Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
PLoS One ; 18(1): e0278844, 2023.
Article En | MEDLINE | ID: mdl-36701370

Leiomyosarcoma (LMS) is an aggressive, often poorly differentiated cancer of the smooth muscle (SM) lineage for which the molecular drivers of transformation and progression are poorly understood. In microRNA (miRNA) profiling studies, miR-130b was previously found to be upregulated in LMS vs. normal SM, and down-regulated during the differentiation of mesenchymal stem cells (MSCs) into SM, suggesting a role in LMS tumor progression. In the present study, the effects of miR-130b on human LMS tumorigenesis were investigated. Stable miR-130b overexpression enhanced invasion of LMS cells in vitro, and led to the formation of undifferentiated, pleomorphic tumors in vivo, with increased growth and metastatic potential compared to control LMS cells. TSC1 was identified as a direct miR-130b target in luciferase-3'UTR assays, and shRNA-mediated knockdown of TSC1 replicated miR-130b effects. Loss-of-function and gain-of-function studies showed that miR-130b levels regulate cell morphology and motility. Following miR-130b suppression, LMS cells adopted a rounded morphology, amoeboid mode of cell movement and enhanced invasive capacity that was Rho/ROCK dependent. Conversely, miR-130b-overexpressing LMS cells exhibited Rho-independent invasion, accompanied by down-regulation of Rho-pathway effectors. In mesenchymal stem cells, both miR-130b overexpression and TSC1 silencing independently impaired SM differentiation in vitro. Together, the data reveal miR-130b as a pro-oncogenic miRNA in LMS and support a miR-130b-TSC1 regulatory network that enhances tumor progression via inhibition of SM differentiation.


Leiomyosarcoma , MicroRNAs , Humans , Cell Line, Tumor , Leiomyosarcoma/genetics , MicroRNAs/genetics , RNA, Small Interfering , Down-Regulation , Gene Expression Regulation, Neoplastic , Cell Proliferation
2.
Cells ; 9(9)2020 09 10.
Article En | MEDLINE | ID: mdl-32927859

TSC1 is a tumor suppressor that inhibits cell growth via negative regulation of the mammalian target of rapamycin complex (mTORC1). TSC1 mutations are associated with Tuberous Sclerosis Complex (TSC), characterized by multiple benign tumors of mesenchymal and epithelial origin. TSC1 modulates self-renewal and differentiation in hematopoietic stem cells; however, its effects on mesenchymal stem cells (MSCs) are unknown. We investigated the impact of Tsc1 inactivation in murine bone marrow (BM)-MSCs, using tissue-specific, transgelin (Tagln)-mediated cre-recombination, targeting both BM-MSCs and smooth muscle cells. Tsc1 mutants were viable, but homozygous inactivation led to a dwarfed appearance with TSC-like pathologies in multiple organs and reduced survival. In young (28 day old) mice, Tsc1 deficiency-induced significant cell expansion of non-hematopoietic BM in vivo, and MSC colony-forming potential in vitro, that was normalized upon treatment with the mTOR inhibitor, everolimus. The hyperproliferative BM-MSC phenotype was lost in aged (1.5 yr) mice, and Tsc1 inactivation was also accompanied by elevated ROS and increased senescence. ShRNA-mediated knockdown of Tsc1 in BM-MSCs replicated the hyperproliferative BM-MSC phenotype and led to impaired adipogenic and myogenic differentiation. Our data show that Tsc1 is a negative regulator of BM-MSC proliferation and support a pivotal role for the Tsc1-mTOR axis in the maintenance of the mesenchymal progenitor pool.


Cell Proliferation , Mesenchymal Stem Cells/cytology , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis/metabolism , Animals , Cell Differentiation , Cells, Cultured , Cellular Senescence , Female , Mice , Mice, Knockout , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/pathology
3.
Cancer Res ; 79(20): 5382-5393, 2019 10 15.
Article En | MEDLINE | ID: mdl-31405846

Neuroblastoma is a pediatric cancer that is frequently metastatic and resistant to conventional treatment. In part, a lack of natively metastatic, chemoresistant in vivo models has limited our insight into the development of aggressive disease. The Th-MYCN genetically engineered mouse model develops rapidly progressive chemosensitive neuroblastoma and lacks clinically relevant metastases. To study tumor progression in a context more reflective of clinical therapy, we delivered multicycle treatment with cyclophosphamide to Th-MYCN mice, individualizing therapy using MRI, to generate the Th-MYCN CPM32 model. These mice developed chemoresistance and spontaneous bone marrow metastases. Tumors exhibited an altered immune microenvironment with increased stroma and tumor-associated fibroblasts. Analysis of copy number aberrations revealed genomic changes characteristic of human MYCN-amplified neuroblastoma, specifically copy number gains at mouse chromosome 11, syntenic with gains on human chromosome 17q. RNA sequencing revealed enriched expression of genes associated with 17q gain and upregulation of genes associated with high-risk neuroblastoma, such as the cell-cycle regulator cyclin B1-interacting protein 1 (Ccnb1ip1) and thymidine kinase (TK1). The antiapoptotic, prometastatic JAK-STAT3 pathway was activated in chemoresistant tumors, and treatment with the JAK1/JAK2 inhibitor CYT387 reduced progression of chemoresistant tumors and increased survival. Our results highlight that under treatment conditions that mimic chemotherapy in human patients, Th-MYCN mice develop genomic, microenvironmental, and clinical features reminiscent of human chemorefractory disease. The Th-MYCN CPM32 model therefore is a useful tool to dissect in detail mechanisms that drive metastasis and chemoresistance, and highlights dysregulation of signaling pathways such as JAK-STAT3 that could be targeted to improve treatment of aggressive disease. SIGNIFICANCE: An in vivo mouse model of high-risk treatment-resistant neuroblastoma exhibits changes in the tumor microenvironment, widespread metastases, and sensitivity to JAK1/2 inhibition.


Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Genes, myc , Neoplasm Metastasis/drug therapy , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Benzamides/therapeutic use , Child , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Disease Models, Animal , Disease Progression , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Janus Kinases/antagonists & inhibitors , Magnetic Resonance Imaging , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Metastasis/diagnostic imaging , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neuroblastoma/diagnostic imaging , Neuroblastoma/genetics , Neuroblastoma/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction , Synteny , Tumor Burden , Tumor Microenvironment
4.
Brain Pathol ; 28(4): 475-483, 2018 07.
Article En | MEDLINE | ID: mdl-28481062

To assess the clinical relevance of transgenic and patient-derived xenograft models of adamantinomatous craniopharyngioma (ACP) using serial magnetic resonance imaging (MRI) and high resolution post-mortem microcomputed tomography (µ-CT), with correlation with histology and human ACP imaging. The growth patterns and radiological features of tumors arising in Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ transgenic mice, and of patient-derived ACP xenografts implanted in the cerebral cortex, were monitored longitudinally in vivo with anatomical and functional MRI, and by ex vivo µ-CT at study end. Pathological correlates with hematoxylin and eosin stained sections were investigated. Early enlargement and heterogeneity of Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ mouse pituitaries was evident at initial imaging at 8 weeks, which was followed by enlargement of a solid tumor, and development of cysts and hemorrhage. Tumors demonstrated MRI features that recapitulated those of human ACP, specifically, T1 -weighted signal enhancement in the solid tumor component following Gd-DTPA administration, and in some animals, hyperintense cysts on FLAIR and T1 -weighted images. Ex vivo µ-CT correlated with MRI findings and identified smaller cysts, which were confirmed by histology. Characteristic histological features, including wet keratin and calcification, were visible on µ-CT and verified by histological sections of patient-derived ACP xenografts. The Hesx1Cre/+ ;Ctnnb1lox(ex3)/+ transgenic mouse model and cerebral patient-derived ACP xenografts recapitulate a number of the key radiological features of the human disease and provide promising foundations for in vivo trials of novel therapeutics for the treatment of these tumors.


Craniopharyngioma/diagnostic imaging , Craniopharyngioma/pathology , Disease Models, Animal , Animals , Craniopharyngioma/genetics , Heterografts/diagnostic imaging , Heterografts/pathology , Homeodomain Proteins/genetics , Humans , Magnetic Resonance Imaging , Male , Mice, Transgenic , Middle Aged , Repressor Proteins/genetics , X-Ray Microtomography , beta Catenin/genetics
5.
Mol Oncol ; 11(8): 996-1006, 2017 08.
Article En | MEDLINE | ID: mdl-28432815

Targeted inhibition of anaplastic lymphoma kinase (ALK) is a successful approach for the treatment of many ALK-aberrant malignancies; however, the presence of resistant mutations necessitates both the development of more potent compounds and pharmacodynamic methods with which to determine their efficacy. We describe immunoassays designed to quantitate phosphorylation of ALK, and their use in preclinical models of neuroblastoma, a pediatric malignancy in which gain-of-function ALK mutations predict a poor overall outcome to conventional treatment. Validation of the immunoassays is presented using a panel of neuroblastoma cell lines and evidence of on-target ALK inhibition provided by treatment of a genetically engineered murine model of neuroblastoma with two clinical ALK inhibitors, crizotinib and ceritinib, highlighting the superior efficacy of ceritinib.


Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neuroblastoma/enzymology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Sulfones/pharmacology , Anaplastic Lymphoma Kinase , Crizotinib , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Immunoassay , Phosphorylation/drug effects
6.
Cancer Res ; 75(14): 2770-4, 2015 Jul 15.
Article En | MEDLINE | ID: mdl-26122839

Neuroblastoma is a childhood malignancy that has not yet benefitted from the rapid progress in the development of small-molecule therapeutics for cancer. An opportunity to take advantage of pharmaceutical innovation in this area arose when the identification of ALK fusion proteins in non-small cell lung cancer (NSCLC) occurred in parallel to the discovery of point mutations of ALK in neuroblastomas. ALK is now known to be a marker of poor outcome in neuroblastoma, and therefore, urgent development of specific ALK inhibitors to treat this devastating disease is a necessity. However, the translation of small molecules from adult directly into pediatric practice has thus far been challenging, due to mutation-specific structural variances in the ALK kinase domain. We discuss how the most recent structural and biological characterizations of ALK are directing preclinical and clinical studies of ALK inhibitors for both NSCLC and neuroblastoma.


Drug Discovery , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Adult , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Child , Crizotinib , Drug Discovery/methods , Drug Resistance, Neoplasm/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Receptor Protein-Tyrosine Kinases/physiology
7.
Leuk Res ; 39(3): 335-41, 2015 Mar.
Article En | MEDLINE | ID: mdl-25597017

The overexpression of microRNA cluster miR-17-92 has been implicated in development of solid tumors and hematological malignancies. The role of miR-17-92 in lymphomagenesis has been extensively investigated; however, because of the developmental defects caused by miR-17-92 dysregulation, its ability to drive tumorigenesis has remained undetermined until recently. Here we demonstrate that overexpression of miR-17-92 in a limited number of hematopoietic cells is sufficient to cause B cell malignancies. In sum, our study provides a novel and physiologically relevant model that exposes the potent ability of miR-17-92 to act as a driver of tumorigenesis.


Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/pathology , MicroRNAs/physiology , Microfilament Proteins/physiology , Muscle Proteins/physiology , Animals , Blotting, Western , Cell Proliferation , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Immunoenzyme Techniques , Integrases/metabolism , Mice , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
8.
Am J Pathol ; 182(6): 2015-27, 2013 Jun.
Article En | MEDLINE | ID: mdl-23708211

Soft tissue sarcomas are a heterogeneous group of tumors associated with poor clinical outcome. Although a subset of soft tissue sarcomas is characterized by simple karyotypes and recurrent chromosomal translocations, the mechanisms driving cytogenetically complex sarcomas are largely unknown. Clinical evidence led us to partially inactivate Pten and Tp53 in the smooth muscle lineage of mice, which developed high-grade undifferentiated pleomorphic sarcomas, leiomyosarcomas, and carcinosarcomas that widely recapitulate the human disease, including the aberrant karyotype and metastatic behavior. Pten was found haploinsufficient, whereas the wild-type allele of Tp53 invariably gained point mutations. Gene expression profiles showed up-regulated Notch signaling in Pten(Δ/+)Tp53(Δ/+) tumors compared with Pten(+/+)Tp53(Δ/+) tumors. Consistently, Pten silencing exacerbated the clonogenic and invasive potential of Tp53-deficient bone marrow-derived mouse mesenchymal stem cells and tumor cells and activated the Notch pathway. Moreover, the increased oncogenic behavior of Pten(Δ/+)Tp53(Δ/+) and shPten-transduced Pten(+/+)Tp53(Δ/+) tumor cells was counteracted by treatment with a γ-secretase inhibitor, suggesting that the aggressiveness of those tumors can be attributed, at least in part, to enhanced Notch signaling. This study demonstrates a cooperative role for Pten and Tp53 suppression in complex karyotype sarcomas while establishing Notch as an important functional player in the cross talk of these pathways during tumor progression. Our results highlight the importance of molecularly subclassifying patients with high-grade sarcoma for targeted treatments.


Genes, p53 , PTEN Phosphohydrolase/genetics , Receptors, Notch/metabolism , Sarcoma/genetics , Soft Tissue Neoplasms/genetics , Animals , DNA Mutational Analysis/methods , Disease Progression , Down-Regulation/physiology , Gene Deletion , Genotype , Haploinsufficiency , Humans , Leiomyosarcoma/genetics , Leiomyosarcoma/metabolism , Leiomyosarcoma/secondary , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/biosynthesis , Sarcoma/metabolism , Sarcoma, Experimental/genetics , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Sarcoma, Experimental/secondary , Signal Transduction/physiology , Soft Tissue Neoplasms/metabolism , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
9.
FASEB J ; 27(4): 1460-7, 2013 Apr.
Article En | MEDLINE | ID: mdl-23271053

MicroRNA cluster miR-17-92 has been implicated in cardiovascular development and function, yet its precise mechanisms of action in these contexts are uncertain. This study aimed to investigate the role of miR-17-92 in morphogenesis and function of cardiac and smooth muscle tissues. To do so, a mouse model of conditional overexpression of miR-17-92 in cardiac and smooth muscle tissues was generated. Extensive cardiac functional studies identified a dose-dependent induction of dilated, hypertrophic cardiomyopathy, and arrhythmia inducibility in transgenic animals, which correlated with premature mortality (98.3 ± 42.5 d, P<0.0001). Expression analyses revealed the abundance of Pten transcript, a known miR-17-92 target, to be inversely correlated with miR-17-92 expression levels and heart size. In addition, we demonstrated through 3'-UTR luciferase assays and expression analyses that Connexin43 (Cx43) is a novel direct target of miR-19a/b and its expression is suppressed in transgenic hearts. Taken together, these data demonstrate that dysregulated expression of miR-17-92 during cardiovascular morphogenesis results in a lethal cardiomyopathy, possibly in part through direct repression of Pten and Cx43. This study highlights the importance of miR-17-92 in both normal and pathological functions of the heart, and provides a model that may serve as a useful platform to test novel antiarrhythmic therapeutics.


Arrhythmias, Cardiac/genetics , Cardiomyopathy, Hypertrophic/genetics , MicroRNAs/genetics , Animals , Arrhythmias, Cardiac/physiopathology , Cardiomyopathy, Hypertrophic/mortality , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Connexin 43/genetics , Connexin 43/metabolism , Disease Models, Animal , Heart Defects, Congenital/genetics , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
10.
Am J Pathol ; 177(2): 908-17, 2010 Aug.
Article En | MEDLINE | ID: mdl-20558575

Smooth muscle (SM) is a spontaneously contractile tissue that provides physical support and function to organs such as the uterus. Uterine smooth muscle-related neoplasia comprise common well-differentiated benign lesions called leiomyomas (ULM), and rare, highly aggressive and pleomorphic tumors named leiomyosarcomas (ULMS). MicroRNAs (miRNAs) are small non-coding RNAs that play essential roles in normal cellular development and tissue homeostasis that can be used to accurately subclassify different tumor types. Here, we demonstrate that miRNAs are required for full smooth muscle cell (SMC) differentiation of bone marrow-derived human mesenchymal stem cells (hMSCs). We also report a miRNA signature associated with this process. Moreover, we show that this signature, along with miRNA profiles for ULMS and ULM, are able to subclassify tumors of smooth muscle origin along SM differentiation. Using multiple computational analyses, we determined that ULMS are more similar to hMSCs as opposed to ULM, which are linked with more mature SMCs and myometrium. Furthermore, a comparison of the SM differentiation and ULMS miRNA signatures identified miRNAs strictly associated with SM maturation or transformation, as well as those modulated in both processes indicating a possible dual role. These results support separate origins and/or divergent transformation pathways for ULM and ULMS, resulting in drastically different states of differentiation. In summary, this work expands on our knowledge of the regulation of SM differentiation and sarcoma pathogenesis.


Cell Differentiation/physiology , Leiomyosarcoma , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology , MicroRNAs , Smooth Muscle Tumor , Uterine Neoplasms , Cell Transformation, Neoplastic , Cluster Analysis , Female , Humans , Leiomyosarcoma/genetics , Leiomyosarcoma/pathology , Mesenchymal Stem Cells/metabolism , MicroRNAs/classification , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/pathology , Muscle, Smooth/physiology , Myometrium/cytology , Myometrium/pathology , Myometrium/physiology , Phylogeny , Smooth Muscle Tumor/genetics , Smooth Muscle Tumor/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
11.
Genetics ; 180(4): 1833-47, 2008 Dec.
Article En | MEDLINE | ID: mdl-18832352

Temporal regulation of origin activation is widely thought to explain the pattern of early- and late-replicating domains in the Saccharomyces cerevisiae genome. Recently, single-molecule analysis of replication suggested that stochastic processes acting on origins with different probabilities of activation could generate the observed kinetics of replication without requiring an underlying temporal order. To distinguish between these possibilities, we examined a clb5Delta strain, where origin firing is largely limited to the first half of S phase, to ask whether all origins nonspecifically show decreased firing (as expected for disordered firing) or if only some origins ("late" origins) are affected. Approximately half the origins in the mutant genome show delayed replication while the remainder replicate largely on time. The delayed regions can encompass hundreds of kilobases and generally correspond to regions that replicate late in wild-type cells. Kinetic analysis of replication in wild-type cells reveals broad windows of origin firing for both early and late origins. Our results are consistent with a temporal model in which origins can show some heterogeneity in both time and probability of origin firing, but clustering of temporally like origins nevertheless yields a genome that is organized into blocks showing different replication times.


Chromosomes, Fungal/genetics , Cyclin B/genetics , DNA Replication , Genome, Fungal , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Cyclin B/metabolism , DNA, Fungal/genetics , DNA, Fungal/metabolism , Models, Genetic , Mutation , S Phase , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
...