Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(18): eadf0138, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37134162

ABSTRACT

Proliferating cells rely on acetyl-CoA to support membrane biogenesis and acetylation. Several organelle-specific pathways are available for provision of acetyl-CoA as nutrient availability fluctuates, so understanding how cells maintain acetyl-CoA homeostasis under such stresses is critically important. To this end, we applied 13C isotope tracing cell lines deficient in these mitochondrial [ATP-citrate lyase (ACLY)]-, cytosolic [acetyl-CoA synthetase (ACSS2)]-, and peroxisomal [peroxisomal biogenesis factor 5 (PEX5)]-dependent pathways. ACLY knockout in multiple cell lines reduced fatty acid synthesis and increased reliance on extracellular lipids or acetate. Knockout of both ACLY and ACSS2 (DKO) severely stunted but did not entirely block proliferation, suggesting that alternate pathways can support acetyl-CoA homeostasis. Metabolic tracing and PEX5 knockout studies link peroxisomal oxidation of exogenous lipids as a major source of acetyl-CoA for lipogenesis and histone acetylation in cells lacking ACLY, highlighting a role for inter-organelle cross-talk in supporting cell survival in response to nutrient fluctuations.


Subject(s)
Acetates , Lipogenesis , Acetyl Coenzyme A/metabolism , Acetates/metabolism , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Mitochondria/metabolism , Homeostasis , Stress, Physiological
2.
Sci Adv ; 5(7): eaaw6455, 2019 07.
Article in English | MEDLINE | ID: mdl-31355336

ABSTRACT

The LKB1 tumor suppressor is often mutationally inactivated in non-small cell lung cancer (NSCLC). LKB1 phosphorylates and activates members of the AMPK family of Ser/Thr kinases. Within this family, the salt-inducible kinases (SIKs) modulate gene expression in part via the inhibitory phosphorylation of the CRTCs, coactivators for CREB (cAMP response element-binding protein). The loss of LKB1 causes SIK inactivation and the induction of the CRTCs, leading to the up-regulation of CREB target genes. We identified CRTC2 as a critical factor in LKB1-deficient NSCLC. CRTC2 is unphosphorylated and therefore constitutively activated in LKB1-mutant NSCLC, where it promotes tumor growth, in part via the induction of the inhibitor of DNA binding 1 (ID1), a bona fide CREB target gene. As ID1 expression is up-regulated and confers poor prognosis in LKB1-deficient NSCLC, our results suggest that small molecules that inhibit CRTC2 and ID1 activity may provide therapeutic benefit to individuals with NSCLC.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Lung Neoplasms/genetics , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/metabolism , AMP-Activated Protein Kinase Kinases , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Protein 1/metabolism , Lung Neoplasms/pathology , Mice, SCID , Prognosis , Signal Transduction
3.
Cancer Discov ; 9(11): 1606-1627, 2019 11.
Article in English | MEDLINE | ID: mdl-31350328

ABSTRACT

Mutations in the LKB1 (also known as STK11) tumor suppressor are the third most frequent genetic alteration in non-small cell lung cancer (NSCLC). LKB1 encodes a serine/threonine kinase that directly phosphorylates and activates 14 AMPK family kinases ("AMPKRs"). The function of many of the AMPKRs remains obscure, and which are most critical to the tumor-suppressive function of LKB1 remains unknown. Here, we combine CRISPR and genetic analysis of the AMPKR family in NSCLC cell lines and mouse models, revealing a surprising critical role for the SIK subfamily. Conditional genetic loss of Sik1 revealed increased tumor growth in mouse models of Kras-dependent lung cancer, which was further enhanced by loss of the related kinase Sik3. As most known substrates of the SIKs control transcription, gene-expression analysis was performed, revealing upregulation of AP1 and IL6 signaling in common between LKB1- and SIK1/3-deficient tumors. The SIK substrate CRTC2 was required for this effect, as well as for proliferation benefits from SIK loss. SIGNIFICANCE: The tumor suppressor LKB1/STK11 encodes a serine/threonine kinase frequently inactivated in NSCLC. LKB1 activates 14 downstream kinases in the AMPK family controlling growth and metabolism, although which kinases are critical for LKB1 tumor-suppressor function has remained an enigma. Here we unexpectedly found that two understudied kinases, SIK1 and SIK3, are critical targets in lung cancer.This article is highlighted in the In This Issue feature, p. 1469.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , A549 Cells , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Animals , CRISPR-Cas Systems , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Gene Editing , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Neoplasm Transplantation , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Tumor Burden
4.
Cell Metab ; 29(2): 285-302.e7, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30415923

ABSTRACT

AMPK, a conserved sensor of low cellular energy, can either repress or promote tumor growth depending on the context. However, no studies have examined AMPK function in autochthonous genetic mouse models of epithelial cancer. Here, we examine the role of AMPK in murine KrasG12D-mediated non-small-cell lung cancer (NSCLC), a cancer type in humans that harbors frequent inactivating mutations in the LKB1 tumor suppressor-the predominant upstream activating kinase of AMPK and 12 related kinases. Unlike LKB1 deletion, AMPK deletion in KrasG12D lung tumors did not accelerate lung tumor growth. Moreover, deletion of AMPK in KrasG12D p53f/f tumors reduced lung tumor burden. We identified a critical role for AMPK in regulating lysosomal gene expression through the Tfe3 transcription factor, which was required to support NSCLC growth. Thus, AMPK supports the growth of KrasG12D-dependent lung cancer through the induction of lysosomes, highlighting an unrecognized liability of NSCLC.


Subject(s)
AMP-Activated Protein Kinases/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line, Tumor , Loss of Function Mutation , Mice
5.
Cell ; 171(3): 696-709.e23, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28965760

ABSTRACT

The transcription factor NRF2 is a master regulator of the cellular antioxidant response, and it is often genetically activated in non-small-cell lung cancers (NSCLCs) by, for instance, mutations in the negative regulator KEAP1. While direct pharmacological inhibition of NRF2 has proven challenging, its aberrant activation rewires biochemical networks in cancer cells that may create special vulnerabilities. Here, we use chemical proteomics to map druggable proteins that are selectively expressed in KEAP1-mutant NSCLC cells. Principal among these is NR0B1, an atypical orphan nuclear receptor that we show engages in a multimeric protein complex to regulate the transcriptional output of KEAP1-mutant NSCLC cells. We further identify small molecules that covalently target a conserved cysteine within the NR0B1 protein interaction domain, and we demonstrate that these compounds disrupt NR0B1 complexes and impair the anchorage-independent growth of KEAP1-mutant cancer cells. Our findings designate NR0B1 as a druggable transcriptional regulator that supports NRF2-dependent lung cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Proteome/analysis , Transcriptome , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cysteine/metabolism , DAX-1 Orphan Nuclear Receptor/metabolism , Gene Regulatory Networks , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Ligands , Lung Neoplasms/metabolism
6.
Metab Eng ; 43(Pt B): 208-217, 2017 09.
Article in English | MEDLINE | ID: mdl-28034771

ABSTRACT

The Liver Kinase B1 (LKB1) tumor suppressor acts as a metabolic energy sensor to regulate AMP-activated protein kinase (AMPK) signaling and is commonly mutated in various cancers, including non-small cell lung cancer (NSCLC). Tumor cells deficient in LKB1 may be uniquely sensitized to metabolic stresses, which may offer a therapeutic window in oncology. To address this question we have explored how functional LKB1 impacts the metabolism of NSCLC cells using 13C metabolic flux analysis. Isogenic NSCLC cells expressing functional LKB1 exhibited higher flux through oxidative mitochondrial pathways compared to those deficient in LKB1. Re-expression of LKB1 also increased the capacity of cells to oxidize major mitochondrial substrates, including pyruvate, fatty acids, and glutamine. Furthermore, LKB1 expression promoted an adaptive response to energy stress induced by anchorage-independent growth. Finally, this diminished adaptability sensitized LKB1-deficient cells to combinatorial inhibition of mitochondrial complex I and glutaminase. Together, our data implicate LKB1 as a major regulator of adaptive metabolic reprogramming and suggest synergistic pharmacological strategies for mitigating LKB1-deficient NSCLC tumor growth.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Energy Metabolism , Lung Neoplasms/metabolism , Mitochondria/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological , A549 Cells , AMP-Activated Protein Kinase Kinases , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mitochondria/genetics , Mitochondria/pathology , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/genetics
7.
Nat Med ; 22(10): 1108-1119, 2016 10.
Article in English | MEDLINE | ID: mdl-27643638

ABSTRACT

Continuous de novo fatty acid synthesis is a common feature of cancer that is required to meet the biosynthetic demands of a growing tumor. This process is controlled by the rate-limiting enzyme acetyl-CoA carboxylase (ACC), an attractive but traditionally intractable drug target. Here we provide genetic and pharmacological evidence that in preclinical models ACC is required to maintain the de novo fatty acid synthesis needed for growth and viability of non-small-cell lung cancer (NSCLC) cells. We describe the ability of ND-646-an allosteric inhibitor of the ACC enzymes ACC1 and ACC2 that prevents ACC subunit dimerization-to suppress fatty acid synthesis in vitro and in vivo. Chronic ND-646 treatment of xenograft and genetically engineered mouse models of NSCLC inhibited tumor growth. When administered as a single agent or in combination with the standard-of-care drug carboplatin, ND-646 markedly suppressed lung tumor growth in the Kras;Trp53-/- (also known as KRAS p53) and Kras;Stk11-/- (also known as KRAS Lkb1) mouse models of NSCLC. These findings demonstrate that ACC mediates a metabolic liability of NSCLC and that ACC inhibition by ND-646 is detrimental to NSCLC growth, supporting further examination of the use of ACC inhibitors in oncology.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acids/biosynthesis , Lipid Metabolism/drug effects , Lung Neoplasms/metabolism , Pyrimidinones/pharmacology , Thiophenes/pharmacology , AMP-Activated Protein Kinases , Acetyltransferases/antagonists & inhibitors , Allosteric Regulation , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation/genetics , Humans , Lipid Metabolism/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Mice, Knockout , Molecular Targeted Therapy , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
8.
Article in English | MEDLINE | ID: mdl-28062532

ABSTRACT

The renaissance in the study of cancer metabolism has refocused efforts to identify and target metabolic dependencies of tumors as an approach for cancer therapy. One of the unique metabolic requirements that cancer cells possess to sustain their biosynthetic growth demands is altered fatty acid metabolism, in particular the synthesis of de novo fatty acids that are required as cellular building blocks to support cell division. Enhanced fatty acid synthesis that is observed in many tumor types has been postulated to open a therapeutic window for cancer therapy and, correspondingly, efforts to pharmacologically inhibit key enzymes of fatty acid synthesis are being pursued. However, despite these efforts, whether inhibition of fatty acid synthesis stunts tumor growth in vivo has been poorly understood. In this review, we focus on the recent evidence that pharmacologic inhibition of acetyl-CoA carboxylase, the enzyme that regulates the rate-limiting step of de novo fatty acid synthesis, exposes a metabolic liability of non-small cell lung cancer and represses tumor growth in preclinical models.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Fatty Acids/metabolism , Gene Regulatory Networks/physiology , Pyrimidinones/metabolism , Thiophenes/metabolism , Acetyl-CoA Carboxylase/genetics , Animals , Fatty Acids/genetics , Humans , Models, Animal
9.
Mol Cell ; 55(3): 436-50, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25042806

ABSTRACT

The serine/threonine kinase LKB1 is a tumor suppressor whose loss is associated with increased metastatic potential. In an effort to define biochemical signatures of metastasis associated with LKB1 loss, we discovered that the epithelial-to-mesenchymal transition transcription factor Snail1 was uniquely upregulated upon LKB1 deficiency across cell types. The ability of LKB1 to suppress Snail1 levels was independent of AMPK but required the related kinases MARK1 and MARK4. In a screen for substrates of these kinases involved in Snail regulation, we identified the scaffolding protein DIXDC1. Similar to loss of LKB1, DIXDC1 depletion results in upregulation of Snail1 in a FAK-dependent manner, leading to increased cell invasion. MARK1 phosphorylation of DIXDC1 is required for its localization to focal adhesions and ability to suppress metastasis in mice. DIXDC1 is frequently downregulated in human cancers, which correlates with poor survival. This study defines an AMPK-independent phosphorylation cascade essential for LKB1-dependent control of metastatic behavior.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Neoplasm Invasiveness/genetics , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms , Mice , Microfilament Proteins/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Proc Natl Acad Sci U S A ; 111(7): 2554-9, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550282

ABSTRACT

One of the major metabolic changes associated with cellular transformation is enhanced nutrient utilization, which supports tumor progression by fueling both energy production and providing biosynthetic intermediates for growth. The liver kinase B1 (LKB1) is a serine/threonine kinase and tumor suppressor that couples bioenergetics to cell-growth control through regulation of mammalian target of rapamycin (mTOR) activity; however, the influence of LKB1 on tumor metabolism is not well defined. Here, we show that loss of LKB1 induces a progrowth metabolic program in proliferating cells. Cells lacking LKB1 display increased glucose and glutamine uptake and utilization, which support both cellular ATP levels and increased macromolecular biosynthesis. This LKB1-dependent reprogramming of cell metabolism is dependent on the hypoxia-inducible factor-1α (HIF-1α), which accumulates under normoxia in LKB1-deficient cells and is antagonized by inhibition of mTOR complex I signaling. Silencing HIF-1α reverses the metabolic advantages conferred by reduced LKB1 signaling and impairs the growth and survival of LKB1-deficient tumor cells under low-nutrient conditions. Together, our data implicate the tumor suppressor LKB1 as a central regulator of tumor metabolism and growth control through the regulation of HIF-1α-dependent metabolic reprogramming.


Subject(s)
Energy Metabolism/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Metabolic Networks and Pathways/genetics , Protein Serine-Threonine Kinases/deficiency , AMP-Activated Protein Kinase Kinases , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Fibroblasts , Gas Chromatography-Mass Spectrometry , Glucose/metabolism , Glutamine/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Metabolic Networks and Pathways/physiology , Mice , Multiprotein Complexes/metabolism , Oxygen Consumption/physiology , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
J Environ Pathol Toxicol Oncol ; 32(1): 29-39, 2013.
Article in English | MEDLINE | ID: mdl-23758150

ABSTRACT

Environmental factors are likely to interact with genetic determinants to influence prostate cancer progression. The Agricultural Health Study has identified an association between exposure to organophosphorous pesticides including chlorpyrifos, and increased prostate cancer risk in pesticide applicators with a first-degree family history of this disease. Exploration of this potential gene-environment interaction would benefit from the development of a suitable animal model. Utilizing a previously described mouse model that is genetically predisposed to prostate cancer through a prostate-specific heterozygous PTEN deletion, termed C57/Luc/Ptenp+/-, we used bioluminescence imaging and histopathological analyses to test whether chronic exposure to chlorpyrifos in a grain-based diet for 32 weeks was able to promote prostate cancer development. Chronic exposure to chlorpyrifos in the diet did not promote prostate cancer development in C57/Luc/Ptenp+/- mice despite achieving sufficient levels to inhibit acetylcholinesterase activity in plasma. We found no significant differences in numbers of murine prostatic intraepithelial neoplasia lesions or disease progression in chlorpyrifos versus control treated animals up to 32 weeks. The mechanistic basis of pesticide-induced prostate cancer may be complex and may involve other genetic variants, multiple genes, or nongenetic factors that might alter prostate cancer risk during pesticide exposure in agricultural workers.


Subject(s)
Chlorpyrifos/adverse effects , Gene-Environment Interaction , Insecticides/adverse effects , Mutation/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/genetics , Acetylcholinesterase/blood , Administration, Oral , Animals , Chlorpyrifos/administration & dosage , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Gene Deletion , Insecticides/administration & dosage , Luminescent Measurements , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Prostatic Neoplasms/epidemiology , Risk Factors
12.
Clin Exp Metastasis ; 30(4): 541-52, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23224938

ABSTRACT

Integrin α3ß1 promotes tumor cell adhesion, migration, and invasion on laminin isoforms, and several clinical studies have indicated a correlation between increased tumoral α3ß1 integrin expression and tumor progression, metastasis, and poor patient outcomes. However, several other clinical and experimental studies have suggested that α3ß1 can possess anti-metastatic activity in certain settings. To help define the range of α3ß1 functions in tumor cells in vivo, we used RNAi to silence the α3 integrin subunit in an aggressive, in vivo-passaged subline of PC-3 prostate carcinoma cells. Loss of α3 integrin impaired adhesion and proliferation on the α3ß1 integrin ligand, laminin-332 in vitro. Despite these deficits in vitro, the α3-silenced cells were significantly more aggressive in a lung colonization model in vivo, with a substantially increased rate of tumor growth that significantly reduced survival. In contrast, silencing the related α6 integrin subunit delayed metastatic growth in vivo. The increased colonization of α3-silenced tumor cells in vivo was recapitulated in 3D collagen co-cultures with lung fibroblasts or pre-osteoblast-like cells, where α3-silenced cells showed dramatically enhanced growth. The increased response of α3-silenced tumor cells to stromal cells in co-culture could be reproduced by fibroblast conditioned medium, which contains one or more heparin-binding factors that selectively favor the growth of α3-silenced cells. Our new data suggest a scenario in which α3ß1 regulates tumor-host interactions within the metastatic tumor microenvironment to limit growth, providing some of the first direct evidence that specific loss of α3 function in tumor cells can have pro-metastatic consequences in vivo.


Subject(s)
Fibroblasts/immunology , Integrin alpha3beta1/metabolism , Lung Neoplasms/prevention & control , Prostatic Neoplasms/prevention & control , Stromal Cells/immunology , Animals , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Coculture Techniques , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Immunoprecipitation , Integrin alpha3beta1/antagonists & inhibitors , Integrin alpha3beta1/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Cells, Cultured
14.
Am J Pathol ; 179(1): 502-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703427

ABSTRACT

Prostate-specific deletion of Pten in mice has been reported to recapitulate histological progression of human prostate cancer. To improve on this model, we introduced the conditional ROSA26 luciferase reporter allele to monitor prostate cancer progression via bioluminescence imaging and extensively backcrossed mice onto the albino C57BL/6 genetic background to address variability in tumor kinetics and to enhance imaging sensitivity. Bioluminescence signal increased rapidly in Pten(p-/-) mice from 3 to 11 weeks, but was much slower from 11 to 52 weeks. Changes in bioluminescence signal were correlated with epithelial proliferation. Magnetic resonance imaging revealed progressive increases in prostate volume, which were attributed to excessive fluid retention in the anterior prostate and to expansion of the stroma. Development of invasive prostate cancer in 52-week-old Pten(p-/-) mice was rare, indicating that disease progression was slowed relative to that in previous reports. Tumors in these mice exhibited a spontaneous inflammatory phenotype and were rapidly infiltrated by myeloid-derived suppressor cells. Although Pten(p-/-) tumors responded to androgen withdrawal, they failed to exhibit relapsed growth for up to 1 year. Taken together, these data identify a mild prostate cancer phenotype in C57BL/6 prostate-specific Pten-deficient mice, reflecting effects of the C57BL/6 genetic background on cancer progression. This model provides a platform for noninvasive assessment of how genetic and environmental risk factors may affect disease progression.


Subject(s)
Disease Models, Animal , Inflammation/pathology , PTEN Phosphohydrolase/physiology , Prostate/pathology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Animals , Cell Proliferation , Disease Progression , Fluorescent Antibody Technique , Humans , Integrases , Luciferases/metabolism , Lymphatic Metastasis , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Prostate/metabolism , Prostatic Neoplasms/genetics , Time Factors
15.
Prostate ; 71(11): 1139-50, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21656824

ABSTRACT

BACKGROUND: Prostatitis is a poorly understood disease and increasing evidence suggests inflammation is involved in other prostatic diseases including prostate cancer. METHODS: The ability of pre-activated CD8 T cells to induce prostatitis was examined by adoptive transfer of prostate antigen specific CD8 T cells into POET-3 mice or POET-3/Luc/Pten(-/+) mice. Characterization of the inflammatory response was determined by examining leukocyte infiltration by histological analysis, flow cytometry and by evaluating cytokine and chemokine levels in prostate tissue. The impact of inflammation on the prostate was evaluated by monitoring epithelial cell proliferation over time. RESULTS: Initiation of inflammation by ovalbumin specific CD8⁺ T cells (OT-I cells) resulted in development of acute prostatitis in the anterior, dorsolateral and ventral prostate of POET-3 and POET-3/Luc/Pten(-/+) mice. Acute prostatitis was characterized by recruitment of adoptively transferred OT-I cells and importantly, autologous CD4⁺ and CD8⁺ T cells, myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg). In concert with leukocyte infiltration elevated levels of pro-inflammatory cytokines and chemokines were observed. Inflammation also resulted in marked epithelial cell proliferation that was sustained up to 80 days post adoptive transfer of OT-I cells. CONCLUSIONS: The POET-3 model represents a novel mouse model to study both acute and chronic prostate inflammation in an antigen-specific system. Further, the POET-3 mouse model can be crossed with other genetic models of disease such as the C57/Luc/Pten(-/-) model of prostate cancer, allowing the impact of prostatitis on other prostatic diseases to be evaluated.


Subject(s)
Cell Proliferation , Disease Models, Animal , Epitopes/biosynthesis , Prostate/immunology , Prostate/pathology , Prostatitis/immunology , Prostatitis/pathology , Acute Disease , Animals , Chronic Disease , Epitopes/immunology , Inflammation/diagnosis , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic
16.
Mol Ther ; 16(12): 1995-2001, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18781145

ABSTRACT

A significant barrier to the successful general development of small-interfering RNA (siRNA) therapeutics is the ability to deliver them systemically to target organs and cell types. In this study, we have developed a mouse strain that will facilitate the evaluation of the efficacy of siRNA delivery strategies. This strain contains robust ubiquitous expression of firefly luciferase from germ line Cre-mediated recombination of the ROSA26-LSL-Luc allele. We show that luciferase is highly and uniformly expressed in all tissues examined. Using this mouse model, we describe a facile assay that enables the assessment of the pharmacodynamics of a systemically delivered siRNA formulation. These mice can also be used as universal donors, enabling the efficient and sensitive monitoring of cell trafficking or tissue transplantation. The primary advantage of this approach is that siRNA efficacy against a nonessential target can be easily evaluated in any tissue. This strain should generally enhance the ability to rapidly screen, compare and optimize various siRNA formulations for tissue-targeted or -enhanced systemic delivery in a preclinical development setting.


Subject(s)
Genes, Reporter/genetics , Luciferases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Animals , Female , Gene Expression/drug effects , Gene Expression/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Luciferases/metabolism , Male , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology
17.
Cancer Res ; 67(21): 10445-54, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17974988

ABSTRACT

Bioluminescence imaging is widely used to evaluate tumor growth and response to therapy in living animals. In cells expressing luciferase under the control of a constitutive promoter, light output in part depends on viable cell number, so that changes in bioluminescence intensity may be correlated with changes in viable tumor mass over time. We have found that treatment of cancer cell lines expressing luciferase under control of the cytomegalovirus (CMV) promoter with staurosporine, doxorubicin, and paclitaxel results in a transient increase in bioluminescence, which is positively correlated with apoptosis and inversely correlated with cell viability. In contrast, similar treatment of cell lines expressing luciferase under control of the SV40 promoter did not exhibit this result. We found that low doses of staurosporine induced bioluminescence in CMV- but not SV40-driven luciferase cell lines, whereas high doses elicited induction in both, indicating promoter-dependent and promoter-independent mechanisms of bioluminescence induction. The promoter-dependent increase in bioluminescence intensity from CMV-driven luciferase is a result of induction of luciferase mRNA and protein expression. We extended these findings in vivo; doxorubicin treatment resulted in a transient induction in bioluminescence when normalized to tumor volume in CMV- but not SV40-driven luciferase-expressing xenografts. We found that inhibition of the p38 mitogen-activated protein kinase pathway blocked bioluminescence induction by doxorubicin, paclitaxel, and staurosporine in CMV-driven luciferase-expressing cells. These findings have important implications when using bioluminescence to monitor the efficacy of anticancer therapy and underscore the complex regulation of the CMV promoter, which is widely used for high-level protein expression in mammalian cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cytomegalovirus/genetics , Neoplasms/drug therapy , Promoter Regions, Genetic/drug effects , Animals , Apoptosis/drug effects , Cell Line , Doxorubicin/pharmacology , Humans , Luminescence , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Biosynthesis , Staurosporine/pharmacology , Up-Regulation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...