1
|
Methods for Evaluation of a Snake Venom-Derived Disintegrin in Animal Models of Human Cancer. Methods Mol Biol 2020; 2068:185-204. [PMID: 31576529 DOI: 10.1007/978-1-4939-9845-6_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Integrin targeting has been shown to be an effective approach for anticancer therapy. We engineered a recombinant disintegrin, vicrostatin (VCN), that binds with high affinity and specificity to the Arg-Gly-Asp (RGD) class of integrins, including αvβ3, αvβ5, and α5β1, involved in tumor invasion and metastasis. We used three different delivery modalities to examine anticancer activity of VCN in mouse models of human ovarian cancer, glioma, and prostate cancer. A female mouse model was used to examine the treatment of established ovarian cancer (OC) using VCN delivered intraperitoneally (IP) weekly either in saline or impregnated in a viscoelastic gel. SKOV3luc cells (a human OC cell line) were directly injected IP into immunodeficient mice. We also examined the antitumor activity of radioiodinated VCN delivered intravenously in a human glioma model in nude mice. We evaluated the effectiveness of 131I-VCN in combination with the DNA alkylating agent temozolomide in limiting glioma growth. Finally, treatment of a bone metastatic model of human prostate cancer (PC) in immunodeficient mice was examined using a liposomal formulation of VCN (LVCN) delivered intravenously. Human PC cells were suspended in a solution of Matrigel and injected into the left tibia of immunodeficient mice. Diameters of both the left and right (control) tibias were measured by caliper repeatedly after VCN treatment was initiated.
Collapse
|
2
|
Kadomatsu M, Nakajima S, Kato H, Gu L, Chi Y, Yao J, Kitamura M. Cordycepin as a sensitizer to tumour necrosis factor (TNF)-α-induced apoptosis through eukaryotic translation initiation factor 2α (eIF2α)- and mammalian target of rapamycin complex 1 (mTORC1)-mediated inhibition of nuclear factor (NF)-κB. Clin Exp Immunol 2012; 168:325-32. [PMID: 22519596 DOI: 10.1111/j.1365-2249.2012.04580.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cordycepin (3'-deoxyadenosine) is one of the major bioactive substances produced by Cordyceps militaris, a traditional medicinal mushroom. Cordycepin possesses several biological activities, including both pro-apoptotic and anti-apoptotic properties. In the present report, we investigated an effect of cordycepin on the survival of cells exposed to tumour necrosis factor (TNF)-α. We found that subtoxic doses of cordycepin increased susceptibility of cells to TNF-α-induced apoptosis. It was associated with suppression of nuclear factor-κB (NF-κB), a major prosurvival component involved in TNF-α signalling. The adenosine transporter and A₃ adenosine receptor, but not A₁ and A₂ adenosine receptors, mediated both anti-NF-κB and pro-apoptotic effects. We found that cordycepin had the potential to phosphorylate eukaryotic translation initiation factor 2α (eIF2α) and that activation of eIF2α mimicked the suppressive effect of cordycepin on the NF-κB pathway. Furthermore, activation of eIF2α sensitized cells to TNF-α-induced apoptosis. To identify molecular events downstream of eIF2α, the role of mammalian target of rapamycin complex 1 (mTORC1) was examined. Selective activation of ₃eIF2α, as well as treatment with cordycepin, caused phosphorylation of mTORC1. Rapamycin, an inhibitor of mTORC1, significantly reversed the suppressive effects of eIF2α on NF-κB. These results suggest that cordycepin sensitizes cells to TNF-α-induced apoptosis, at least in part, via induction of the eIF2α-mTORC1 pathway and consequent suppression of NF-κB.
Collapse
Affiliation(s)
- M Kadomatsu
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | | | | | | | | | | | | |
Collapse
|
3
|
Liggett A, Crawford L, Walker B, Morris T, Irvine A. Methods for measuring proteasome activity: Current limitations and future developments. Leuk Res 2010; 34:1403-9. [DOI: 10.1016/j.leukres.2010.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 06/10/2010] [Accepted: 07/03/2010] [Indexed: 10/19/2022]
|
4
|
Zhou P, Gross S, Liu JH, Yu BY, Feng LL, Nolta J, Sharma V, Piwnica-Worms D, Qiu SX. Flavokawain B, the hepatotoxic constituent from kava root, induces GSH‐sensitive oxidative stress through modulation of IKK/NF‐κB and MAPK signaling pathways. FASEB J 2010. [DOI: 10.1096/fj.10.163311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Ping Zhou
- Division of OncologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - Shimon Gross
- Division of OncologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Department of Internal Medicine, Stem Cell ProgramUniversity of California Davis Sacramento CA USA
| | - Ji-Hua Liu
- Department of Developmental BiologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Molecular Imaging CenterMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - Bo-Yang Yu
- School of Traditional Chinese PharmacyChina Pharmaceutical University Nanjing China
| | - Ling-Ling Feng
- Key Laboratory of Plant Resources Conservation and Sustainable UtilizationSouth China Botanical GardenChinese Academy of Sciences Guangzhou China
| | - Jan Nolta
- Division of OncologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Department of Internal Medicine, Stem Cell ProgramUniversity of California Davis Sacramento CA USA
| | - Vijay Sharma
- Molecular Imaging CenterMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - David Piwnica-Worms
- Department of Developmental BiologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Molecular Imaging CenterMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - Samuel X. Qiu
- Key Laboratory of Plant Resources Conservation and Sustainable UtilizationSouth China Botanical GardenChinese Academy of Sciences Guangzhou China
| |
Collapse
|
5
|
Zhou P, Gross S, Liu JH, Yu BY, Feng LL, Nolta J, Sharma V, Piwnica-Worms D, Qiu SX. Flavokawain B, the hepatotoxic constituent from kava root, induces GSH-sensitive oxidative stress through modulation of IKK/NF-kappaB and MAPK signaling pathways. FASEB J 2010; 24:4722-32. [PMID: 20696856 DOI: 10.1096/fj.10-163311] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Kava (Piper methysticum Foster, Piperaceae) organic solvent-extract has been used to treat mild to moderate anxiety, insomnia, and muscle fatigue in Western countries, leading to its emergence as one of the 10 best-selling herbal preparations. However, several reports of severe hepatotoxicity in kava consumers led the U.S. Food and Drug Administration and authorities in Europe to restrict sales of kava-containing products. Herein we demonstrate that flavokawain B (FKB), a chalcone from kava root, is a potent hepatocellular toxin, inducing cell death in HepG2 (LD(50)=15.3 ± 0.2 μM) and L-02 (LD(50)=32 μM) cells. Hepatocellular toxicity of FKB is mediated by induction of oxidative stress, depletion of reduced glutathione (GSH), inhibition of IKK activity leading to NF-κB transcriptional blockade, and constitutive TNF-α-independent activation of mitogen-activated protein kinase (MAPK) signaling pathways, namely, ERK, p38, and JNK. We further demonstrate by noninvasive bioluminescence imaging that oral consumption of FKB leads to inhibition of hepatic NF-κB transcriptional activity in vivo and severe liver damage. Surprisingly, replenishment with exogenous GSH normalizes both TNF-α-dependent NF-κB as well as MAPK signaling and rescues hepatocytes from FKB-induced death. Our data identify FKB as a potent GSH-sensitive hepatotoxin, levels of which should be specifically monitored and controlled in kava-containing herb products.
Collapse
Affiliation(s)
- Ping Zhou
- Division of Oncology, Washington University, School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Vikis HG, Jackson EN, Krupnick AS, Franklin A, Gelman AE, Chen Q, Piwnica-Worms D, You M. Strain-specific susceptibility for pulmonary metastasis of sarcoma 180 cells in inbred mice. Cancer Res 2010; 70:4859-67. [PMID: 20501835 DOI: 10.1158/0008-5472.can-09-4177] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most cancer deaths are a result of metastasis. To extend our understanding of the factors that influence the process, we aimed to develop a mouse model of pulmonary metastasis that can be assayed in multiple inbred mouse strains for further use in identification of host genetic variants that influence metastasis. We used i.v. injection of Sarcoma 180 (S180) cells, which can be tracked and quantified by bioluminescence imaging. We observed growth of S180 cells solely in the lung and observed a wide range of pulmonary metastasis among inbred mouse strains. Interestingly, we noted that the BTBRT+tf/J strain exhibited complete clearance and provide evidence that the mechanism of resistance may involve immune factors, as strains subjected to whole-body irradiation are significantly more susceptible to tumor growth. One possible mechanism of resistance to pulmonary metastasis in BTBRT+tf/J mice may require T-cell function. Our experiments present a new mouse model for further characterization of the genetics and mechanisms of pulmonary metastasis.
Collapse
Affiliation(s)
- Haris G Vikis
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
LRP6 overexpression defines a class of breast cancer subtype and is a target for therapy. Proc Natl Acad Sci U S A 2010; 107:5136-41. [PMID: 20194742 DOI: 10.1073/pnas.0911220107] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The Wnt/beta-catenin signaling pathway is activated in breast cancer, a leading cause of cancer mortality in women. Because mutations in the key intracellular components of this pathway are rare, identifying the molecular mechanisms of aberrant Wnt activation in breast cancer is critical for development of pathway-targeted therapy. Here, we show that expression of the Wnt signaling coreceptor LRP6 is up-regulated in a subpopulation of human breast cancers. LRP6 silencing in breast cancer cells reduces Wnt signaling, cell proliferation, and in vivo tumor growth. In vivo administration of an LRP6 antagonist, Mesd, markedly suppressed growth of MMTV-Wnt1 tumors without causing undesirable side effects. These results demonstrate that Wnt activation at the cell surface contributes to breast cancer tumorigenesis. Together, our studies highlight LRP6 as a potential therapeutic target in breast cancer, and introduce Mesd as a promising antitumor agent for treating breast cancer subtypes with Wnt activation at the cell surface.
Collapse
|
8
|
Jost SC, Collins L, Travers S, Piwnica-Worms D, Garbow JR. Measuring Brain Tumor Growth: Combined Bioluminescence Imaging–Magnetic Resonance Imaging Strategy. Mol Imaging 2009. [DOI: 10.2310/7290.2009.00023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Small-animal tumor models are essential for developing translational therapeutic strategies in oncology research, with imaging having an increasingly important role. Magnetic resonance imaging (MRI) offers tumor localization, volumetric measurement, and the potential for advanced physiologic imaging but is less well suited to high-throughput studies and has limited capacity to assess early tumor growth. Bioluminescence imaging (BLI) identifies tumors early, monitors tumor growth, and efficiently measures response to therapeutic intervention. Generally, BLI signals have been found to correlate well with magnetic resonance measurements of tumor volume. However, in our studies of small-animal models of malignant brain tumors, we have observed specific instances in which BLI data do not correlate with corresponding MRIs. These observations led us to hypothesize that use of BLI and MRI together, rather than in isolation, would allow more effective and efficient measures of tumor growth in preclinical studies. Herein we describe combining BLI and MRI studies to characterize tumor growth in a mouse model of glioblastoma. The results led us to suggest a cost-effective, multimodality strategy for selecting cohorts of animals with similar tumor growth patterns that improves the accuracy of longitudinal in vivo measurements of tumor growth and treatment response in preclinical therapeutic studies.
Collapse
Affiliation(s)
- Sarah C. Jost
- From the Department of Neurosurgery; Molecular Imaging Center, Department of Radiology, and Department of Developmental Biology; Biomedical MR Laboratory, Department of Radiology; Alvin J Siteman Cancer Center; Washington University School of Medicine, Saint Louis, MO
| | - Lynne Collins
- From the Department of Neurosurgery; Molecular Imaging Center, Department of Radiology, and Department of Developmental Biology; Biomedical MR Laboratory, Department of Radiology; Alvin J Siteman Cancer Center; Washington University School of Medicine, Saint Louis, MO
| | - Sarah Travers
- From the Department of Neurosurgery; Molecular Imaging Center, Department of Radiology, and Department of Developmental Biology; Biomedical MR Laboratory, Department of Radiology; Alvin J Siteman Cancer Center; Washington University School of Medicine, Saint Louis, MO
| | - David Piwnica-Worms
- From the Department of Neurosurgery; Molecular Imaging Center, Department of Radiology, and Department of Developmental Biology; Biomedical MR Laboratory, Department of Radiology; Alvin J Siteman Cancer Center; Washington University School of Medicine, Saint Louis, MO
| | - Joel R. Garbow
- From the Department of Neurosurgery; Molecular Imaging Center, Department of Radiology, and Department of Developmental Biology; Biomedical MR Laboratory, Department of Radiology; Alvin J Siteman Cancer Center; Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
9
|
Carreno BM, Garbow JR, Kolar GR, Jackson EN, Engelbach JA, Becker-Hapak M, Carayannopoulos LN, Piwnica-Worms D, Linette GP. Immunodeficient mouse strains display marked variability in growth of human melanoma lung metastases. Clin Cancer Res 2009; 15:3277-86. [PMID: 19447870 DOI: 10.1158/1078-0432.ccr-08-2502] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunodeficient mice serve as critical hosts for transplantation of xenogeneic cells for in vivo analysis of various biological processes. Because investigators typically select one or two immunodeficient mouse strains as recipients, no comprehensive study has been published documenting differences in human tumor engraftment. Taking advantage of the increased metastatic potential of RhoC-expressing human (A375) melanoma cells, we evaluate four immunodeficient mouse strains: severe combined immunodeficiency (scid), nonobese diabetic (NOD)-scid, NOD-scid beta2m(null), and NOD-scid IL2Rgamma(null) as xenograft tumor recipients. EXPERIMENTAL DESIGN Bioluminescence, magnetic resonance imaging, and histopathology were used to monitor serial tumor growth. Natural killer (NK) cell function was examined in each mouse strain using standard (51)Chromium release assays. RESULTS Melanoma metastases growth is delayed and variable in scid and NOD-scid mice. In contrast, NOD-scid beta2m(null) and NOD-scid IL2Rgamma(null) mice show rapid tumor engraftment, although tumor growth is variable in NOD-scid beta2m(null) mice. NK cells were detected in all strains except NOD-scid IL2Rgamma(null), and in vitro activated scid, NOD-scid, and NOD-scid beta2m(null) NK cells kill human melanoma lines and primary melanoma cells. Expression of human NKG2D ligands MHC class I chain-related A and B molecules renders melanoma susceptible to murine NK cell-mediated cytotoxicity and killing is inhibited by antibody blockade of murine NKG2D. CONCLUSIONS Murine NKG2D recognition of MICA/B is an important receptor-ligand interaction used by NK cells in immunodeficient strains to limit engraftment of human tumors. The absolute NK deficiency in NOD-scid IL2Rgamma(null) animals makes this strain an excellent recipient of melanoma and potentially other human malignancies.
Collapse
Affiliation(s)
- Beatriz M Carreno
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Gross S, Gammon ST, Moss BL, Rauch D, Harding J, Heinecke JW, Ratner L, Piwnica-Worms D. Bioluminescence imaging of myeloperoxidase activity in vivo. Nat Med 2009; 15:455-61. [PMID: 19305414 PMCID: PMC2831476 DOI: 10.1038/nm.1886] [Citation(s) in RCA: 259] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Accepted: 10/18/2008] [Indexed: 12/17/2022]
Abstract
The myeloperoxidase (MPO) system of activated phagocytes is central to normal host defense mechanisms, and dysregulated MPO contributes to the pathogenesis of inflammatory disease states ranging from atherosclerosis to cancer. Here we show that upon systemic administration, the small molecule luminol enables noninvasive bioluminescence imaging (BLI) of MPO activity in vivo. Luminol-BLI allowed quantitative longitudinal monitoring of MPO activity in animal models of acute dermatitis, mixed allergic contact hypersensitivity, focal arthritis and spontaneous large granular lymphocytic tumors. Bioluminescence colocalized with histological sites of inflammation and was totally abolished in gene-deleted Mpo(-/-) mice, despite massive tissue infiltration of neutrophils and activated eosinophils, indicating that eosinophil peroxidase did not contribute to luminol-BLI in vivo. Thus, luminol-BLI provides a noninvasive, specific and highly sensitive optical readout of phagocyte-mediated MPO activity in vivo and may enable new diagnostic applications in a wide range of acute and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Shimon Gross
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Flentie KN, Qi M, Gammon ST, Razia Y, Lui F, Marpegan L, Manglik A, Piwnica-Worms D, McKinney JS. Stably IntegratedluxCDABEfor Assessment ofSalmonellaInvasion Kinetics. Mol Imaging 2008. [DOI: 10.2310/7290.2008.00024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Kelly N. Flentie
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Min Qi
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Seth T. Gammon
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Yasmin Razia
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Felix Lui
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Luciano Marpegan
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Aashish Manglik
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - David Piwnica-Worms
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | - Jeffrey S. McKinney
- From the Departments of Molecular Microbiology, Pediatrics, Biology, and Developmental Biology and Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
12
|
Coppola JM, Ross BD, Rehemtulla A. Noninvasive imaging of apoptosis and its application in cancer therapeutics. Clin Cancer Res 2008; 14:2492-501. [PMID: 18413842 DOI: 10.1158/1078-0432.ccr-07-0782] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Activation of the apoptotic cascade plays an important role in the response of tumors to therapy. Noninvasive imaging of apoptosis facilitates optimization of therapeutic protocols regarding dosing and schedule and enables identification of efficacious combination therapies. EXPERIMENTAL DESIGN We describe a hybrid polypeptide that reports on caspase-3 activity in living cells and animals in a noninvasive manner. This reporter, ANLucBCLuc, constitutes a fusion of small interacting peptides, peptide A and peptide B, with the NLuc and CLuc fragments of luciferase with a caspase-3 cleavage site (DEVD) between pepANLuc (ANLuc) and pepBCLuc (BCLuc). During apoptosis, caspase-3 cleaves the reporter, enabling separation of ANLuc from BCLuc. A high-affinity interaction between peptide A and peptide B restores luciferase activity by NLuc and CLuc complementation. Using a D54 glioma model, we show the utility of the reporter in imaging of apoptosis in living subjects in response to various chemotherapy and radiotherapy regimens. RESULTS Treatment of live cells and mice carrying D54 tumor xenografts with chemotherapeutic agents such as temozolomide and perifosine resulted in induction of bioluminescence activity, which correlated with activation of caspase-3. Treatment of mice with combination therapy of temozolomide and radiation resulted in increased bioluminescence activity over individual treatments and increased therapeutic response due to enhanced apoptosis. CONCLUSION The data provided show the utility of the ANLucBCLuc reporter in dynamic, noninvasive imaging of apoptosis and provides a rationale for use of this technology to optimize dose and schedule of novel therapies or to develop novel combination therapies using existing drugs.
Collapse
Affiliation(s)
- Julia M Coppola
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
13
|
Robers MB, Horton RA, Bercher MR, Vogel KW, Machleidt T. High-throughput cellular assays for regulated posttranslational modifications. Anal Biochem 2007; 372:189-97. [PMID: 17961489 DOI: 10.1016/j.ab.2007.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Revised: 08/21/2007] [Accepted: 09/07/2007] [Indexed: 10/22/2022]
Abstract
We have developed a set of high-throughput screening (HTS)-compatible assays capable of measuring regulated, target-specific posttranslational modifications in a mammalian cell-based format. We chose the NFkappaB signal transduction cascade as a model system to validate this approach because specific target proteins in this signaling pathway undergo a multitude of posttranslational modifications in response to pathway stimulation. In this pathway, TNFalpha induces the phosphorylation, ubiquitination, and proteasomal degradation of IkappaBalpha, which leads to the release and translocation of the NFkappaB transcriptional complex into the nucleus. To measure these cellular processes, we describe the use of a stable cell line expressing a fusion of green fluorescent protein (GFP) with IkappaBalpha that can be interrogated for either ubiquitination or phosphorylation using a unique set of terbium-labeled antibodies in a time-resolved Förster resonance energy transfer (TR-FRET)-based readout. Concurrently, we have engineered a beta-lactamase-IkappaBalpha reporter cell line that can be used to quantify proteasomal degradation of IkappaBalpha in living cells. Both TR-FRET and beta-lactamase reporter technologies provide a convenient, sensitive, and robust means to interrogate the chronological steps in NFkappaB signaling in a physiologically relevant cellular context without the need to overexpress any enzyme involved in this pathway. Cellular HTS assays that interrogate such processes will provide a unique integrated approach to dissecting intermediate steps in NFkappaB activation and could serve as examples of broadly applicable pathway analysis tools for target-based drug discovery.
Collapse
|
14
|
Gross S, Abraham U, Prior JL, Herzog ED, Piwnica-Worms D. Continuous Delivery of D-Luciferin by Implanted Micro-osmotic Pumps Enables True Real-Time Bioluminescence Imaging of Luciferase Activity in Vivo. Mol Imaging 2007. [DOI: 10.2310/7290.2007.00009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Shimon Gross
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology, Department of Molecular Biology and Pharmacology, and Department of Biology, Washington University in St. Louis, St. Louis, MO
| | - Ute Abraham
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology, Department of Molecular Biology and Pharmacology, and Department of Biology, Washington University in St. Louis, St. Louis, MO
| | - Julie L. Prior
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology, Department of Molecular Biology and Pharmacology, and Department of Biology, Washington University in St. Louis, St. Louis, MO
| | - Erik D. Herzog
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology, Department of Molecular Biology and Pharmacology, and Department of Biology, Washington University in St. Louis, St. Louis, MO
| | - David Piwnica-Worms
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology, Department of Molecular Biology and Pharmacology, and Department of Biology, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
15
|
Hirbe AC, Uluçkan O, Morgan EA, Eagleton MC, Prior JL, Piwnica-Worms D, Trinkaus K, Apicelli A, Weilbaecher K. Granulocyte colony-stimulating factor enhances bone tumor growth in mice in an osteoclast-dependent manner. Blood 2006; 109:3424-31. [PMID: 17192391 PMCID: PMC1852257 DOI: 10.1182/blood-2006-09-048686] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Inhibition of osteoclast (OC) activity has been associated with decreased tumor growth in bone in animal models. Increased recognition of factors that promote osteoclastic bone resorption in cancer patients led us to investigate whether increased OC activation could enhance tumor growth in bone. Granulocyte colony-stimulating factor (G-CSF) is used to treat chemotherapy-induced neutropenia, but is also associated with increased markers of OC activity and decreased bone mineral density (BMD). We used G-CSF as a tool to investigate the impact of increased OC activity on tumor growth in 2 murine osteolytic tumor models. An 8-day course of G-CSF alone (without chemotherapy) significantly decreased BMD and increased OC perimeter along bone in mice. Mice administered G-CSF alone demonstrated significantly increased tumor growth in bone as quantitated by in vivo bioluminescence imaging and histologic bone marrow tumor analysis. Short-term administration of AMD3100, a CXCR4 inhibitor that mobilizes neutrophils with little effect on bone resorption, did not lead to increased tumor burden. However, OC-defective osteoprotegerin transgenic (OPG(Tg)) mice and bisphosphonate-treated mice were resistant to the effects of G-CSF administration upon bone tumor growth. These data demonstrate a G-CSF-induced stimulation of tumor growth in bone that is OC dependent.
Collapse
Affiliation(s)
- Angela C Hirbe
- Department of Medicine and Division of Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ilagan R, Pottratz J, Le K, Zhang L, Wong SG, Ayala R, Iyer M, Wu L, Gambhir SS, Carey M. Imaging Mitogen-Activated Protein Kinase Function in Xenograft Models of Prostate Cancer. Cancer Res 2006; 66:10778-85. [PMID: 17108114 DOI: 10.1158/0008-5472.can-05-3577] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mitogen-activated protein kinases (MAPK) play important roles in malignancy. The ability to detect and quantitate MAPKs in live animal models of cancer will facilitate an understanding of disease progression. We have developed a gene expression-based imaging system that detects and quantifies MAPK activity in prostate cancer tumors implanted into severe combined immunodeficient mice. The imaging technology uses a modified version of two-step transcriptional amplification (TSTA). The tissue specificity of gene expression is imparted by an enhanced version of the prostate-specific antigen regulatory region that expresses GAL4-ELK1. GAL4-ELK1 confers MAPK specificity by activating a firefly luciferase (FLuc) reporter gene when the Ets-like transcription factor (ELK) 1 activation domain is phosphorylated by MAPK. FLuc activity in live animals was detected using the Xenogen In vivo Imaging System. We validated the TSTA-ELK1 system by analyzing its response to epidermal growth factor treatment in transfected tissue culture cells and in adenovirus (AdTSTA-ELK1)-injected prostate cancer xenograft tumors. We measured MAPK activity in two well-characterized xenograft models, CWR22 and LAPC9. Although no significant differences in MAPK levels were detected between androgen-dependent and androgen-independent xenografts, the CWR22 models display significantly higher levels of AdTSTA-ELK1 activity versus LAPC9. Western blots of tumor extracts showed that the elevated imaging signal in CWR22 xenografts correlated with elevated levels of phosphorylated extracellular signal-regulated kinase 1/2 but not p38 or c-Jun NH(2)-terminal kinase. We conclude that a gene expression-based optical imaging system can accurately detect and quantify MAPK activity in live animals.
Collapse
Affiliation(s)
- Romyla Ilagan
- Department of Biological Chemistry, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yuan L, Siegel M, Choi K, Khosla C, Miller CR, Jackson EN, Piwnica-Worms D, Rich KM. Transglutaminase 2 inhibitor, KCC009, disrupts fibronectin assembly in the extracellular matrix and sensitizes orthotopic glioblastomas to chemotherapy. Oncogene 2006; 26:2563-73. [PMID: 17099729 DOI: 10.1038/sj.onc.1210048] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transglutaminase 2 (TG2, a.k.a. tissue transglutaminase) belongs to a family of transglutaminase enzymes that stabilize proteins by affecting covalent crosslinking via formation of amide bonds. Cell surface TG2 is directly involved as an adhesive receptor in cell-extracellular matrix (ECM) interactions. Here, we show that TG2 activity is elevated in glioblastomas compared with non-neoplastic brain. Immunofluorescent studies showed increased staining of fibronectin colocalized with TG2 in the ECM in glioblastomas. In addition, small clusters of invading human glioblastoma cells present in non-neoplastic brain parenchyma secrete high levels of TG2 and fibronectin that distinguish them from normal brain stroma. Downregulation of TG2 in U87MG glioblastoma cells with RNAi demonstrated decreased assembly of fibronectin in the ECM. Treatment with KCC009 blocked the remodeling of fibronectin in the ECM in glioblastomas in both in vitro and in vivo studies. KCC009 treatment in mice harboring orthotopic glioblastomas (DBT-FG) sensitized the tumors to N,N'-bis(2-chloroethyl)-N-nitrosourea chemotherapy, as measured by reduced bioluminescence, increased apoptosis and prolonged survival. The ability of KCC009 to interfere with the permissive remodeling of fibronectin in the ECM in glioblastomas suggests a novel target to enhance sensitivity to chemotherapy directed not only at the tumor mass, but also invading glioblastoma cells.
Collapse
Affiliation(s)
- L Yuan
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kesarwala AH, Prior JL, Sun J, Harpstrite SE, Sharma V, Piwnica-Worms D. Second-Generation Triple Reporter for Bioluminescence, Micro–Positron Emission Tomography, and Fluorescence Imaging. Mol Imaging 2006. [DOI: 10.2310/7290.2006.00024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Aparna H. Kesarwala
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO
| | - Julie L. Prior
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO
| | - Jinwu Sun
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO
| | - Scott E. Harpstrite
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO
| | - Vijay Sharma
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO
| | - David Piwnica-Worms
- From the Molecular Imaging Center, Mallinckrodt Institute of Radiology and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
19
|
Hajitou A, Trepel M, Lilley CE, Soghomonyan S, Alauddin MM, Marini FC, Restel BH, Ozawa MG, Moya CA, Rangel R, Sun Y, Zaoui K, Schmidt M, von Kalle C, Weitzman MD, Gelovani JG, Pasqualini R, Arap W. A hybrid vector for ligand-directed tumor targeting and molecular imaging. Cell 2006; 125:385-98. [PMID: 16630824 DOI: 10.1016/j.cell.2006.02.042] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2005] [Revised: 01/11/2006] [Accepted: 02/01/2006] [Indexed: 11/21/2022]
Abstract
Merging tumor targeting and molecular-genetic imaging into an integrated platform is limited by lack of strategies to enable systemic yet ligand-directed delivery and imaging of specific transgenes. Many eukaryotic viruses serve for transgene delivery but require elimination of native tropism for mammalian cells; in contrast, prokaryotic viruses can be adapted to bind to mammalian receptors but are otherwise poor vehicles. Here we introduce a system containing cis-elements from adeno-associated virus (AAV) and single-stranded bacteriophage. Our AAV/phage (AAVP) prototype targets an integrin. We show that AAVP provides superior tumor transduction over phage and that incorporation of inverted terminal repeats is associated with improved fate of the delivered transgene. Moreover, we show that the temporal dynamics and spatial heterogeneity of gene expression mediated by targeted AAVP can be monitored by positron emission tomography. This new class of targeted hybrid viral particles will enable a wide range of applications in biology and medicine.
Collapse
Affiliation(s)
- Amin Hajitou
- Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Genetically encoded imaging reporters introduced into cells and transgenic animals enable noninvasive, longitudinal studies of dynamic biological processes in vivo. The most common reporters include firefly luciferase (bioluminescence imaging), green fluorescence protein (fluorescence imaging), herpes simplex virus-1 thymidine kinase (positron emission tomography), and variants with enhanced spectral and kinetic properties. When cloned into promoter/enhancer sequences or engineered into fusion proteins, imaging reporters allow transcriptional regulation, signal transduction, protein-protein interactions, oncogenic transformation, cell trafficking, and targeted drug action to be spatiotemporally resolved in vivo. Spying on cancer with genetically encoded imaging reporters provides insight into cancer-specific molecular machinery within the context of the whole animal.
Collapse
Affiliation(s)
- Shimon Gross
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110, USA
| | | |
Collapse
|