Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Sci Rep ; 9(1): 2084, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30765854

ABSTRACT

Bladder cancer (BC) is heterogeneous and expresses various cell surface targets. Photoimmunotherapy (PIT) involves monoclonal antibodies (MAbs) conjugated to a photoabsorber (PA), IR Dye 700Dx, and then activated by near infra-red light (NIR) to specifically target tumors. We have demonstrated that tumors expressing EGFR can be targeted with PIT. However, PIT may be less effective when a tumor lacks "overwhelming" expression of a single target such as EGFR. We present a combinatorial PIT approach for targeting BC expressing EGFR and HER2, using PA- labeled panitumumab (pan) and trastuzumab (tra), respectively. Human BC tissues and cell lines were analyzed for EGFR and HER2 expression. Efficacy of PA-labeled MAbs singly and in combination was analyzed. About 45% of BC tissues stain for both EGFR and HER2. In vitro, the combination of pan IR700 and tra IR700 with NIR was more efficacious than either agent alone. Tumor xenografts treated with combination PIT showed significant tumor growth retardation. Combination PIT is a promising approach for treating BC with low/moderate expression of surface receptors. In addition, given the molecular heterogeneity of bladder cancer, targeting more than one surface receptor may allow for more effective cell death across different bladder tumors.


Subject(s)
ErbB Receptors/metabolism , Phototherapy/methods , Receptor, ErbB-2/metabolism , Urinary Bladder Neoplasms/genetics , Animals , Antibodies, Monoclonal , Antineoplastic Agents, Immunological , Cell Line, Tumor , ErbB Receptors/genetics , Female , Humans , Immunotherapy/methods , Infrared Rays , Mice, Nude , Panitumumab/pharmacology , Photosensitizing Agents , Receptor, ErbB-2/genetics , Trastuzumab/pharmacology , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor Assays
2.
J Clin Invest ; 128(7): 2927-2943, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29863500

ABSTRACT

Although nonmalignant stromal cells facilitate tumor growth and can occupy up to 90% of a solid tumor mass, better strategies to exploit these cells for improved cancer therapy are needed. Here, we describe a potent MMAE-linked antibody-drug conjugate (ADC) targeting tumor endothelial marker 8 (TEM8, also known as ANTXR1), a highly conserved transmembrane receptor broadly overexpressed on cancer-associated fibroblasts, endothelium, and pericytes. Anti-TEM8 ADC elicited potent anticancer activity through an unexpected killing mechanism we term DAaRTS (drug activation and release through stroma), whereby the tumor microenvironment localizes active drug at the tumor site. Following capture of ADC prodrug from the circulation, tumor-associated stromal cells release active MMAE free drug, killing nearby proliferating tumor cells in a target-independent manner. In preclinical studies, ADC treatment was well tolerated and induced regression and often eradication of multiple solid tumor types, blocked metastatic growth, and prolonged overall survival. By exploiting TEM8+ tumor stroma for targeted drug activation, these studies reveal a drug delivery strategy with potential to augment therapies against multiple cancer types.


Subject(s)
Immunoconjugates/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Cell Surface/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Brentuximab Vedotin , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, SCID , Microfilament Proteins , Neoplasms/metabolism , Receptors, Peptide/antagonists & inhibitors , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , Stromal Cells/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
3.
Mol Carcinog ; 56(12): 2643-2662, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28762556

ABSTRACT

NADPH oxidase 5 (NOX5) generated reactive oxygen species (ROS) have been implicated in signaling cascades that regulate cancer cell proliferation. To evaluate and validate NOX5 expression in human tumors, we screened a broad range of tissue microarrays (TMAs), and report substantial overexpression of NOX5 in malignant melanoma and cancers of the prostate, breast, and ovary. In human UACC-257 melanoma cells that possesses high levels of functional endogenous NOX5, overexpression of NOX5 resulted in enhanced cell growth, increased numbers of BrdU positive cells, and increased γ-H2AX levels. Additionally, NOX5-overexpressing (stable and inducible) UACC-257 cells demonstrated increased normoxic HIF-1α expression and decreased p27Kip1 expression. Similarly, increased normoxic HIF-1α expression and decreased p27Kip1 expression were observed in stable NOX5-overexpressing clones of KARPAS 299 human lymphoma cells and in the human prostate cancer cell line, PC-3. Conversely, knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased cell growth, decreased HIF-1α expression, and increased p27Kip1 expression. Likewise, in an additional human melanoma cell line, WM852, and in PC-3 cells, transient knockdown of endogenous NOX5 resulted in increased p27Kip1 and decreased HIF-1α expression. Knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased Akt and GSK3ß phosphorylation, signaling pathways known to modulate p27Kip1 levels. In summary, our findings suggest that NOX5 expression in human UACC-257 melanoma cells could contribute to cell proliferation due, in part, to the generation of high local concentrations of extracellular ROS that modulate multiple pathways that regulate HIF-1α and networks that signal through Akt/GSK3ß/p27Kip1 .


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NADPH Oxidase 5/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , NADPH Oxidase 5/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
4.
Mol Cancer Ther ; 16(10): 2201-2214, 2017 10.
Article in English | MEDLINE | ID: mdl-28619755

ABSTRACT

The use of light as a means of therapy for bladder cancer has a long history but has been hampered by a lack of tumor specificity and therefore, damage to the normal bladder mucosa. Here, we describe a targeted form of phototherapy called photoimmunotherapy (PIT), which targets EGFR-expressing bladder cancer. Anti-EGFR antibody panitumumab was labeled with the photoabsorber (PA), IRDye 700Dx (IR700), to create a panitumumab-IR700 antibody-PA conjugate that is activated by near-infrared radiation (NIR). Bladder cancer tissue microarray (TMA) and bladder cancer cell lines were analyzed for expression of EGFR. Mechanism of PIT-induced cell death was studied using proliferation assays, transmission electron microscopy (TEM), and production of reactive oxygen species. Finally, the in vivo effect was studied in xenografts. EGFR staining of TMAs showed that while most bladder cancers have expression of EGFR to a varying degree, squamous cell carcinomas (SCC) have the highest expression of EGFR. Panitumumab-IR700 activated by NIR light rapidly killed UMUC-5 cells, a bladder SCC line. Panitumumab alone, panitumumab-IR700 without NIR, or NIR alone had no effect on cells. TEM demonstrated that cell death is due to necrosis. Singlet oxygen species contributed toward cell death. NIR-PIT with panitumumab-IR700 reduced growth compared with only panitumumab-IR700-treated UMUC-5 xenograft tumors. PIT is a new targeted treatment for bladder cancer. Panitumumab-IR700-induced PIT selectively kills EGFR-expressing bladder cancer cells in vitro and in vivo and therefore warrants further therapeutic studies in orthotopic xenografts of bladder cancer and ultimately in patients. Mol Cancer Ther; 16(10); 2201-14. ©2017 AACR.


Subject(s)
ErbB Receptors/genetics , Immunotherapy , Phototherapy , Urinary Bladder Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Humans , Immunoconjugates/administration & dosage , Infrared Rays , Mice , Panitumumab , Photosensitizing Agents/administration & dosage , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
Oncotarget ; 8(19): 30621-30643, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28430642

ABSTRACT

Effective drug development to combat metastatic disease in breast cancer would be aided by the availability of well-characterized preclinical animal models that (a) metastasize with high efficiency, (b) metastasize in a reasonable time-frame, (c) have an intact immune system, and (d) capture some of the heterogeneity of the human disease. To address these issues, we have assembled a panel of twelve mouse mammary cancer cell lines that can metastasize efficiently on implantation into syngeneic immunocompetent hosts. Genomic characterization shows that more than half of the 30 most commonly mutated genes in human breast cancer are represented within the panel. Transcriptomically, most of the models fall into the luminal A or B intrinsic molecular subtypes, despite the predominance of an aggressive, poorly-differentiated or spindled histopathology in all models. Patterns of immune cell infiltration, proliferation rates, apoptosis and angiogenesis differed significantly among models. Inherent within-model variability of the metastatic phenotype mandates large cohort sizes for intervention studies but may also capture some relevant non-genetic sources of variability. The varied molecular and phenotypic characteristics of this expanded panel of models should aid in model selection for development of antimetastatic therapies in vivo, and serve as a useful platform for predictive biomarker identification.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , Mammary Neoplasms, Experimental , Allografts , Animals , Biomarkers, Tumor , Biopsy , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Genomics/methods , Heterografts , Humans , Immunohistochemistry , Mice , Molecular Targeted Therapy , Neoplasm Metastasis , Polymorphism, Single Nucleotide
6.
J Biol Chem ; 292(19): 7866-7887, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28330872

ABSTRACT

Reactive oxygen species (ROS) play a critical role in cell signaling and proliferation. NADPH oxidase 1 (NOX1), a membrane-bound flavin dehydrogenase that generates O2̇̄, is highly expressed in colon cancer. To investigate the role that NOX1 plays in colon cancer growth, we used shRNA to decrease NOX1 expression stably in HT-29 human colon cancer cells. The 80-90% decrease in NOX1 expression achieved by RNAi produced a significant decline in ROS production and a G1/S block that translated into a 2-3-fold increase in tumor cell doubling time without increased apoptosis. The block at the G1/S checkpoint was associated with a significant decrease in cyclin D1 expression and profound inhibition of mitogen-activated protein kinase (MAPK) signaling. Decreased steady-state MAPK phosphorylation occurred concomitant with a significant increase in protein phosphatase activity for two colon cancer cell lines in which NOX1 expression was knocked down by RNAi. Diminished NOX1 expression also contributed to decreased growth, blood vessel density, and VEGF and hypoxia-inducible factor 1α (HIF-1α) expression in HT-29 xenografts initiated from NOX1 knockdown cells. Microarray analysis, supplemented by real-time PCR and Western blotting, revealed that the expression of critical regulators of cell proliferation and angiogenesis, including c-MYC, c-MYB, and VEGF, were down-regulated in association with a decline in hypoxic HIF-1α protein expression downstream of silenced NOX1 in both colon cancer cell lines and xenografts. These studies suggest a role for NOX1 in maintaining the proliferative phenotype of some colon cancers and the potential of NOX1 as a therapeutic target in this disease.


Subject(s)
Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Cyclin D1/metabolism , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , NADPH Oxidase 1 , Neoplasm Transplantation , Phenotype , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
PLoS One ; 12(2): e0171510, 2017.
Article in English | MEDLINE | ID: mdl-28158249

ABSTRACT

Thymic epithelial cells (TEC), as part of thymic stroma, provide essential growth factors/cytokines and self-antigens to support T cell development and selection. Deletion of Rb family proteins in adult thymic stroma leads to T cell hyperplasia in vivo. To determine whether deletion of Rb specifically in keratin (K) 18 positive TEC was sufficient for thymocyte hyperplasia, we conditionally inactivated Rb and its family members p107 and p130 in K18+ TEC in genetically engineered mice (TgK18GT121; K18 mice). We found that thymocyte hyperproliferation was induced in mice with Rb inactivation in K18+ TEC, while normal T cell development was maintained; suggesting that inactivation of Rb specifically in K18+ TEC was sufficient and responsible for the phenotype. Transplantation of wild type bone marrow cells into mice with Rb inactivation in K18+ TEC resulted in donor T lymphocyte hyperplasia confirming the non-cell autonomous requirement for Rb proteins in K18+ TEC in regulating T cell proliferation. Our data suggests that thymic epithelial cells play an important role in regulating lymphoid proliferation and thymus size.


Subject(s)
Cell Proliferation , Keratin-18/metabolism , Retinoblastoma Protein/physiology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Female , Gene Expression , Male , Mice , Mice, Transgenic , Retinoblastoma Protein/genetics , Stromal Cells/metabolism , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Transgenes
8.
Hum Mol Genet ; 26(2): 354-366, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28007907

ABSTRACT

Germline H255Y and K508R missense mutations in the folliculin (FLCN) gene have been identified in patients with bilateral multifocal (BMF) kidney tumours and clinical manifestations of Birt-Hogg-Dubé (BHD) syndrome, or with BMF kidney tumours as the only manifestation; however, their impact on FLCN function remains to be determined. In order to determine if FLCN H255Y and K508R missense mutations promote aberrant kidney cell proliferation leading to pathogenicity, we generated mouse models expressing these mutants using BAC recombineering technology and investigated their ability to rescue the multi-cystic phenotype of Flcn-deficient mouse kidneys. Flcn H255Y mutant transgene expression in kidney-targeted Flcn knockout mice did not rescue the multi-cystic kidney phenotype. However, expression of the Flcn K508R mutant transgene partially, but not completely, abrogated the phenotype. Notably, expression of the Flcn K508R mutant transgene in heterozygous Flcn knockout mice resulted in development of multi-cystic kidneys and cardiac hypertrophy in some mice. These results demonstrate that both FLCN H255Y and K508R missense mutations promote aberrant kidney cell proliferation, but to different degrees. Based on the phenotypes of our preclinical models, the FLCN H255Y mutant protein has lost it tumour suppressive function leading to the clinical manifestations of BHD, whereas the FLCN K508R mutant protein may have a dominant negative effect on the function of wild-type FLCN in regulating kidney cell proliferation and, therefore, act as an oncoprotein. These findings may provide mechanistic insight into the role of FLCN in regulating kidney cell proliferation and facilitate the development of novel therapeutics for FLCN-deficient kidney cancer.


Subject(s)
Birt-Hogg-Dube Syndrome/genetics , Kidney Diseases, Cystic/genetics , Kidney Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Birt-Hogg-Dube Syndrome/pathology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Humans , Kidney/pathology , Kidney Diseases, Cystic/pathology , Kidney Neoplasms/pathology , Mice , Mice, Knockout , Mutation, Missense
9.
Oncotarget ; 7(42): 68412-68433, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27637085

ABSTRACT

Several NADPH oxidase family members, including dual oxidase 2 [DUOX2], are expressed in human tumors, particularly gastrointestinal cancers associated with long-standing chronic inflammation. We found previously that exposure of pancreatic ductal adenocarcinoma cells to the pro-inflammatory cytokine IFN-γ increased DUOX2 expression (but not other NADPH oxidases) leading to long-lived H2O2 production. To elucidate the pathophysiology of DUOX2-mediated H2O2 formation in the pancreas further, we demonstrate here that IFN-γ-treated BxPC-3 and CFPAC-1 pancreatic cancer cells (known to increase DUOX2 expression) produce significant levels of intracellular oxidants and extracellular H2O2 which correlate with concomitant up-regulation of VEGF-A and HIF-1α transcription. These changes are not observed in the PANC-1 line that does not increase DUOX2 expression following IFN-γ treatment. DUOX2 knockdown with short interfering RNA significantly decreased IFN-γ-induced VEGF-A or HIF-1α up-regulation, as did treatment of pancreatic cancer cells with the NADPH oxidase inhibitor diphenylene iodonium, the multifunctional reduced thiol N-acetylcysteine, and the polyethylene glycol-modified form of the hydrogen peroxide detoxifying enzyme catalase. Increased DUOX2-related VEGF-A expression appears to result from reactive oxygen-mediated activation of ERK signaling that is responsible for AP-1-related transcriptional effects on the VEGF-A promoter. To clarify the relevance of these observations in vivo, we demonstrate that many human pre-malignant pancreatic intraepithelial neoplasms and frank pancreatic cancers express substantial levels of DUOX protein compared to histologically normal pancreatic tissues, and that expression of both DUOX2 and VEGF-A mRNAs is significantly increased in surgically-resected pancreatic cancers compared to the adjacent normal pancreas.


Subject(s)
Adenocarcinoma/genetics , Dual Oxidases/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Pancreatic Neoplasms/genetics , Vascular Endothelial Growth Factor A/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Dual Oxidases/antagonists & inhibitors , Dual Oxidases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interferon-gamma/pharmacology , Mice, Nude , Onium Compounds/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA Interference , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism
10.
PLoS Genet ; 12(8): e1006236, 2016 08.
Article in English | MEDLINE | ID: mdl-27490902

ABSTRACT

Human breast cancer susceptibility gene, BRCA2, encodes a 3418-amino acid protein that is essential for maintaining genomic integrity. Among the proteins that physically interact with BRCA2, Partner and Localizer of BRCA2 (PALB2), which binds to the N-terminal region of BRCA2, is vital for its function by facilitating its subnuclear localization. A functional redundancy has been reported between this N-terminal PALB2-binding domain and the C-terminal DNA-binding domain of BRCA2, which undermines the relevance of the interaction between these two proteins. Here, we describe a genetic approach to examine the functional significance of the interaction between BRCA2 and PALB2 by generating a knock-in mouse model of Brca2 carrying a single amino acid change (Gly25Arg, Brca2G25R) that disrupts this interaction. In addition, we have combined Brca2G25R homozygosity as well as hemizygosity with Palb2 and Trp53 heterozygosity to generate an array of genotypically and phenotypically distinct mouse models. Our findings reveal defects in body size, fertility, meiotic progression, and genome stability, as well as increased tumor susceptibility in these mice. The severity of the phenotype increased with a decrease in the interaction between BRCA2 and PALB2, highlighting the significance of this interaction. In addition, our findings also demonstrate that hypomorphic mutations such as Brca2G25R have the potential to be more detrimental than the functionally null alleles by increasing genomic instability to a level that induces tumorigenesis, rather than apoptosis.


Subject(s)
BRCA2 Protein/genetics , Breast Neoplasms/genetics , Nuclear Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Apoptosis/genetics , BRCA1 Protein/genetics , BRCA2 Protein/metabolism , Breast Neoplasms/pathology , Carcinogenesis/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Fanconi Anemia Complementation Group N Protein , Female , Gene Knock-In Techniques , Genetic Predisposition to Disease , Genomic Instability/genetics , Humans , Mice , Mutation , Nuclear Proteins/metabolism , Protein Interaction Domains and Motifs , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
11.
Oncotarget ; 7(10): 11094-112, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26848978

ABSTRACT

The oncogenic role of microRNA-155 (miR-155) in leukemia is well established but its role in other cancers, especially breast cancer, is gradually emerging. In this study we examined the effect of mir-155 loss in a well-characterized spontaneous breast cancer mouse model where Brca1 and Trp53 are deleted by K14-Cre. miR-155 is known to be up-regulated in BRCA1-deficient tumors. Surprisingly, complete loss of miR-155 (miR-155ko/ko) did not alter the tumor free survival of the mutant mice. However, we found increased infiltration of myeloid derived suppressor cells (MDSCs) in miR-155 deficient tumors. In addition, cytokine/chemokine array analysis revealed altered level of cytokines that are implicated in the recruitment of MDSCs. Mechanistically, we identified C/EBP-ß, a known miR-155 target, to regulate the expression of these cytokines in the miR-155-deficient cells. Furthermore, using an allograft model, we showed that inhibition of miR-155 in cancer cells suppressed in vivo growth, which was restored by the loss of miR-155 in the microenvironment. Taken together, we have uncovered a novel tumor suppressive function of miR-155 in the tumor microenvironment, which is also dependent on miR-155 expression in the tumor cells. Because of the oncogenic as well as tumor suppressive roles of miR-155, our findings warrant caution against a systemic inhibition of miR-155 for anticancer therapy.


Subject(s)
Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , MicroRNAs , Myeloid-Derived Suppressor Cells/immunology , Tumor Microenvironment/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Female , Gene Knockdown Techniques , Mice , Mice, Mutant Strains , Oncogenes
12.
Hum Mol Genet ; 25(10): 1934-1945, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26920070

ABSTRACT

The breast cancer gene, BRCA2, is essential for viability, yet patients with Fanconi anemia-D1 subtype are born alive with biallelic mutations in this gene. The hypomorphic nature of the mutations is believed to support viability, but this is not always apparent. One such mutation is IVS7+2T>G, which causes premature protein truncation due to skipping of exon 7. We previously identified a transcript lacking exons 4-7, which restores the open-reading frame, encodes a DNA repair proficient protein and is expressed in IVS7+2T>G carriers. However, because the exons 4-7 encoded region contains several residues required for normal cell-cycle regulation and cytokinesis, this transcript's ability to support viability can be argued. To address this, we generated a Brca2 knock-in mouse model lacking exons 4-7 and demonstrated that these exons are dispensable for viability as well as tumor-free survival. This study provides the first in vivo evidence of the functional significance of a minor transcript of BRCA2 that can play a major role in the survival of humans who are homozygous for a clearly pathogenic mutation. Our results highlight the importance of assessing protein function restoration by premature truncating codon bypass by alternative splicing when evaluating the functional significance of variants such as nonsense and frame-shift mutations that are assumed to be clearly pathogenic. Our findings will impact not only the assessment of variants that map to this region, but also influence counseling paradigms and treatment options for such mutation carriers.


Subject(s)
BRCA2 Protein/genetics , Breast Neoplasms/genetics , Fanconi Anemia/genetics , Genetic Predisposition to Disease , Alternative Splicing/genetics , Animals , Breast Neoplasms/pathology , Exons/genetics , Fanconi Anemia/pathology , Gene Knock-In Techniques , Germ-Line Mutation , Humans , Mice , Mutation , Pedigree , RNA Splice Sites
13.
Cancer Res ; 76(5): 1055-1065, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26719530

ABSTRACT

Smokers develop metastatic prostate cancer more frequently than nonsmokers, suggesting that a tobacco-derived factor is driving metastatic progression. To identify smoking-induced alterations in human prostate cancer, we analyzed gene and protein expression patterns in tumors collected from current, past, and never smokers. By this route, we elucidated a distinct pattern of molecular alterations characterized by an immune and inflammation signature in tumors from current smokers that were either attenuated or absent in past and never smokers. Specifically, this signature included elevated immunoglobulin expression by tumor-infiltrating B cells, NF-κB activation, and increased chemokine expression. In an alternate approach to characterize smoking-induced oncogenic alterations, we also explored the effects of nicotine in human prostate cancer cells and prostate cancer-prone TRAMP mice. These investigations showed that nicotine increased glutamine consumption and invasiveness of cancer cells in vitro and accelerated metastatic progression in tumor-bearing TRAMP mice. Overall, our findings suggest that nicotine is sufficient to induce a phenotype resembling the epidemiology of smoking-associated prostate cancer progression, illuminating a novel candidate driver underlying metastatic prostate cancer in current smokers.


Subject(s)
Inflammation/metabolism , Prostatic Neoplasms/immunology , Smoking/adverse effects , Transcriptome , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Immunoglobulins/genetics , Interleukin-8/blood , Male , Mice , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Nicotine/pharmacology , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism
14.
Mol Cell Biol ; 36(5): 693-713, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26667036

ABSTRACT

The integrated stress response (ISR) controls cellular adaptations to nutrient deprivation, redox imbalances, and endoplasmic reticulum (ER) stress. ISR genes are upregulated in stressed cells, primarily by the bZIP transcription factor ATF4 through its recruitment to cis-regulatory C/EBP:ATF response elements (CAREs) together with a dimeric partner of uncertain identity. Here, we show that C/EBPγ:ATF4 heterodimers, but not C/EBPß:ATF4 dimers, are the predominant CARE-binding species in stressed cells. C/EBPγ and ATF4 associate with genomic CAREs in a mutually dependent manner and coregulate many ISR genes. In contrast, the C/EBP family members C/EBPß and C/EBP homologous protein (CHOP) were largely dispensable for induction of stress genes. Cebpg(-/-) mouse embryonic fibroblasts (MEFs) proliferate poorly and exhibit oxidative stress due to reduced glutathione levels and impaired expression of several glutathione biosynthesis pathway genes. Cebpg(-/-) mice (C57BL/6 background) display reduced body size and microphthalmia, similar to ATF4-null animals. In addition, C/EBPγ-deficient newborns die from atelectasis and respiratory failure, which can be mitigated by in utero exposure to the antioxidant, N-acetyl-cysteine. Cebpg(-/-) mice on a mixed strain background showed improved viability but, upon aging, developed significantly fewer malignant solid tumors than WT animals. Our findings identify C/EBPγ as a novel antioxidant regulator and an obligatory ATF4 partner that controls redox homeostasis in normal and cancerous cells.


Subject(s)
Activating Transcription Factor 4/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Oxidative Stress , Activating Transcription Factor 4/analysis , Activating Transcription Factor 4/genetics , Animals , CCAAT-Enhancer-Binding Proteins/analysis , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Female , Fetus/abnormalities , Fetus/metabolism , Gene Deletion , Gene Expression Regulation , Glutathione/metabolism , Humans , Male , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/metabolism , Protein Multimerization , Response Elements , Transcription Factor CHOP/metabolism
15.
Proc Natl Acad Sci U S A ; 112(13): E1624-31, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25775561

ABSTRACT

Folliculin (FLCN)-interacting proteins 1 and 2 (FNIP1, FNIP2) are homologous binding partners of FLCN, a tumor suppressor for kidney cancer. Recent studies have revealed potential functions for Flcn in kidney; however, kidney-specific functions for Fnip1 and Fnip2 are unknown. Here we demonstrate that Fnip1 and Fnip2 play critical roles in kidney tumor suppression in cooperation with Flcn. We observed no detectable phenotype in Fnip2 knockout mice, whereas Fnip1 deficiency produced phenotypes similar to those seen in Flcn-deficient mice in multiple organs, but not in kidneys. We found that absolute Fnip2 mRNA copy number was low relative to Fnip1 in organs that showed phenotypes under Fnip1 deficiency but was comparable to Fnip1 mRNA copy number in mouse kidney. Strikingly, kidney-targeted Fnip1/Fnip2 double inactivation produced enlarged polycystic kidneys, as was previously reported in Flcn-deficient kidneys. Kidney-specific Flcn inactivation did not further augment kidney size or cystic histology of Fnip1/Fnip2 double-deficient kidneys, suggesting pathways dysregulated in Flcn-deficient kidneys and Fnip1/Fnip2 double-deficient kidneys are convergent. Heterozygous Fnip1/homozygous Fnip2 double-knockout mice developed kidney cancer at 24 mo of age, analogous to the heterozygous Flcn knockout mouse model, further supporting the concept that Fnip1 and Fnip2 are essential for the tumor-suppressive function of Flcn and that kidney tumorigenesis in human Birt-Hogg-Dubé syndrome may be triggered by loss of interactions among Flcn, Fnip1, and Fnip2. Our findings uncover important roles for Fnip1 and Fnip2 in kidney tumor suppression and may provide molecular targets for the development of novel therapeutics for kidney cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Alleles , Animals , Apoptosis Regulatory Proteins/genetics , Birt-Hogg-Dube Syndrome/genetics , Carrier Proteins/genetics , Disease Models, Animal , Female , Kidney/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Polycystic Kidney Diseases/metabolism , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
16.
Cancer Lett ; 355(2): 232-41, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25242356

ABSTRACT

Cancer pain is a deleterious consequence of tumor growth and related inflammation. Opioids and anti-inflammatory drugs provide first line treatment for cancer pain, but both are limited by side effects. Fufang Kushen injection (FKI) is GMP produced, traditional Chinese medicine used alone or with chemotherapy to reduce cancer-associated pain. FKI limited mouse sarcoma growth both in vivo and in vitro, in part, by reducing the phosphorylation of ERK and AKT kinases and BAD. FKI inhibited TRPV1 mediated capsaicin-induced ERK phosphorylation and reduced tumor-induced proinflammatory cytokine production. Thus, FKI limited cancer pain both directly by blocking TRPV1 signaling and indirectly by reducing tumor growth.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hyperalgesia/drug therapy , MAP Kinase Signaling System/drug effects , Sarcoma/complications , Sarcoma/drug therapy , TRPV Cation Channels/metabolism , Animals , Capsaicin/metabolism , Cell Line, Tumor , Cytokines/metabolism , Female , Hyperalgesia/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Sarcoma/enzymology , Sarcoma/metabolism
17.
Hum Mol Genet ; 23(21): 5706-19, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24908670

ABSTRACT

Cardiac hypertrophy, an adaptive process that responds to increased wall stress, is characterized by the enlargement of cardiomyocytes and structural remodeling. It is stimulated by various growth signals, of which the mTORC1 pathway is a well-recognized source. Here, we show that loss of Flcn, a novel AMPK-mTOR interacting molecule, causes severe cardiac hypertrophy with deregulated energy homeostasis leading to dilated cardiomyopathy in mice. We found that mTORC1 activity was upregulated in Flcn-deficient hearts, and that rapamycin treatment significantly reduced heart mass and ameliorated cardiac dysfunction. Phospho-AMP-activated protein kinase (AMPK)-alpha (T172) was reduced in Flcn-deficient hearts and nonresponsive to various stimulations including metformin and AICAR (5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide). ATP levels were elevated and mitochondrial function was increased in Flcn-deficient hearts, suggesting that excess energy resulting from up-regulated mitochondrial metabolism under Flcn deficiency might attenuate AMPK activation. Expression of Ppargc1a, a central molecule for mitochondrial metabolism, was increased in Flcn-deficient hearts and indeed, inactivation of Ppargc1a in Flcn-deficient hearts significantly reduced heart mass and prolonged survival. Ppargc1a inactivation restored phospho-AMPK-alpha levels and suppressed mTORC1 activity in Flcn-deficient hearts, suggesting that up-regulated Ppargc1a confers increased mitochondrial metabolism and excess energy, leading to inactivation of AMPK and activation of mTORC1. Rapamycin treatment did not affect the heart size of Flcn/Ppargc1a doubly inactivated hearts, further supporting the idea that Ppargc1a is the critical element leading to deregulation of the AMPK-mTOR-axis and resulting in cardiac hypertrophy under Flcn deficiency. These data support an important role for Flcn in cardiac homeostasis in the murine model.


Subject(s)
Cardiomegaly/genetics , Cardiomegaly/metabolism , Estrone/genetics , Gene Silencing , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cardiomegaly/complications , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cell Line , Disease Models, Animal , Enzyme Activation , Heart Failure/etiology , Heart Failure/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Mitochondrial Turnover , Organ Size/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Signal Transduction , Sirolimus/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Ventricular Function/drug effects
18.
Sci Transl Med ; 6(242): 242ra84, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24964992

ABSTRACT

Antiangiogenic agents that block vascular endothelial growth factor (VEGF) signaling are important components of current cancer treatment modalities but are limited by alternative ill-defined angiogenesis mechanisms that allow persistent tumor vascularization in the face of continued VEGF pathway blockade. We identified prostaglandin E2 (PGE2) as a soluble tumor-derived angiogenic factor associated with VEGF-independent angiogenesis. PGE2 production in preclinical breast and colon cancer models was tightly controlled by cyclooxygenase-2 (COX-2) expression, and COX-2 inhibition augmented VEGF pathway blockade to suppress angiogenesis and tumor growth, prevent metastasis, and increase overall survival. These results demonstrate the importance of the COX-2/PGE2 pathway in mediating resistance to VEGF pathway blockade and could aid in the rapid development of more efficacious anticancer therapies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Mammary Neoplasms, Experimental/prevention & control , Mammary Neoplasms, Experimental/secondary , Xenograft Model Antitumor Assays , Angiogenesis Inhibitors/pharmacology , Animals , Axitinib , Carcinogenesis/pathology , Cell Line, Tumor , Clone Cells , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Female , Humans , Imidazoles/pharmacology , Indazoles/pharmacology , Liver Neoplasms/blood supply , Liver Neoplasms/drug therapy , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mice , Neoadjuvant Therapy , Signal Transduction/drug effects , Survival Analysis , Vascular Endothelial Growth Factor A/metabolism
19.
Int J Mol Imaging ; 2014: 102702, 2014.
Article in English | MEDLINE | ID: mdl-24724022

ABSTRACT

Metastatic spread is the leading cause of death from cancer. Early detection of cancer at primary and metastatic sites by noninvasive imaging modalities would be beneficial for both therapeutic intervention and disease management. Noninvasive imaging modalities such as bioluminescence (optical), positron emission tomography (PET)/X-ray computed tomography (CT), and magnetic resonance imaging (MRI) can provide complementary information and accurately measure tumor growth as confirmed by histopathology. Methods. We validated two metastatic tumor models, MDA-MD-231-Luc and B16-F10-Luc intravenously injected, and 4T1-Luc cells orthotopically implanted into the mammary fat pad. Longitudinal whole body bioluminescence imaging (BLI) evaluated metastasis, and tumor burden of the melanoma cell line (B16-F10-Luc) was correlated with (PET)/CT and MRI. In addition, ex vivo imaging evaluated metastasis in relevant organs and histopathological analysis was used to confirm imaging. Results. BLI revealed successful colonization of cancer cells in both metastatic tumor models over a 4-week period. Furthermore, lung metastasis of B16-F10-Luc cells imaged by PET/CT at week four showed a strong correlation (R (2) = 0.9) with histopathology. The presence and degree of metastasis as determined by imaging correlated (R (2) = 0.7) well with histopathology findings. Conclusions. We validated two metastatic tumor models by longitudinal noninvasive imaging with good histopathology correlation.

20.
Mol Cancer Res ; 12(1): 82-90, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24296759

ABSTRACT

UNLABELLED: We report that HMGN1, a nucleosome-binding protein that affects chromatin structure and function, affects the growth of N-nitrosodiethylamine (DEN)-induced liver tumors. Following a single DEN injection at 2 weeks of age, Hmgn1(tm1/tm1) mice, lacking the nucleosome-binding domain of HMGN1, had earlier signs of liver tumorigenesis than their Hmgn1(+/+) littermates. Detailed gene expression profiling revealed significant differences between DEN-injected and control saline-injected mice, but only minor differences between the injected Hmgn1(tm1/tm1) mice and their Hmgn1(+/+) littermates. Pathway analysis revealed that the most significant process affected by loss of HMGN1 involves the lipid/sterol metabolic pathway. Our study indicates that in mice, loss of HMGN1 leads to transcription changes that accelerate the progression of DEN-induced hepatocarcinogenesis, without affecting the type of tumors or the final total tumor burden of these mice. IMPLICATIONS: Loss of HMGN1 leads to accelerated progression of DEN-induced hepatocarcinogenesis in mice.


Subject(s)
Cell Transformation, Neoplastic/genetics , Diethylnitrosamine/pharmacology , HMGN1 Protein/genetics , Lipid Metabolism/genetics , Liver Neoplasms/genetics , Animals , Chromatin/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Liver/pathology , Liver Neoplasms/chemically induced , Mice , Mice, Knockout , Tumor Burden
SELECTION OF CITATIONS
SEARCH DETAIL
...