Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
BMC Cancer ; 17(1): 485, 2017 Jul 14.
Article in English | MEDLINE | ID: mdl-28709411

ABSTRACT

BACKGROUND: The serine/threonine protein kinases ROCK1 and 2 are key RhoA-mediated regulators of cell shape and cytoskeletal dynamics. These proteins perform multiple functions in vascular endothelial cell physiology and are attractive targets for cancer therapy based on their roles as oncogenes and metastatic promoters. Given their critical functions in both of these processes, we hypothesized that molecular targeting of ROCK proteins would be exceedingly effective against vascular tumors such as hemangiomas and angiosarcomas, which are neoplasms composed of aberrant endothelial cells. METHODS: In this study, we compared ROCK1 and 2 protein expression in a large panel of benign and malignant vascular tumors to that of normal vasculature. We then utilized shRNA technology to knockdown the expression of ROCK1 and 2 in SVR tumor-forming vascular cells, and evaluated tumor size and proliferation rate in a xenograft model. Finally, we employed proteomics and metabolomics to assess how knockdown of the ROCK paralogs induced alterations in protein expression/phosphorylation and metabolite concentrations in the xenograft tumors. RESULTS: Our findings revealed that ROCK1 was overexpressed in malignant vascular tumors such as hemangioendotheliomas and angiosarcomas, and ROCK2 was overexpressed in both benign and malignant vascular tumors including hemangiomas, hemangioendotheliomas, hemangiopericytomas, and angiosarcomas. shRNA-mediated knockdown of ROCK2, but not ROCK1, in xenograft vascular tumors significantly reduced tumor size and proliferative index compared to control tumors. Proteomics and metabolomics analysis of the xenograft tumors revealed both overlapping as well as unique roles for the ROCK paralogs in regulating signal transduction and metabolite concentrations. CONCLUSIONS: Collectively, these data indicate that ROCK proteins are overexpressed in diverse vascular tumors and suggest that specific targeting of ROCK2 proteins may show efficacy against malignant vascular tumors.


Subject(s)
Neoplasms/genetics , Vascular Neoplasms/genetics , rho-Associated Kinases/genetics , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/pathology , RNA, Small Interfering/genetics , Signal Transduction/genetics , Transcriptional Activation/genetics , Vascular Neoplasms/classification , Vascular Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Am J Pathol ; 185(3): 717-28, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25622542

ABSTRACT

Liposarcomas, which are malignant fatty tumors, are the second most common soft-tissue sarcomas. Several histologically defined liposarcoma subtypes exist, yet little is known about the molecular pathology that drives the diversity in these tumors. We used functional genomics to classify a panel of diverse liposarcoma cell lines based on hierarchical clustering of their gene expression profiles, indicating that liposarcoma gene expression profiles and histologic classification are not directly correlated. Boolean probability approaches based on cancer-associated properties identified differential expression in multiple genes, including MYC, as potentially affecting liposarcoma signaling networks and cancer outcome. We confirmed our method with a large panel of lipomatous tumors, revealing that MYC protein expression is correlated with patient survival. These data encourage increased reliance on genomic features in conjunction with histologic features for liposarcoma clinical characterization and lay the groundwork for using Boolean-based probabilities to identify prognostic biomarkers for clinical outcome in tumor patients.


Subject(s)
Liposarcoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Soft Tissue Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Genomics , Humans , Liposarcoma/mortality , Liposarcoma/pathology , Male , Middle Aged , Prognosis , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/pathology , Survival Rate , Transcriptome
3.
BMC Cancer ; 14: 81, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24511912

ABSTRACT

BACKGROUND: Preclinical and clinical studies have shown for decades that tumor cells demonstrate significantly enhanced sensitivity to "fever range" hyperthermia (increasing the intratumoral temperature to 42-45°C) than normal cells, although it is unknown why cancer cells exhibit this distinctive susceptibility. METHODS: To address this issue, mammary epithelial cells and three malignant breast cancer lines were subjected to hyperthermic shock and microarray, bioinformatics, and network analysis of the global transcription changes was subsequently performed. RESULTS: Bioinformatics analysis differentiated the gene expression patterns that distinguish the heat shock response of normal cells from malignant breast cancer cells, revealing that the gene expression profiles of mammary epithelial cells are completely distinct from malignant breast cancer lines following this treatment. Using gene network analysis, we identified altered expression of transcripts involved in mitotic regulators, histones, and non-protein coding RNAs as the significant processes that differed between the hyperthermic response of mammary epithelial cells and breast cancer cells. We confirmed our data via qPCR and flow cytometric analysis to demonstrate that hyperthermia specifically disrupts the expression of key mitotic regulators and G2/M phase progression in the breast cancer cells. CONCLUSION: These data have identified molecular mechanisms by which breast cancer lines may exhibit enhanced susceptibility to hyperthermic shock.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Genomics/methods , Hot Temperature , Hyperthermia, Induced/methods , Cell Line, Tumor , Disease Susceptibility/diagnosis , Female , Fever , Gene Regulatory Networks/genetics , Humans , MCF-7 Cells , Protein Array Analysis
4.
FASEB J ; 24(9): 3186-95, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20400538

ABSTRACT

The small GTPase RhoA and its downstream effectors, ROCK1 and ROCK2, regulate a number of cellular processes, including cell motility, proliferation, survival, and permeability. Pharmacological inhibitors of the Rho pathway reportedly block angiogenesis; however, the molecular details of this inhibition are largely unknown. We demonstrate that vascular endothelial growth factor-A (VEGF) rapidly induces RhoA activation in endothelial cells (ECs). Moreover, the pharmacological inhibition of ROCK1/2 using 10 microM Y-27632 (the IC(50) for this compound in ECs) strongly disrupts vasculogenesis in pluripotent embryonic stem cell cultures, VEGF-mediated regenerative angiogenesis in ex vivo retinal explants, and VEGF-mediated in vitro EC tube formation. Furthermore, using small interfering RNA knockdown and mouse heterozygote knockouts of ROCK1 and ROCK2, we provide data indicating that VEGF-driven angiogenesis is largely mediated through ROCK2. These data demonstrate that Rho/ROCK signaling is an important mediator in a number of angiogenic processes, including EC migration, survival, and cell permeability, and suggest that Rho/ROCK inhibition may prove useful for the treatment of angiogenesis-related disorders.


Subject(s)
Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Signal Transduction , Vascular Endothelial Growth Factor A/pharmacology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Amides/pharmacology , Animals , Apoptosis , Blotting, Western , Cattle , Cells, Cultured , Enzyme Activation/drug effects , Humans , Mice , Microscopy, Fluorescence , Pyridines/pharmacology , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , rho-Associated Kinases/genetics
5.
J Cell Biochem ; 111(3): 543-53, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20626031

ABSTRACT

Healthy cells, as well as benign and malignant tumors, depend upon the body's blood supply to bring in oxygen and nutrients and carry away waste products. Using this property against tumors, anti-angiogenic therapy targets the tumor vasculature with the aim of starving the tumor, and has demonstrated exceptional clinical efficacy against a number of tumors. This review discusses the current state of knowledge regarding anti-angiogenic therapies presently available to patients, and garners from both preclinical and clinical literature the benefits and side effects associated with anti-angiogenic therapies, the unfortunate mechanisms of acquired resistance to these novel therapeutics, and highlights promising next generation anti-angiogenics that may overcome the limitations encountered with first generation therapies.


Subject(s)
Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Humans , Neoplasms/blood supply
6.
Mol Cell Biol ; 25(24): 11089-101, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314529

ABSTRACT

Rho family guanine nucleotide exchange factors (GEFs) regulate diverse cellular processes including cytoskeletal reorganization, cell adhesion, and differentiation via activation of the Rho GTPases. However, no studies have yet implicated Rho-GEFs as molecular regulators of the mesenchymal cell fate decisions which occur during development and repair of tissue damage. In this study, we demonstrate that the steady-state protein level of the Rho-specific GEF GEFT is modulated during skeletal muscle regeneration and that gene transfer of GEFT into cardiotoxin-injured mouse tibialis anterior muscle exerts a powerful promotion of skeletal muscle regeneration in vivo. In order to molecularly characterize this regenerative effect, we extrapolate the mechanism of action by examining the consequence of GEFT expression in multipotent cell lines capable of differentiating into a number of cell types, including muscle and adipocyte lineages. Our data demonstrate that endogenous GEFT is transcriptionally upregulated during myogenic differentiation and downregulated during adipogenic differentiation. Exogenous expression of GEFT promotes myogenesis of C2C12 cells via activation of RhoA, Rac1, and Cdc42 and their downstream effector proteins, while a dominant-negative mutant of GEFT inhibits this process. Moreover, we show that GEFT inhibits insulin-induced adipogenesis in 3T3L1 preadipocytes. In summary, we provide the first evidence that the Rho family signaling pathways act as potential regulators of skeletal muscle regeneration and provide the first reported molecular mechanism illustrating how a mammalian Rho family GEF controls this process by modulating mesenchymal cell fate decisions.


Subject(s)
Adipogenesis , Guanine Nucleotide Exchange Factors/metabolism , Muscle Development , Muscle, Skeletal/physiology , Regeneration , 3T3-L1 Cells , Adipogenesis/genetics , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cytoplasm/chemistry , Enzyme Activation , Gene Transfer Techniques , Guanine Nucleotide Exchange Factors/analysis , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice , Muscle Development/genetics , MyoD Protein/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Regeneration/genetics , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , Transcription, Genetic , Up-Regulation , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
7.
Mol Vis ; 13: 1144-53, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17653061

ABSTRACT

PURPOSE: The small GTPases function as "molecular switches" by binding and releasing GTP to mediate downstream signaling effects. The Rho-family of GTPases is central in modulating cell differentiation and cytoskeletal changes. Since eye development requires comprehensive morphogenetic movements and extensive cellular differentiation, we hypothesize that different small GTPases may play important roles during morphogenesis of eye development. To explore this possibility, we examined the expression patterns of three major Rho-GTPases: RhoA, Rac1, and Cdc42 in embryonic, postnatal (one day after birth), and adult (two-month old) mouse eye. METHODS: Various ocular tissues were collected from embryonic, postnatal, and adult C57BL/6 mice. Western blots were conducted using total proteins extracted from cornea, retina, lens epithelial cells, and lens fiber cells of the adult mice or different fractions of rat lenses. Immunohistochemistry (IHC) was performed with 6 mum thick sections cut through the eye ball region of 11.5 pc, 14.5 pc, 17.5 pc, postnatal, and adult mice. Parallel controls were run using the rabbit preimmune and GTPase-specific antibodies blocked with saturating levels of corresponding peptide antigen. RESULTS: In the embryonic mouse eye, RhoA and Cdc42 expressions were initially detectable in all three compartments at 11.5 pc. However, Rac1 became easily detectable in these compartments at 14.5 pc. Increased levels of RhoA, Rac1, and Cdc42 were detected in the three compartments at 17.5 pc and the strongest signals for RhoA, Rac1, and Cdc42 were observed in the primary lens fiber cells at 17.5 pc. In the postnatal mouse eye, the three small GTPases were significantly expressed in both endothelial and epithelial cells of mouse cornea, epithelial cells of the ocular lens, photoreceptors, horizontal/amacrine/Muller's cells, and some ganglian cells of the retina. Much lower level of expression was observed in the corneal stroma fibroblasts, lens fiber cells, and the inner and outer plexiform layers of the mouse retina. In the adult mouse eye, all three Rho-GTPases were expressed in corneal epithelial cells and retina. However, only RhoA protein was detected in corneal endothelial cells and Rac1 protein detected in the ocular lens. CONCLUSIONS: The strong expression of the three small GTPases in the cornea, lens, and retina of mouse eye at embryonic 17.5 pc and postnatal stage suggests their important functions for the morphogenesis of the different compartments of the mouse eye. Particularly, high levels of expression of RhoA, Rac1, and Cdc42 in embryonic lens fiber cells suggest their involvement in differentiation of primary lens fiber cells. In the adult mouse eye, all three Rho-GTPases seem to be involved in differentiation of corneal epithelial cells and retina, however, RhoA alone may be required for endothelial cell differentiation and Rac1 likely plays an important role in supporting continuous lens growth and maintenance of lens transparency.


Subject(s)
Aging/metabolism , Eye/embryology , Eye/enzymology , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Cornea/embryology , Cornea/metabolism , Embryo, Mammalian/enzymology , Eye/growth & development , Immunohistochemistry , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Retina/embryology , Retina/metabolism , Tissue Distribution
8.
Oncotarget ; 7(25): 38467-86, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27509260

ABSTRACT

Angiogenesis is hallmark of clear cell renal cell carcinogenesis. Anti-angiogenic therapies have been successful in improving disease outcome; however, most patients treated with anti-angiogenic agents will eventually progress. In this study we report that clear cell renal cell carcinoma was associated with vasculogenic mimicry in both mice and human with tumor cells expressing endothelial markers in the vicinity of tumor vessels. We show that vasculogenic mimicry was efficiently targeted by sunitinib but eventually associated with tumor resistance and a more aggressive phenotype both in vitro and in vivo. Re-challenging these resistant tumors in mice, we showed that second-line treatment with everolimus particularly affected vasculogenic mimicry and tumor cell differentiation compared to sorafenib and axitinib. Finally, our results highlighted the phenotypic and genotypic changes at the tumor cell and microenvironment levels during sunitinib response and progression and the subsequent improvement second-line therapies bring to the current renal cell carcinoma treatment paradigm.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Everolimus/pharmacology , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Pyrroles/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Axitinib , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/pathology , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm , Female , Humans , Imidazoles/pharmacology , Indazoles/pharmacology , Kidney Neoplasms/blood supply , Kidney Neoplasms/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Random Allocation , Sunitinib , Xenograft Model Antitumor Assays
9.
Mol Endocrinol ; 17(4): 562-74, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12554762

ABSTRACT

The preovulatory surge of estrogen up-regulates estrogen receptor-alpha (ER) gene expression in the uterus during the estrous/menstrual cycles of female mammals. Previously, we demonstrated that the 5-fold increase in ER mRNA levels in endometrium of ovariectomized ewes treated with a physiological dose of estradiol (E2) is entirely due to an increase in ER mRNA stability. Our current work confirms that the E2 effect is specific to ER mRNA. The sequence of ER mRNA, cloned from sheep endometrium, shows a high degree of conservation with those of other species, even in the 5'- and the very long 3'-untranslated regions. In a cell-free assay, ER mRNA demonstrates greater stability with endometrial extracts from E2-treated ewes compared with those from untreated ovariectomized ewes. The E2-enhanced stability of ER mRNA was ablated by prior treatment of the extracts with proteinase K, 70 C heat, and oxidizing and alkylating reagents, indicating that a protein is responsible for stabilization of the message. The 3'-untranslated region of ER mRNA contains discrete sequences required for E2-enhanced stability, four of which were identified by extensive deletion mutant analyses. Transfer of two of the four minimal E2-modulated stability sequences conferred E2-enhanced stability to a heterologous RNA. These minimal E2-modulated stability sequences contain a common 10-base, uridine-rich sequence that is predicted to reside in a loop structure. Throughout our studies, estrogen stabilization of ER mRNA in sheep endometrium resembled that of vitellogenin mRNA in frog liver, indicating conservation of this ancient mechanism for enhancing gene expression in response to estrogen.


Subject(s)
3' Untranslated Regions , Carrier Proteins , Endometrium/physiology , Estradiol/metabolism , RNA Stability , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Animals , Base Sequence , Conserved Sequence , Estradiol/pharmacology , Female , Magnesium/metabolism , Magnesium/pharmacology , Molecular Sequence Data , Polyadenylation , Proteins/metabolism , RNA, Messenger/drug effects , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Estrogen/drug effects , Regulatory Sequences, Ribonucleic Acid , Sheep , Up-Regulation , Uterus/physiology
10.
Cancer Genomics Proteomics ; 10(6): 251-63, 2013.
Article in English | MEDLINE | ID: mdl-24336634

ABSTRACT

BACKGROUND: Therapeutic targeting of Rho-Associated, Coiled-Coil Containing Protein Kinase (ROCK) signaling for tumor cells and tumor endothelium has shown efficacy in pre-clinical tumors models, and a better understanding of how proteins regulate tumor progression will strengthen our knowledge over disease etiology and treatment of patients with cancer. Recent reports have shown that ROCK activity is critical for the expression of a large number of mRNA transcripts across multiple cell types including endothelial cells. MATERIALS AND METHODS: To examine the effects of ROCK proteins on microRNA (miRNA) expression in tumor-forming endothelial cells, we utilized microarrays to evaluate expression levels of 1088 miRNAs in vascular tumor-forming endothelial cells knocked-down for ROCK1 or ROCK2 or treated with a pharmacological inhibitor of ROCK activity. RESULTS: Microarray analysis demonstrated that inhibiting ROCK activity altered global miRNA expression. We confirmed our findings using qPCR and identified cell-cycle progression, calcium transport, and neurogenesis/synaptogenesis as the most highly overrepresented predicted target gene networks for the identified miRNAs whose expression was altered by ROCK inhibition. CONCLUSION: ROCK signaling induces large-scale changes in global miRNA expression and may lead to a better understanding of how these proteins affect aberrant vascular states.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/biosynthesis , rho-Associated Kinases/metabolism , Animals , Cell Line , Endothelial Cells/enzymology , Gene Knockdown Techniques , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Transfection , rho-Associated Kinases/deficiency , rho-Associated Kinases/genetics
11.
Vasc Cell ; 5(1): 6, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23531100

ABSTRACT

BACKGROUND: Infantile hemangiomas are benign vascular tumors primarily found on the skin in 10% of the pediatric population. The etiology of this disease is largely unknown and while large scale genomic studies have examined the transcriptomes of infantile hemangioma tumors as a whole, no study to date has compared the global gene expression profiles of pure infantile hemangioma endothelial cells (HEMECs) to that of normal human dermal microvascular endothelial cells (HDMVECs). METHODS: To shed light on the molecular differences between these normal and aberrant dermal endothelial cell types, we performed whole genome microarray analysis on purified cultures of HEMECs and HDMVECs. We then utilized qPCR and immunohistochemistry to confirm our microarray results. RESULTS: Our array analysis identified 125 genes whose expression was upregulated and 104 genes whose expression was downregulated by greater than two fold in HEMECs compared to HDMVECs. Bioinformatics analysis revealed three major classifications of gene functions that were altered in HEMECs including cell adhesion, cell cycle, and arachidonic acid production. Several of these genes have been reported to be critical regulators and/or mutated in cancer, vascular tumors, and vascular malformations. We confirmed the expression of a subset of these differentially expressed genes (ANGPT2, ANTXR1, SMARCE1, RGS5, CTAG2, LTBP2, CLDN11, and KISS1) using qPCR and utilized immunohistochemistry on a panel of paraffin embedded infantile hemangioma tumor tissues to demonstrate that the cancer/testis antigen CTAG2 is highly abundant in vessel-dense proliferating infantile hemangiomas and with significantly reduced levels during tumor involution as vascular density decreases. CONCLUSION: Our data reveal that the transcriptome of HEMECs is reflective of a pro-proliferative cell type with altered adhesive characteristics. Moveover, HEMECs show altered expression of many genes that are important in the progression and prognosis of metastatic cancers.

12.
FEBS J ; 280(18): 4474-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23802622

ABSTRACT

Alterations in cell shape have been shown to modulate chromatin condensation and cell lineage specification; however, the mechanisms controlling these processes are largely unknown. Because endothelial cells experience cyclic mechanical changes from blood flow during normal physiological processes and disrupted mechanical changes as a result of abnormal blood flow, cell shape deformation and loss of polarization during coronary artery disease, we aimed to determine how morphological restriction affects global gene expression patterns. Human coronary artery endothelial cells (HCAECs) were cultured on spatially defined adhesive micropatterns, forcing them to conform to unique cellular morphologies differing in cellular polarization and angularity. We utilized pattern recognition algorithms and statistical analysis to validate the cytoskeletal pattern reproducibility and uniqueness of each micropattern, and performed microarray analysis on normal-shaped and micropatterned HCAECs to determine how constrained cellular morphology affects gene expression patterns. Analysis of the data revealed that forcing HCAECs to conform to geometrically-defined shapes significantly affects their global transcription patterns compared to nonrestricted shapes. Interestingly, gene expression patterns were altered in response to morphological restriction in general, although they were consistent regardless of the particular shape the cells conformed to. These data suggest that the ability of HCAECs to spread, although not necessarily their particular morphology, dictates their genomics patterns.


Subject(s)
Actin Cytoskeleton/genetics , Coronary Vessels/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Microfilament Proteins/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Algorithms , Cell Adhesion , Cell Polarity/genetics , Cell Shape/genetics , Coronary Vessels/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression Profiling , Humans , Image Processing, Computer-Assisted , Mechanotransduction, Cellular , Microfilament Proteins/metabolism , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Primary Cell Culture
13.
PLoS One ; 8(3): e60021, 2013.
Article in English | MEDLINE | ID: mdl-23555867

ABSTRACT

Therapeutic targeting of the beta-adrenergic receptors has recently shown remarkable efficacy in the treatment of benign vascular tumors such as infantile hemangiomas. As infantile hemangiomas are reported to express high levels of beta adrenergic receptors, we examined the expression of these receptors on more aggressive vascular tumors such as hemangioendotheliomas and angiosarcomas, revealing beta 1, 2, and 3 receptors were indeed present and therefore aggressive vascular tumors may similarly show increased susceptibility to the inhibitory effects of beta blockade. Using a panel of hemangioendothelioma and angiosarcoma cell lines, we demonstrate that beta adrenergic inhibition blocks cell proliferation and induces apoptosis in a dose dependent manner. Beta blockade is selective for vascular tumor cells over normal endothelial cells and synergistically effective when combined with standard chemotherapeutic or cytotoxic agents. We demonstrate that inhibition of beta adrenergic signaling induces large scale changes in the global gene expression patterns of vascular tumors, including alterations in the expression of established cell cycle and apoptotic regulators. Using in vivo tumor models we demonstrate that beta blockade shows remarkable efficacy as a single agent in reducing the growth of angiosarcoma tumors. In summary, these experiments demonstrate the selective cytotoxicity and tumor suppressive ability of beta adrenergic inhibition on malignant vascular tumors and have laid the groundwork for a promising treatment of angiosarcomas in humans.


Subject(s)
Hemangioendothelioma/drug therapy , Hemangioendothelioma/metabolism , Hemangiosarcoma/drug therapy , Propranolol/therapeutic use , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytoskeleton/metabolism , Female , Fluorescent Antibody Technique , Hemangiosarcoma/metabolism , Humans , Immunohistochemistry , Mice , Propranolol/pharmacology
14.
Exp Ther Med ; 4(4): 594-604, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23170111

ABSTRACT

Infantile hemangiomas (IHs) are non-malignant, largely cutaneous vascular tumors affecting approximately 5-10% of children to varying degrees. During the first year of life, these tumors are strongly proliferative, reaching an average size ranging from 2 to 20 cm. These lesions subsequently stabilize, undergo a spontaneous slow involution and are fully regressed by 5 to 10 years of age. Systemic treatment of infants with the non-selective ß-adrenergic receptor blocker, propranolol, has demonstrated remarkable efficacy in reducing the size and appearance of IHs. However, the mechanism by which this occurs is largely unknown. In this study, we sought to understand the molecular mechanisms underlying the effectiveness of ß blocker treatment in IHs. Our data reveal that propranolol treatment of IH endothelial cells, as well as a panel of normal primary endothelial cells, blocks endothelial cell proliferation, migration, and formation of the actin cytoskeleton coincident with alterations in vascular endothelial growth factor receptor-2 (VEGFR-2), p38 and cofilin signaling. Moreover, propranolol induces major alterations in the protein levels of key cyclins and cyclin-dependent kinase inhibitors, and modulates global gene expression patterns with a particular affect on genes involved in lipid/sterol metabolism, cell cycle regulation, angiogenesis and ubiquitination. Interestingly, the effects of propranolol were endothelial cell-type independent, affecting the properties of IH endothelial cells at similar levels to that observed in neonatal dermal microvascular and coronary artery endothelial cells. This data suggests that while propranolol markedly inhibits hemangioma and normal endothelial cell function, its lack of endothelial cell specificity hints that the efficacy of this drug in the treatment of IHs may be more complex than simply blockage of endothelial function as previously believed.

15.
Mol Biol Cell ; 19(3): 994-1006, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094043

ABSTRACT

Angiogenesis is largely controlled by hypoxia-driven transcriptional up-regulation and secretion of vascular endothelial growth factor (VEGF) and its binding to the endothelial cell tyrosine receptor kinases, VEGFR1 and VEGFR2. Recent expression analysis suggests that VEGF is expressed in a cell-specific manner in normoxic adult tissue; however, the transcriptional regulation and role of VEGF in these tissues remains fundamentally unknown. In this report we demonstrate that VEGF is coordinately up-regulated during terminal skeletal muscle differentiation. We reveal that this regulation is mediated in part by MyoD homo- and hetero-dimeric transcriptional mechanisms. Serial deletions of the VEGF promoter elucidated a region containing three tandem CANNTG consensus MyoD sites serving as essential sites of direct interaction for MyoD-mediated up-regulation of VEGF transcription. VEGF-null embryonic stem (ES) cells exhibited reduced myogenic differentiation compared with wild-type ES cells, suggesting that VEGF may serve a role in skeletal muscle differentiation. We demonstrate that VEGFR1 and VEGFR2 are expressed at low levels in myogenic precursor cells and are robustly activated upon VEGF stimulation and that their expression is coordinately regulated during skeletal muscle differentiation. VEGF stimulation of differentiating C2C12 cells promoted myotube hypertrophy and increased myogenic differentiation, whereas addition of sFlt1, a VEGF inhibitor, resulted in myotube hypotrophy and inhibited myogenic differentiation. We further provide evidence indicating VEGF-mediated myogenic marker expression, mitogenic activity, migration, and prosurvival functions may contribute to increased myogenesis. These data suggest a novel mechanism whereby VEGF is coordinately regulated as part of the myogenic differentiation program and serves an autocrine function regulating skeletal myogenesis.


Subject(s)
Muscle Development , Muscle, Skeletal/embryology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Mice, Inbred C57BL , Muscle Development/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , MyoD Protein/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology
16.
J Biol Chem ; 281(1): 51-8, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16260418

ABSTRACT

KiSS-1 has been shown to function as a tumor metastasis suppressor gene and reduce the number of metastases in different cancers. The expression of KiSS-1 or KiSS1, like other tumor suppressor, is commonly reduced or completely ablated in a variety of cancers via an unknown mechanism. Here we show that the loss of KiSS-1 expression in highly metastatic breast cancer cell lines correlates directly with the expression levels of two transcription factors, activator protein-2alpha (AP-2alpha) and specificity protein 1 (Sp1), which synergistically activate the transcriptional regulation of KiSS-1 in breast cancer cells. Although the KiSS-1 promoter contains multiple AP-2alpha binding elements, AP-2alpha-mediated regulation occurs indirectly through Sp1 sites, as determined by deletion and mutation analysis. Overexpression of AP-2alpha into highly metastatic breast cell lines did not alter KiSS-1 promoter-driven luciferase gene activity. However, co-transfection of AP-2alpha wild-type or the dominant negative form of AP-2 lacking its C-terminal DNA-binding domain, AP-2B, together with Sp1, increased KiSS-1 promoter activity dramatically, suggesting that AP-2alpha regulation of KiSS-1 transcription does not require direct binding to the KiSS-1 promoter. Furthermore, we demonstrated that AP-2alpha directly interacted with Sp1 to form transcription complexes at two tandem Sp1-binding sites of the promoter to activate KiSS-1 transcription. Together, our results indicate that AP-2alpha and Sp1 are strong transcriptional regulators of KiSS-1 and that loss or decreased expression of AP-2alpha in breast cancer may account for the loss of tumor metastasis suppressor KiSS-1 expression and thus increased cancer metastasis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/secondary , Proteins/genetics , Sp1 Transcription Factor/metabolism , Transcription Factor AP-2/metabolism , Breast Neoplasms/physiopathology , Cell Line, Tumor , Consensus Sequence , Gene Expression Regulation, Neoplastic/physiology , Humans , Kisspeptins , Promoter Regions, Genetic/physiology , Transcriptional Activation/physiology , Tumor Suppressor Proteins
17.
Funct Integr Genomics ; 3(3): 112-6, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12687409

ABSTRACT

We are interested in the collagen gene superfamily and its involvement in hereditary diseases of the human and domestic dog. Presented here is radiation hybrid mapping of the type I and type IV collagen gene subfamilies on the most recent version of the canine map. The col1A1 gene was mapped to chromosome 9, col1A2 was mapped to chromosome 14, col4A1 and col4A2 were mapped to chromosome 22 and col4A3 and col4A4 were mapped to chromosome 25. The col4A5 and col4A6 genes, while linked to one another, are not linked in the present version of the canine map but likely are present on the X chromosome. These data provide an insight into the molecular evolution of these subfamilies and increase the number of mapped genes in discrete regions of the canine genome.


Subject(s)
Collagen Type IV/genetics , Collagen Type I/genetics , Dogs/genetics , Radiation Hybrid Mapping , Animals , Genetic Linkage , Humans , Mice , Molecular Sequence Data , Multigene Family , X Chromosome
SELECTION OF CITATIONS
SEARCH DETAIL