Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
PLoS Genet ; 14(9): e1007589, 2018 09.
Article in English | MEDLINE | ID: mdl-30188888

ABSTRACT

Canine malignant melanoma, a significant cause of mortality in domestic dogs, is a powerful comparative model for human melanoma, but little is known about its genetic etiology. We mapped the genomic landscape of canine melanoma through multi-platform analysis of 37 tumors (31 mucosal, 3 acral, 2 cutaneous, and 1 uveal) and 17 matching constitutional samples including long- and short-insert whole genome sequencing, RNA sequencing, array comparative genomic hybridization, single nucleotide polymorphism array, and targeted Sanger sequencing analyses. We identified novel predominantly truncating mutations in the putative tumor suppressor gene PTPRJ in 19% of cases. No BRAF mutations were detected, but activating RAS mutations (24% of cases) occurred in conserved hotspots in all cutaneous and acral and 13% of mucosal subtypes. MDM2 amplifications (24%) and TP53 mutations (19%) were mutually exclusive. Additional low-frequency recurrent alterations were observed amidst low point mutation rates, an absence of ultraviolet light mutational signatures, and an abundance of copy number and structural alterations. Mutations that modulate cell proliferation and cell cycle control were common and highlight therapeutic axes such as MEK and MDM2 inhibition. This mutational landscape resembles that seen in BRAF wild-type and sun-shielded human melanoma subtypes. Overall, these data inform biological comparisons between canine and human melanoma while suggesting actionable targets in both species.


Subject(s)
Melanoma/genetics , Melanoma/veterinary , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Skin Neoplasms/genetics , Skin Neoplasms/veterinary , Animals , Cell Cycle/genetics , Cell Proliferation/genetics , Comparative Genomic Hybridization , DNA Mutational Analysis , Dog Diseases/genetics , Dogs , Female , Male , Melanoma/blood , Melanoma/pathology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Signal Transduction/genetics , Skin Neoplasms/blood , Skin Neoplasms/pathology , Tissue Array Analysis
2.
Am J Cancer Res ; 5(4): 1507-18, 2015.
Article in English | MEDLINE | ID: mdl-26101714

ABSTRACT

Variable clinical responses, tumor heterogeneity, and drug resistance reduce long-term survival outcomes for metastatic melanoma patients. To guide and accelerate drug development, we characterized tumor responses for five melanoma patient derived xenograft models treated with Vemurafenib. Three BRAF(V600E) models showed acquired drug resistance, one BRAF(V600E) model had a complete and durable response, and a BRAF(V600V) model was expectedly unresponsive. In progressing tumors, a variety of resistance mechanisms to BRAF inhibition were uncovered, including mutant BRAF alternative splicing, NRAS mutation, COT (MAP3K8) overexpression, and increased mutant BRAF gene amplification and copy number. The resistance mechanisms among the patient derived xenograft models were similar to the resistance pathways identified in clinical specimens from patients progressing on BRAF inhibitor therapy. In addition, there was both inter- and intra-patient heterogeneity in resistance mechanisms, accompanied by heterogeneous pERK expression immunostaining profiles. MEK monotherapy of Vemurafenib-resistant tumors caused toxicity and acquired drug resistance. However, tumors were eradicated when Vemurafenib was combined the MEK inhibitor. The diversity of drug responses among the xenograft models; the distinct mechanisms of resistance; and the ability to overcome resistance by the addition of a MEK inhibitor provide a scheduling rationale for clinical trials of next-generation drug combinations.

3.
Int J Oncol ; 47(1): 71-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25955301

ABSTRACT

Angiosarcoma (AS) is a rare neoplasm of endothelial origin that has limited treatment options and poor five-year survival. Using tumorgraft models, we previously showed that AS is sensitive to small-molecule inhibitors that target mitogen-activated/extracellular-signal-regulated protein kinase kinases 1 and 2 (MEK). The objective of this study was to identify drugs that combine with MEK inhibitors to more effectively inhibit AS growth. We examined the in vitro synergy between the MEK inhibitor PD0325901 and inhibitors of eleven common cancer pathways in melanoma cell lines and canine angiosarcoma cell isolates. Combination indices were calculated using the Chou-Talalay method. Optimized combination therapies were evaluated in vivo for toxicity and efficacy using canine angiosarcoma tumorgrafts. Among the drugs we tested, rapamycin stood out because it showed strong synergy with PD0325901 at nanomolar concentrations. We observed that angiosarcomas are insensitive to mTOR inhibition. However, treatment with nanomolar levels of mTOR inhibitor renders these cells as sensitive to MEK inhibition as a melanoma cell line with mutant BRAF. Similar results were observed in B-Raf wild-type melanoma cells as well as in vivo, where treatment of canine AS tumorgrafts with MEK and mTOR inhibitors was more effective than monotherapy. Our data show that a low dose of an mTOR inhibitor can dramatically enhance angiosarcoma and melanoma response to MEK inhibition, potentially widening the field of applications for MEK-targeted therapy.


Subject(s)
Benzamides/administration & dosage , Diphenylamine/analogs & derivatives , Hemangiosarcoma/drug therapy , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols , Benzamides/pharmacology , Cell Line, Tumor , Diphenylamine/administration & dosage , Diphenylamine/pharmacology , Dogs , Dose-Response Relationship, Drug , Drug Synergism , Humans , Mice , Sirolimus/pharmacology , Xenograft Model Antitumor Assays
4.
Am J Med Genet A ; 167(7): 1429-35, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808193

ABSTRACT

Oculoectodermal syndrome (OES) is a rare disease characterized by a combination of congenital scalp lesions and ocular dermoids, with additional manifestations including non-ossifying fibromas and giant cell granulomas of the jaw occurring during the first decade of life. To identify the genetic etiology of OES, we conducted whole-genome sequencing of several tissues in an affected individual. Comparison of DNA from a non-ossifying fibroma to blood-derived DNA allowed identification of a somatic missense alteration in KRAS NM_033360.3(KRAS):c.38G>A, resulting in p.Gly13Asp. This alteration was also observed in the patient's other affected tissues including the skin and muscle. Targeted sequencing in a second, unrelated OES patient identified an NM_033360.3(KRAS):c.57G>C, p.Leu19Phe alteration. Allelic frequencies fell below 40% in all tissues examined in both patients, suggesting that OES is a mosaic RAS-related disorder, or RASopathy. The characteristic findings in OES, including scalp lesions, ocular dermoids, and benign tumors, are found in other mosaic and germline RASopathies. This discovery also broadens our understanding of the spectrum of phenotypes resulting from KRAS alterations. Future research into disease progression with regard to malignancy risk and investigation of RAS-targeted therapies in OES is warranted. KRAS sequencing is clinically available and may also now improve OES diagnostic criteria.


Subject(s)
Dermoid Cyst/genetics , Dermoid Cyst/pathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Genome, Human/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Base Sequence , Child , Child, Preschool , Choristoma/pathology , Corneal Diseases/pathology , Female , Gene Frequency , Growth Disorders/pathology , Humans , Male , Molecular Sequence Data , Mutation, Missense/genetics , Scalp/pathology , Sequence Analysis, DNA
5.
Invest Ophthalmol Vis Sci ; 54(10): 6472-80, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24003089

ABSTRACT

PURPOSE: To identify inflammatory cytokines significantly elevated and independent of VEGF levels in the vitreous of proliferative diabetic retinopathy (PDR) patients that may serve as novel diagnostic factors or therapeutic targets. METHODS: Thirty-nine cytokines and chemokines were measured from the vitreous of 72 patients undergoing vitrectomy (29 controls and 43 PDR) via a magnetic bead-based immunoassay. Patient information, including sex, age, history of smoking, cancer diagnosis and treatment, and presence of diabetes and hypertension were also collected. Univariate and multivariate logistic regression analyses were performed to assess the association of cytokine concentrations and patient demographics with disease. RESULTS: Nineteen cytokines were significantly elevated in the vitreous of PDR patients compared with controls, including five novel cytokines that have not previously been associated with PDR: sCD40L, GM-CSF, IFNα2, IL-12p40, and MCP-3. Sixteen cytokines were found to be statistically independent of VEGF. Of these, 14 show a statistically significant interaction with VEGF, while two do not. With regards to patient demographics, age and hypertension were statistically significant risk factors with the odds of disease decreasing with increasing age and increasing 3-fold for hypertensive patients. CONCLUSIONS: This is the first report of a comprehensive multiplex analysis to identify novel VEGF-independent cytokines associated with PDR. Of the 39 inflammatory cytokines tested, 16 are predictive of disease risk, independent of VEGF levels. These PDR-associated cytokines represent potential targets in the treatment of PDR, both in conjunction with anti-VEGF therapy, as well as for patients that are nonresponders to such therapy.


Subject(s)
Cytokines/metabolism , Diabetic Retinopathy/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vitreoretinopathy, Proliferative/metabolism , Vitreous Body/chemistry , Aged , Aged, 80 and over , Biomarkers/metabolism , Diabetic Retinopathy/complications , Diabetic Retinopathy/surgery , Female , Humans , Male , Middle Aged , Vitrectomy , Vitreoretinopathy, Proliferative/etiology , Vitreoretinopathy, Proliferative/surgery
6.
Clin Dev Immunol ; 2013: 475809, 2013.
Article in English | MEDLINE | ID: mdl-23861692

ABSTRACT

Human anthrax infection caused by exposure to Bacillus anthracis cannot always be treated by antibiotics. This is mostly because of the effect of the remaining anthrax toxin in the body. Lethal factor (LF) is a component of lethal toxin (LeTx), which is the major virulence of anthrax toxin. A murine IgG monoclonal antibody (mAb) against LF with blocking activity (coded LF8) was produced in a previous study. In this report, a human/murine chimeric Fab mAb (coded LF8-Fab) was developed from LF8 by inserting murine variable regions into human constant regions using antibody engineering to reduce the incompatibility of the murine antibody for human use. The LF8-Fab expressed in Escherichia coli could specifically identify LF with an affinity of 3.46 × 10(7) L/mol and could neutralize LeTx with an EC50 of 85 µ g/mL. Even after LeTx challenge at various time points, the LF8-Fab demonstrated protection of J774A.1 cells in vitro. The results suggest that the LF8-Fab might be further characterized and potentially be used for clinical applications against anthrax infection.


Subject(s)
Antibodies, Neutralizing/pharmacology , Bacillus anthracis/drug effects , Bacterial Toxins/antagonists & inhibitors , Immunoglobulin Fab Fragments/pharmacology , Macrophages/drug effects , Amino Acid Sequence , Animals , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibody Affinity , Antibody Specificity , Antigens, Bacterial/immunology , Bacillus anthracis/growth & development , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Base Sequence , Cell Line , Escherichia coli/genetics , Gene Expression , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Molecular Sequence Data , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology
7.
Mol Cancer Ther ; 12(9): 1701-14, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804705

ABSTRACT

Angiosarcoma is a rare neoplasm of endothelial origin that has limited treatment options and poor five-year survival. As a model for human angiosarcoma, we studied primary cells and tumorgrafts derived from canine hemangiosarcoma (HSA), which is also an endothelial malignancy with similar presentation and histology. Primary cells isolated from HSA showed constitutive extracellular signal-regulated kinase (ERK) activation. The mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitor CI-1040 reduced ERK activation and the viability of primary cells derived from visceral, cutaneous, and cardiac HSA in vitro. HSA-derived primary cells were also sensitive to sorafenib, an inhibitor of B-Raf and multireceptor tyrosine kinases. In vivo, CI-1040 or PD0325901 decreased the growth of cutaneous cell-derived xenografts and cardiac-derived tumorgrafts. Sorafenib decreased tumor size in both in vivo models, although cardiac tumorgrafts were more sensitive. In human angiosarcoma, we noted that 50% of tumors stained positively for phosphorylated ERK1/2 and that the expression of several MEK-responsive transcription factors was upregulated. Our data showed that MEK signaling is essential for the growth of HSA in vitro and in vivo and provided evidence that the same pathways are activated in human angiosarcoma. This indicates that MEK inhibitors may form part of an effective therapeutic strategy for the treatment of canine HSA or human angiosarcoma, and it highlights the use of spontaneous canine cancers as a model of human disease.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Proliferation/drug effects , Diphenylamine/analogs & derivatives , Hemangiosarcoma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Diphenylamine/pharmacology , Disease Models, Animal , Dogs , Drug Screening Assays, Antitumor , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Hemangiosarcoma/drug therapy , Hemangiosarcoma/metabolism , Hemangiosarcoma/veterinary , Humans , Mice , Mice, Nude , Niacinamide/pharmacology , Signal Transduction/drug effects , Sorafenib , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Brief Funct Genomics ; 11(4): 300-10, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22753777

ABSTRACT

The mitogen-activated protein kinase kinases (the MAPK/ERK kinases; MKKs or MEKs) and their downstream substrates, the extracellular-regulated kinases have been intensively studied for their roles in development and disease. Until recently, it had been assumed any mutation affecting their function would have lethal consequences. However, the identification of MEK1 and MEK2 mutations in developmental syndromes as well as chemotherapy-resistant tumors, and the discovery of genomic variants in MEK1 and MEK2 have led to the realization the extent of genomic variation associated with MEKs is much greater than had been appreciated. In this review, we will discuss these recent advances, relating them to what is currently understood about the structure and function of MEKs, and describe how they change our understanding of the role of MEKs in development and disease.


Subject(s)
Ectodermal Dysplasia/genetics , Failure to Thrive/genetics , Genomics , Heart Defects, Congenital/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mutation , Animals , Cluster Analysis , Facies , Genetic Variation , Humans , Mice , Models, Genetic , Neoplasms/genetics , Neoplasms/metabolism , Polymorphism, Single Nucleotide , Protein Conformation , Protein Structure, Tertiary
9.
Invest Ophthalmol Vis Sci ; 52(12): 8979-92, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-21989728

ABSTRACT

PURPOSE: To evaluate the role of mitogen-activated protein kinase kinase (MKK) signaling in a mouse model of oxygen-induced retinopathy (OIR) that mimics retinopathy of prematurity (ROP). METHODS: Postnatal day 7 mice were exposed to elevated oxygen for 5 days to induce retinopathy. Anthrax lethal toxin (LeTx), an MKK inhibitor, was injected into the vitreous after restoration to normoxia, and its effects on vascular growth were analyzed by whole mount immunofluorescence and confocal microscopy. Pericyte coverage was determined by PDGFR-ß and α-SMA staining. Macrophage presence was determined by F4/80 staining. Vitreal cytokine secretion was measured by ELISA and multianalyte profiling. RESULTS: Intravitreal injection of LeTx over a restricted time interval after return to normoxic conditions blocked the progression of OIR. This block was independent of vascular endothelial growth factor (VEGF) release and did not alter the release of cytokines and growth factors associated with OIR. VEGFR2 expression and activation were similarly unaffected. LeTx had no statistically significant effect on macrophage recruitment. LeTx sensitivity correlated with vessel maturity, extent of hypoxia, and growth of the deep vascular plexus network. CONCLUSIONS: Correlation among pericyte coverage, deep vascular plexus growth, and hypoxia after LeTx treatment indicate immature vessels in a hypoxic environment are preferentially sensitive to LeTx-mediated MKK inhibition. The persistence of VEGF without concomitant induction of neovascular growth or revascularization of vaso-obliterated zones suggests MKK inhibition causes an inability of the cells that are present, or a failure to recruit cells able, to respond to proangiogenic stimuli. These results indicate the inhibition of MKK signaling presents a novel strategy for the inhibition of vascular retinopathies such as OIR and ROP.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Oxygen/adverse effects , Retinal Neovascularization/drug therapy , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/prevention & control , Angiopoietins/metabolism , Animals , Animals, Newborn , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells , Humans , Infant, Newborn , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/metabolism , Retinal Neovascularization/metabolism , Retinopathy of Prematurity/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
PLoS One ; 6(2): e17165, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21365009

ABSTRACT

Mitogen-activated protein kinase kinases (MKK or MEK) 1 and 2 are usually treated as redundant kinases. However, in assessing their relative contribution towards ERK-mediated biologic response investigators have relied on tests of necessity, not sufficiency. In response we developed a novel experimental model using lethal toxin (LeTx), an anthrax toxin-derived pan-MKK protease, and genetically engineered protease resistant MKK mutants (MKKcr) to test the sufficiency of MEK signaling in melanoma SK-MEL-28 cells. Surprisingly, ERK activity persisted in LeTx-treated cells expressing MEK2cr but not MEK1cr. Microarray analysis revealed non-overlapping downstream transcriptional targets of MEK1 and MEK2, and indicated a substantial rescue effect of MEK2cr on proliferation pathways. Furthermore, LeTx efficiently inhibited the cell proliferation and anchorage-independent growth of SK-MEL-28 cells expressing MKK1cr but not MEK2cr. These results indicate in SK-MEL-28 cells MEK1 and MEK2 signaling pathways are not redundant and interchangeable for cell proliferation. We conclude that in the absence of other MKK, MEK2 is sufficient for SK-MEL-28 cell proliferation. MEK1 conditionally compensates for loss of MEK2 only in the presence of other MKK.


Subject(s)
Cell Proliferation , MAP Kinase Kinase 2/physiology , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Antigens, Bacterial/metabolism , Antigens, Bacterial/pharmacology , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , CHO Cells , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Proliferation/drug effects , Cluster Analysis , Cricetinae , Cricetulus , Gene Expression Profiling , Humans , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Melanoma/genetics , Microarray Analysis , Neoplasm Invasiveness , Point Mutation/physiology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Skin Neoplasms/genetics , Tumor Cells, Cultured
11.
Mol Cancer Ther ; 9(1): 190-201, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20053778

ABSTRACT

Patients with anaplastic thyroid carcinoma (ATC) typically succumb to their disease months after diagnosis despite aggressive therapy. A large percentage of ATCs have been shown to harbor the V600E B-Raf point mutation, leading to the constitutive activation of the mitogen-activated protein kinase pathway. ATC invasion, metastasis, and angiogenesis are in part dependent on the gelatinase class of matrix metalloproteinases (MMP). The explicit targeting of these two tumor markers may provide a novel therapeutic strategy for the treatment of ATC. The MMP-activated anthrax lethal toxin (LeTx), a novel recombinant protein toxin combination, shows potent mitogen-activated protein kinase pathway inhibition in gelatinase-expressing V600E B-Raf tumor cells in vitro. However, preliminary in vivo studies showed that the MMP-activated LeTx also exhibited dramatic antitumor activity against xenografts that did not show significant antiproliferative responses to the LeTx in vitro. Here, we show that the MMP-activated LeTx inhibits orthotopic ATC xenograft progression in both toxin-sensitive and toxin-resistant ATC cells via reduced endothelial cell recruitment and subsequent tumor vascularization. This in turn translates to an improved long-term survival that is comparable with that produced by the multikinase inhibitor sorafenib. Our results also indicate that therapy with the MMP-activated LeTx is extremely effective against advanced tumors with well-established vascular networks. Taken together, these results suggest that the MMP-activated LeTx-mediated endothelial cell targeting is the primary in vivo antitumor mechanism of this novel toxin. Therefore, the MMP-activated LeTx could be used not only in the clinical management of V600E B-Raf ATC but potentially in any solid tumor.


Subject(s)
Antigens, Bacterial/therapeutic use , Bacterial Toxins/therapeutic use , Carcinoma/blood supply , Matrix Metalloproteinases/metabolism , Neovascularization, Pathologic/drug therapy , Thyroid Neoplasms/blood supply , Xenograft Model Antitumor Assays , Animals , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Benzenesulfonates/pharmacology , Carcinoma/drug therapy , Carcinoma/enzymology , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Endocytosis/drug effects , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/pharmacology , Sorafenib , Survival Analysis , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Time Factors
12.
PLoS One ; 4(9): e6956, 2009 Sep 14.
Article in English | MEDLINE | ID: mdl-19750016

ABSTRACT

Lethal factor, the enzymatic moiety of anthrax lethal toxin (LeTx) is a protease that inactivates mitogen activated protein kinase kinases (MEK or MKK). In vitro and in vivo studies demonstrate LeTx targets endothelial cells. However, the effects of LeTx on endothelial cells are incompletely characterized. To gain insight into this process we used a developmental model of vascularization in the murine retina. We hypothesized that application of LeTx would disrupt normal retinal vascularization, specifically during the angiogenic phase of vascular development. By immunoblotting and immunofluorescence microscopy we observed that MAPK activation occurs in a spatially and temporally regulated manner during retinal vascular development. Intravitreal administration of LeTx caused an early delay (4 d post injection) in retinal vascular development that was marked by reduced penetration of vessels into distal regions of the retina as well as failure of sprouting vessels to form the deep and intermediate plexuses within the inner retina. In contrast, later stages (8 d post injection) were characterized by the formation of abnormal vascular tufts that co-stained with phosphorylated MAPK in the outer retinal region. We also observed a significant increase in the levels of secreted VEGF in the vitreous 4 d and 8 d after LeTx injection. In contrast, the levels of over 50 cytokines other cytokines, including bFGF, EGF, MCP-1, and MMP-9, remained unchanged. Finally, co-injection of VEGF-neutralizing antibodies significantly decreased LeTx-induced neovascular growth. Our studies not only reveal that MAPK signaling plays a key role in retinal angiogenesis but also that perturbation of MAPK signaling by LeTx can profoundly alter vascular morphogenesis.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Retina/drug effects , Retinal Vessels/drug effects , Animals , Anthrax/metabolism , Anthrax/physiopathology , Immunoblotting , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Morphogenesis/drug effects , Neovascularization, Physiologic , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/metabolism
13.
Mol Cancer Res ; 7(4): 452-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19372576

ABSTRACT

Solid tumor growth is dependent on angiogenesis, the formation of neovasculature from existing vessels. Endothelial activation of the extracellular signal-regulated kinase 1/2, c-jun NH(2)-terminal kinase, and p38 mitogen-activated protein kinase pathways is central to this process, and thus presents an attractive target for the development of angiogenesis inhibitors. Anthrax lethal toxin (LeTx) has potent catalytic mitogen-activated protein kinase inhibition activity. Preclinical studies showed that LeTx induced potent tumor growth inhibition via the inhibition of xenograft vascularization. However, LeTx receptors and the essential furin-like activating proteases are expressed in many normal tissues, potentially limiting the specificity of LeTx as an antitumor agent. To circumvent nonspecific LeTx activation and simultaneously enhance tumor vascular targeting, a substrate preferably cleaved by the gelatinases class of matrix metalloproteinases (MMP) was substituted for the furin LeTx activation site. In vivo efficacy studies showed that this MMP-activated LeTx inhibited tumor xenografts growth via the reduced migration of endothelial cells into the tumor parenchyma. Here we have expanded on these initial findings by showing that this MMP-activated LeTx reduces endothelial proangiogenic MMP expression, thus causing a diminished proteolytic capacity for extracellular matrix remodeling and endothelial differentiation into capillary networks. Additionally, our data suggest that inhibition of the c-jun NH(2)-terminal kinase and p38, but not extracellular signal-regulated kinase-1/2, pathways is significant in the antiangiogenic activity of the MMP-activated LeTx. Collectively, these results support the clinical development of the MMP-activated LeTx for the treatment of solid tumors.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Endothelium, Vascular/drug effects , Matrix Metalloproteinases/metabolism , Neovascularization, Pathologic/prevention & control , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Front Biosci (Landmark Ed) ; 14(12): 4516-24, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273366

ABSTRACT

Infection by Bacillus anthracis in animals and humans results from accidental or intentional exposure, by oral, cutaneous or pulmonary routes, to spores, which are normally present in the soil. Treatment includes administration of antibiotics, vaccination or treatment with antibody to the toxin. A better understanding of the molecular basis of the processes involved in the pathogenesis of anthrax namely, spore germination in macrophages and biological effects of the secreted toxins on heart and blood vessels will lead to improved management of infected animals and patients. Controlling germination will be feasible by inhibiting macrophage paralysis and cell death. On the other hand, the control of terminal hypotension might be achieved by inhibition of cardiomyocyte mitogen-activated protein kinase and stimulation of vessel cAMP.


Subject(s)
Anthrax/physiopathology , Antigens, Bacterial/toxicity , Bacillus anthracis/growth & development , Bacterial Toxins/toxicity , Blood Pressure/drug effects , Hemodynamics/drug effects , Humans , Spores, Bacterial
15.
Mol Cancer Ther ; 7(5): 1218-26, 2008 May.
Article in English | MEDLINE | ID: mdl-18483309

ABSTRACT

Anthrax lethal toxin (LeTx) shows potent mitogen-activated protein kinase pathway inhibition and apoptosis in melanoma cells that harbor the activating V600E B-RAF mutation. LeTx is composed of two proteins, protective antigen and lethal factor. Uptake of the toxin into cells is dependent on proteolytic activation of protective antigen by the ubiquitously expressed furin or furin-like proteases. To circumvent nonspecific LeTx activation, a substrate preferably cleaved by gelatinases was substituted for the furin LeTx activation site. Here, we have shown that the toxicity of this matrix metalloproteinase (MMP)-activated LeTx is dependent on host cell surface MMP-2 and MMP-9 activity as well as the presence of the activating V600E B-RAF mutation, making this toxin dual specific. This additional layer of tumor cell specificity would potentially decrease systemic toxicity from the reduction of nonspecific toxin activation while retaining antitumor efficacy in patients with V600E B-RAF melanomas. Moreover, our results indicate that cell surface-associated gelatinase expression can be used to predict sensitivity among V600E B-RAF melanomas. This finding will aid in the better selection of patients that will potentially respond to MMP-activated LeTx therapy.


Subject(s)
Antigens, Bacterial/toxicity , Antineoplastic Agents/toxicity , Bacterial Toxins/toxicity , Gelatinases/metabolism , Matrix Metalloproteinases/metabolism , Melanoma/enzymology , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Cell Line, Tumor , Humans , Melanoma/metabolism , Mutation , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured
16.
Methods Enzymol ; 438: 355-65, 2008.
Article in English | MEDLINE | ID: mdl-18413261

ABSTRACT

The secretion of factors that block critical intracellular signaling pathways is a common strategy used by pathogenic bacteria for disabling host defenses and causing disease. Anthrax lethal toxin (LeTx) has been shown to cleave and inactivate mitogen-activated protein kinase (MAPK) kinases (MKKs or MEKs) and to inhibit MKK signaling. Cleavage of MKKs by LeTx prevents activation of their downstream substrates, the MAPKs. Because MAPK pathways regulate a variety of crucial cellular functions including proliferation, survival, differentiation, adhesion, and motility, LeTx has become a focus of study as an investigative tool as well as for the treatment and prevention of diseases due to malfunctions in MAPK signaling. This chapter describes methods for expressing and purifying the components of LeTx and focuses on techniques available for assessing its activity.


Subject(s)
Antigens, Bacterial/analysis , Bacterial Toxins/analysis , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Animals , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Cell Survival/drug effects , Female , Immunoblotting , Mice , Mitogen-Activated Protein Kinase Kinases/analysis , Mitogen-Activated Protein Kinase Kinases/immunology , Oocytes/drug effects , Xenopus laevis
17.
Mol Cancer Ther ; 7(3): 648-58, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18319331

ABSTRACT

We hypothesized that signaling through multiple mitogen-activated protein kinase (MAPK) kinase (MKK) pathways is essential for the growth and vascularization of soft-tissue sarcomas, which are malignant tumors derived from mesenchymal tissues. We tested this using HT-1080, NCI, and Shac fibrosarcoma-derived cell lines and anthrax lethal toxin (LeTx), a bacterial toxin that inactivates MKKs. Western blots confirmed that LeTx treatment reduced the levels of phosphorylated extracellular signal-regulated kinase and p38 MAPK in vitro. Although short treatments with LeTx only modestly affected cell proliferation, sustained treatment markedly reduced cell numbers. LeTx also substantially inhibited the extracellular release of angioproliferative factors including vascular endothelial growth factor, interleukin-8, and basic fibroblast growth factor. Similar results were obtained with cell lines derived from malignant fibrous histiocytomas, leiomyosarcomas, and liposarcomas. In vivo, LeTx decreased MAPK activity and blocked fibrosarcoma growth. Growth inhibition correlated with decreased cellular proliferation and extensive necrosis, and it was accompanied by a decrease in tumor mean vessel density as well as a reduction in serum expression of angioproliferative cytokines. Vital imaging using high-resolution ultrasound enhanced with contrast microbubbles revealed that the effects of LeTx on tumor perfusion were remarkably rapid (<24 h) and resulted in a marked reduction of perfusion within the tumor but not in nontumor tissues. These results are consistent with our initial hypothesis and lead us to propose that MKK inhibition by LeTx is a broadly effective strategy for targeting neovascularization in fibrosarcomas and other similar proliferative lesions.


Subject(s)
Cell Division , Fibrosarcoma/blood supply , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic , Signal Transduction , Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Fibrosarcoma/pathology , Humans
18.
Int J Oncol ; 32(4): 739-48, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18360701

ABSTRACT

Two of the three components of anthrax toxin, protective antigen (PA) and lethal factor (LF), together known as lethal toxin (LeTx), reportedly show anti-tumor activity in melanoma in vitro and in vivo. The growth inhibitory activity of LeTx in culture was determined in nine human cancer cell lines, including melanoma, neuroblastoma and adenocarcinoma cells, as well as in human umbilical vein endothelial cells (HUVEC). The contribution of the two known PA receptor proteins, ANTXR1/TEM8 and ANTXR2/CMG2, to the sensitivity of the cells was assessed. The efficacy of LeTx was evaluated in vivo in the SK-N-AS neuroblastoma and SK-MEL-28 melanoma tumor xenograft models. Sensitivity to LeTx in vitro was observed in the neuroblastoma and colorectal adenocarcinoma cells and HUVEC, as well as melanoma cells. ANTXR1/TEM8 and ANTXR2/CMG2 protein expression studies suggested that a certain threshold of the PA receptor protein level must be met for sensitivity to LeTx to be observed. However, although the SK-N-AS neuroblastoma cells expressed the highest levels of receptor proteins and achieved the lowest IC50 in vitro (0.1 ng/ml), we observed no correlation between either the ANTXR1/TEM8 or ANTXR2/CMG2 protein levels and sensitivity to LeTx in vitro. In vivo, LeTx was an active anti-tumor agent when administered intravenously to mice bearing the human SK-N-AS or SK-MEL-28 tumor xenografts. The tumor growth delays were 6-8 days with a lower dose regimen and 14-16 days with a higher dose regimen for the two tumor models. These in vitro data suggest that LeTx may have broad therapeutic indications in cancer and the in vivo studies demonstrate that LeTx has systemic efficacy in neuroblastoma as well as melanoma. The therapeutic potential of LeTx needs to be further investigated in non-melanoma tumor models expressing the ANTXR1/TEM8 and/or ANTXR2/CMG2 protein.


Subject(s)
Antigens, Bacterial/therapeutic use , Bacterial Toxins/therapeutic use , Melanoma, Experimental/drug therapy , Neuroblastoma/drug therapy , Receptors, Peptide/analysis , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Female , Humans , Male , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
19.
Cell Cycle ; 7(6): 745-9, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18245947

ABSTRACT

Angiogenesis is a critical step in solid tumor progression. The mitogen-activated protein kinase (MAPK) signaling pathways are central to this process, and thus present attractive targets for angiogenesis inhibition. Anthrax Lethal Toxin (LeTx), secreted from the gram positive Bacillus anthracis, demonstrates potent MAPK pathway inhibition. In vivo efficacy studies revealed that LeTx has broad anti-tumor efficacy via the targeting of angiogenesis. However, specificity in animal models was limited due to the presence of receptors on many normal tissues and the ubiquitous expression of furin in tissues. Further, half-life of LeTx was short due to circulating furin-like proteases. Gelatinases are expressed on tumor angiogenic sprouts and only to a limited extent in normal tissues or blood. In order to circumvent nonspecific LeTx activation, enhance tumor vascular targeting, and improve plasma half-life, a substrate preferably cleaved by gelatinases was substituted for the furin LeTx activation site. The MMP-activated LeTx showed potent angiogenic inhibition in vivo in the absence of systemic toxicity. Based on these studies, this attenuated toxin has clinical potential as a broad anti-tumor agent.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Matrix Metalloproteinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Signal Transduction/drug effects , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Drug Delivery Systems/methods , Humans
20.
Cancer Res ; 68(1): 81-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172299

ABSTRACT

The mitogen-activated protein kinase (MAPK) signaling pathways play essential roles in cell proliferation and differentiation. Recent studies also show the activation of MAPK signaling pathways in tumorigenesis, metastasis, and angiogenesis of multiple human malignancies, including renal cell carcinoma (RCC). To assess the role of this pathway in regulating the proliferation and survival of RCC cells, we first examined the expression of MAPK kinase (MKK) and MAPK in clear cell RCC and confirmed the overexpression of MKK1 and extracellular signal-regulated kinase 2 (ERK2) in these tumors. We then tested the effects of pharmacologic inhibition of MKK on human RCC cell lines, both in vitro and in vivo, using anthrax lethal toxin (LeTx), which cleaves and inactivates several MKKs. Western blotting showed that the phosphorylation levels of ERK, c-Jun-NH(2) kinase, and p38 MAPK decreased after 72 h of LeTx treatment. Exposure to LeTx for 72 h reduced cell proliferation by 20% without significant effects on cell cycle distribution and apoptosis. Anchorage-independent growth of RCC cells was dramatically inhibited by LeTx. In vivo studies showed that tumor growth of RCC xenografts could be suppressed by LeTx. Extensive necrosis and decreased tumor neovascularization were observed after LeTx treatment. LeTx also showed direct inhibition of proliferation of endothelial cells in vitro. Our results suggest that suppression of one or more MAPK signaling pathways may inhibit RCC growth through the disruption of tumor vasculature.


Subject(s)
Antigens, Bacterial/pharmacology , Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/blood supply , Kidney Neoplasms/enzymology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neovascularization, Pathologic/enzymology , Animals , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Kidney Neoplasms/pathology , Mice , Mice, Inbred Strains , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...