes showed that o-ATP increased endosomal pH. Finally, BALB/cJ mice injected with o-ATP and LT were completely protected against lethality.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Antigens, Bacterial/toxicity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/toxicity , Macrophages/physiology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/physiology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Oxidation-Reduction
11.
Mol Cell Biol ; 26(10): 3835-41, 2006 May.
Article in English | MEDLINE | ID: mdl-16648478

ABSTRACT

The translation elongation factor 2 in eukaryotes (eEF-2) contains a unique posttranslationally modified histidine residue, termed diphthamide, which serves as the only target for diphtheria toxin and Pseudomonas aeruginosa exotoxin A. Diphthamide biosynthesis is carried out by five highly conserved proteins, Dph1 to Dph5, and an as-yet-unidentified amidating enzyme. The evolutionary conservation of the complex diphthamide biosynthesis pathway throughout eukaryotes implies a key role for diphthamide in normal cellular physiology. Of the proteins required for diphthamide synthesis, Dph3 is the smallest, containing only 82 residues. In addition to having a role in diphthamide biosynthesis, Dph3 is also involved in modulating the functions of the Elongator complex in yeast. To explore the physiological roles of Dph3 and to begin to investigate the function of diphthamide, we generated dph3 knockout mice and showed that dph3+/- mice are phenotypically normal, whereas dph3-/- mice, which lack the diphthamide modification on eEF-2, are embryonic lethal. Loss of both dph3 alleles causes a general delay in embryonic development accompanied by lack of allantois fusion to the chorion and increased degeneration and necrosis in neural tubes and is not compatible with life beyond embryonic day 11.5. The dph3-/- placentas also developed abnormally, showing a thinner labyrinth lacking embryonic erythrocytes and blood vessels. These results attest to the physiological importance of Dph3 in development. The biological roles of Dph3 are also discussed.


Subject(s)
Carrier Proteins/metabolism , Fetal Development/physiology , Histidine/analogs & derivatives , Proteins/metabolism , Adenosine Diphosphate/analysis , Adenosine Diphosphate/metabolism , Alleles , Amino Acid Sequence , Amino Acids, Acidic , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/physiology , Evolution, Molecular , Exons , Female , Fetal Development/genetics , Gene Deletion , Gene Targeting , Heterozygote , Histidine/biosynthesis , Homozygote , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Molecular Sequence Data , Pregnancy , Proteins/chemistry , Proteins/genetics , Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
12.
Infect Immun ; 74(2): 1266-72, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428776

ABSTRACT

Bacillus anthracis lethal toxin (LT) is a bipartite toxin composed of protective antigen (PA) and lethal factor (LF). Injection of LT produces clinical signs characteristic of anthrax infection, including pleural edema and vascular collapse in various animal models. We utilized the classic Miles leakage assay to quantify vascular leakage in mice. LT injected intradermally induced leakage as early as 15 to 25 min in some inbred mouse strains, but not in others, whereas PA or LF individually did not induce leakage. A third component of anthrax toxin, edema factor, did not induce leakage alone or with PA. Leakage was quantified in eight mouse strains, and no correlation was found between sensitivity to intradermal leakage and sensitivity to the lethality of systemically administered LT. The leakage could be inhibited by ketotifen, an inhibitor of mast cell degranulation, but not by azelastine, a histamine receptor 1 antagonist, or by ketanserin, a serotonin 5-HT2A receptor antagonist. LT was cytotoxic to MC/9 mast cells (in vitro) by 7 h after toxin treatment but did not induce histamine release from these cells. Mast cell-deficient mice exhibited the leakage event and had no increased resistance to systemic LT. Human umbilical vein endothelial cells were resistant to LT over 12 h, with only 20% of cells succumbing by 24 h, suggesting that endothelial cell killing is not the cause of the rapid LT-mediated leakage event. We describe here a ketotifen-sensitive vascular leakage event induced by LT which is the most rapid in vivo or in vitro LT-mediated effect reported to date.


Subject(s)
Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Capillary Permeability/drug effects , Histamine H1 Antagonists/pharmacology , Ketotifen/pharmacology , Skin/blood supply , Animals , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Endothelial Cells/drug effects , Histamine Release , Mast Cells , Mice , Mice, Inbred Strains , Species Specificity , Umbilical Veins
13.
Am J Pathol ; 167(5): 1309-20, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16251415

ABSTRACT

Bacillus anthracis edema toxin (ET), an adenylyl cyclase, is an important virulence factor that contributes to anthrax disease. The role of ET in anthrax pathogenesis is, however, poorly understood. Previous studies using crude toxin preparations associated ET with subcutaneous edema, and ET-deficient strains of B. anthracis showed a reduction in virulence. We report the first comprehensive study of ET-induced pathology in an animal model. Highly purified ET caused death in BALB/cJ mice at lower doses and more rapidly than previously seen with the other major B. anthracis virulence factor, lethal toxin. Observations of gross pathology showed intestinal intralumenal fluid accumulation followed by focal hemorrhaging of the ileum and adrenal glands. Histopathological analyses of timed tissue harvests revealed lesions in several tissues including adrenal glands, lymphoid organs, bone, bone marrow, gastrointestinal mucosa, heart, and kidneys. Concomitant blood chemistry analyses supported the induction of tissue damage. Several cytokines increased after ET administration, including granulocyte colony-stimulating factor, eotaxin, keratinocyte-derived cytokine, MCP-1/JE, interleukin-6, interleukin-10, and interleukin-1beta. Physiological measurements also revealed a concurrent hypotension and bradycardia. These studies detail the extensive pathological lesions caused by ET and suggest that it causes death due to multiorgan failure.


Subject(s)
Antigens, Bacterial/toxicity , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Adenylyl Cyclases/toxicity , Adrenal Glands/pathology , Animals , Bone Marrow/pathology , Bone and Bones/pathology , Cytokines/biosynthesis , Gastric Mucosa/pathology , Hemorrhage , Ileum/pathology , Intestinal Mucosa/pathology , Kidney/pathology , Lymphoid Tissue/pathology , Mice , Mice, Inbred BALB C , Multiple Organ Failure , Myocardium/pathology , Virulence Factors/toxicity
14.
Infect Immun ; 73(7): 4238-44, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15972515

ABSTRACT

Bacillus anthracis lethal toxin (LT) causes vascular collapse and high lethality in BALB/cJ mice, intermediate lethality in C57BL/6J mice, and no lethality in DBA/2J mice. We found that adrenalectomized (ADX) mice of all three strains had increased susceptibility to LT. The increased susceptibility of ADX-DBA/2J mice was not accompanied by changes in their macrophage sensitivity or cytokine response to LT. DBA/2J mice showed no change in serum corticosteroid levels in response to LT injection, while BALB/cJ mice showed a fivefold increase in serum corticosterone. However, LT inhibited dexamethasone (DEX)-induced glucocorticoid receptor gene activation to similar extents in all three strains. DEX treatment did not rescue ADX mice from LT-mediated mortality. Surprisingly, oral DEX treatment also sensitized adrenally intact DBA/2J mice to LT lethality at all doses tested and also exacerbated LT-mediated pathogenesis and mortality in BALB/cJ mice. Aldosterone did not protect ADX mice from toxin challenge. These results indicate that susceptibility to anthrax LT in mice depends on a fine but easily perturbed balance of endocrine functions. Thus, the potentially detrimental consequences of steroid therapy for anthrax must be considered in treatment protocols for this disease.


Subject(s)
Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Adrenalectomy , Aldosterone/therapeutic use , Animals , Corticosterone/blood , Cytokines/biosynthesis , Dexamethasone/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Receptors, Glucocorticoid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...