Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Publication year range
1.
Cell ; 149(6): 1257-68, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682248

ABSTRACT

Thrombospondin (Thbs) proteins are induced in sites of tissue damage or active remodeling. The endoplasmic reticulum (ER) stress response is also prominently induced with disease where it regulates protein production and resolution of misfolded proteins. Here we describe a function for Thbs as ER-resident effectors of an adaptive ER stress response. Thbs4 cardiac-specific transgenic mice were protected from myocardial injury, whereas Thbs4(-/-) mice were sensitized to cardiac maladaptation. Thbs induction produced a unique profile of adaptive ER stress response factors and expansion of the ER and downstream vesicles. Thbs bind the ER lumenal domain of activating transcription factor 6α (Atf6α) to promote its nuclear shuttling. Thbs4(-/-) mice showed blunted activation of Atf6α and other ER stress-response factors with injury, and Thbs4-mediated protection was lost upon Atf6α deletion. Hence, Thbs can function inside the cell during disease remodeling to augment ER function and protect through a mechanism involving regulation of Atf6α.


Subject(s)
Endoplasmic Reticulum Stress , Signal Transduction , Thrombospondins/metabolism , Activating Transcription Factor 6/genetics , Animals , Cardiomyopathies/metabolism , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic , Thrombospondins/genetics
2.
Semin Cell Dev Biol ; 155(Pt B): 3-11, 2024 03 01.
Article in English | MEDLINE | ID: mdl-37286406

ABSTRACT

Many cancers begin with the formation of a small nest of transformed cells that can remain dormant for years. Thrombospondin-1 (TSP-1) initially promotes dormancy by suppressing angiogenesis, a key early step in tumor progression. Over time, increases in drivers of angiogenesis predominate, and vascular cells, immune cells, and fibroblasts are recruited to the tumor mass forming a complex tissue, designated the tumor microenvironment. Numerous factors, including growth factors, chemokine/cytokine, and extracellular matrix, participate in the desmoplastic response that in many ways mimics wound healing. Vascular and lymphatic endothelial cells, and cancer-associated pericytes, fibroblasts, macrophages and immune cells are recruited to the tumor microenvironment, where multiple members of the TSP gene family promote their proliferation, migration and invasion. The TSPs also affect the immune signature of tumor tissue and the phenotype of tumor-associated macrophages. Consistent with these observations, expression of some TSPs has been established to correlate with poor outcomes in specific types of cancer.


Subject(s)
Neoplasms , Thrombospondins , Humans , Thrombospondins/genetics , Thrombospondins/metabolism , Endothelial Cells/metabolism , Tumor Microenvironment , Neoplasms/metabolism , Extracellular Matrix/metabolism
3.
Cell ; 139(2): 380-92, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818485

ABSTRACT

Synapses are asymmetric cellular adhesions that are critical for nervous system development and function, but the mechanisms that induce their formation are not well understood. We have previously identified thrombospondin as an astrocyte-secreted protein that promotes central nervous system (CNS) synaptogenesis. Here, we identify the neuronal thrombospondin receptor involved in CNS synapse formation as alpha2delta-1, the receptor for the anti-epileptic and analgesic drug gabapentin. We show that the VWF-A domain of alpha2delta-1 interacts with the epidermal growth factor-like repeats common to all thrombospondins. alpha2delta-1 overexpression increases synaptogenesis in vitro and in vivo and is required postsynaptically for thrombospondin- and astrocyte-induced synapse formation in vitro. Gabapentin antagonizes thrombospondin binding to alpha2delta-1 and powerfully inhibits excitatory synapse formation in vitro and in vivo. These findings identify alpha2delta-1 as a receptor involved in excitatory synapse formation and suggest that gabapentin may function therapeutically by blocking new synapse formation.


Subject(s)
CD36 Antigens/metabolism , Calcium Channels/metabolism , Neurogenesis , Synapses , Amines/pharmacology , Animals , Calcium Channels, L-Type , Cyclohexanecarboxylic Acids/pharmacology , Gabapentin , Mice , Neuronal Plasticity , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Synapses/drug effects , gamma-Aminobutyric Acid/pharmacology
4.
Semin Cancer Biol ; 86(Pt 2): 126-135, 2022 11.
Article in English | MEDLINE | ID: mdl-36191900

ABSTRACT

Considerable progress has been made in our understanding of the process of angiogenesis in the context of normal and tumor tissue over the last fifty years. Angiogenesis, like most physiological processes, is carefully controlled by dynamic and opposing effects of positive factors, such as vascular endothelial growth factor (VEGF), and negative factors, such as thrombospondin-1. In most cases, the progression of a small mass of cancerous cells to a life-threatening tumor depends upon the initiation of angiogenesis and involves the dysregulation of the angiogenic balance. Whereas our newfound appreciation for the role of angiogenesis in cancer has opened up new avenues for treatment, the success of these treatments, which have focused almost exclusively on antagonizing the VEGF pathway, has been limited to date. It is anticipated that this situation will improve as more therapeutics that target other pathways are developed, more strategies for combination therapies are advanced, more detailed stratification of patient populations occurs, and a better understanding of resistance to anti-angiogenic therapy is gained.


Subject(s)
Neoplasms , Neovascularization, Pathologic , Thrombospondin 1 , Vascular Endothelial Growth Factor A , Humans , Neoplasms/blood supply , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Thrombospondin 1/metabolism
5.
Gynecol Oncol ; 164(1): 154-169, 2022 01.
Article in English | MEDLINE | ID: mdl-34799137

ABSTRACT

OBJECTIVES: Tumor vasculature is structurally abnormal, with anatomical deformities, reduced pericyte coverage and low tissue perfusion. As a result of this vascular dysfunction, tumors are often hypoxic, which is associated with an aggressive tumor phenotype, and reduced delivery of therapeutic compounds to the tumor. We have previously shown that a peptide containing the thrombospondin-1 type I repeats (3TSR) specifically targets tumor vessels and induces vascular normalization in a mouse model of epithelial ovarian cancer (EOC). However, due to its small size, 3TSR is rapidly cleared from circulation. We now introduce a novel construct with the 3TSR peptide fused to the C-terminus of each of the two heavy chains of the Fc region of human IgG1 (Fc3TSR). We hypothesize that Fc3TSR will have greater anti-tumor activity in vitro and in vivo compared to the native compound. METHODS: Fc3TSR was evaluated in vitro using proliferation and apoptosis assays to investigate differences in efficacy compared to native 3TSR. In light of the multivalency of Fc3TSR, we also investigate whether it induces greater clustering of its functional receptor, CD36. We also compare the compounds in vivo using an orthotopic, syngeneic mouse model of advanced stage EOC. The impact of the two compounds on changes to tumor vasculature morphology was also investigated. RESULTS: Fc3TSR significantly decreased the viability and proliferative potential of EOC cells and endothelial cells in vitro compared to native 3TSR. High-resolution imaging followed by image correlation spectroscopy demonstrated enhanced clustering of the CD36 receptor in cells treated with Fc3TSR. This was associated with enhanced downstream signaling and greater in vitro and in vivo cellular responses. Fc3TSR induced greater vascular normalization and disease regression compared to native 3TSR in an orthotopic, syngeneic mouse model of advanced stage ovarian cancer. CONCLUSION: The development of Fc3TSR which is greater in size, stable in circulation and enhances receptor activation compared to 3TSR, facilitates its translational potential as a therapy in the treatment of metastatic advanced stage ovarian cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Immunoglobulin G/therapeutic use , Ovarian Neoplasms/drug therapy , Thrombospondin 1/therapeutic use , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor/drug effects , Disease Models, Animal , Female , Humans , Immunoglobulin G/pharmacology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Ovarian Neoplasms/pathology , Thrombospondin 1/pharmacokinetics , Thrombospondin 1/pharmacology
8.
J Surg Res ; 210: 1-7, 2017 04.
Article in English | MEDLINE | ID: mdl-28457315

ABSTRACT

BACKGROUND: Thrombospondin-1 (TSP-1) is functionally important to intimal hyperplasia (IH) development. Statin drugs have beneficial pleiotropic effects, including reduced IH; however, the effect of statins on IH in a TSP-1-independent setting is unknown. HYPOTHESIS: Statins will be less effective in attenuating IH after vascular injury in TSP-1-null (Thbs1-/-) mice compared with wild-type (WT) mice. MATERIALS AND METHODS: Carotid artery ligation was performed on WT and Thbs1-/- mice. Each strain was divided into two groups: no statin control or standard chow containing fluvastatin (10 or 40 mg/kg/d). After 28 d, analysis included morphometric analysis and real-time quantitative reverse transcription polymerase chain reaction on the arteries and enzyme-linked immunosorbent assay on plasma (TSP-1 WT, TSP-2 WT, and Thbs1-/-). Comparisons were made by analysis of variance, with P < 0.05 considered significant. RESULTS: In no statin controls, WT mice had more IH than Thbs1-/- mice (0.46 ± 0.09 versus 0.15 ± 0.04). Fluvastatin reduced IH in the WT (0.46 ± 0.09 versus 0.23 ± 0.06), but not in Thbs1-/- groups (0.15 ± 0.04 versus 0.22 ± 0.07). No difference in IH existed between Thbs1-/- no statin controls and fluvastatin WT and Thbs1-/- groups. Statin dose did not affect IH. TSP-1 plasma levels were increased in fluvastatin WT. TSP-2 levels were decreased in fluvastatin WT and elevated in fluvastatin Thbs1-/-. Fluvastatin had no effect on tissue Thbs1 or Thbs2 gene expression. CONCLUSIONS: TSP-1 is necessary for robust IH after arterial injury. Because fluvastatin had no effect on IH in Thbs1-/-, the data suggest that the statin effect on IH may be largely TSP-1 dependent. Both statins and the presence of TSP-1 affect TSP-1 and TSP-2 plasma levels.


Subject(s)
Carotid Arteries/pathology , Fatty Acids, Monounsaturated/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperplasia/prevention & control , Indoles/therapeutic use , Thrombospondin 1/metabolism , Tunica Intima/pathology , Animals , Biomarkers/metabolism , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hyperplasia/metabolism , Indoles/pharmacology , Male , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/deficiency , Tunica Intima/drug effects , Tunica Intima/metabolism
9.
FASEB J ; 29(2): 576-88, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25395453

ABSTRACT

Most women are diagnosed with epithelial ovarian cancer (EOC) at advanced stage, where therapies have limited effectiveness and the long-term survival rate is low. We evaluated the effects of combined antiangiogenic and chemotherapy treatments on advanced stage EOC. Treatment of EOC cells with a recombinant version of the thrombospondin-1 type I repeats (3TSR) induced more apoptotic cell death (36.5 ± 9.6%) in vitro compared to untreated controls (4.1 ± 1.4). In vivo, tumors were induced in an orthotopic, syngeneic mouse model of advanced stage EOC. Mice were treated with 3TSR (4 mg/kg per day) alone or in combination with chemotherapy drugs delivered with maximum tolerated dose or metronomic scheduling. Pretreatment with 3TSR induced tumor regression, normalized tumor vasculature, and improved uptake of chemotherapy drugs. Combination 3TSR and metronomic chemotherapy induced the greatest tumor regression (6.2-fold reduction in size compared to PBS-treated controls) and highest survival when treatment was initiated at advanced stage. 3TSR binding to its receptor, CD36 (cluster of differentiation 36), increased binding of CD36 and SHP-1, which significantly inhibited phosphorylation of the VEGF receptor. In this study, we describe a novel treatment approach and mechanism of action with 3TSR and chemotherapy that induces regression of advanced stage EOC and significantly improves survival.-Russell, S., Duquette, M., Liu, J., Drapkin, R., Lawler, J., Petrik, J. Combined therapy with thrombospondin-1 type I repeats (3TSR) and chemotherapy induces regression and significantly improves survival in a preclinical model of advanced stage epithelial ovarian cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Thrombospondin 1/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis , CD36 Antigens/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Combined Modality Therapy , Female , Humans , Hypoxia , Maximum Tolerated Dose , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Thrombospondin 1/genetics , Treatment Outcome
10.
Mol Ther ; 23(2): 235-43, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25358253

ABSTRACT

Three type-1 repeat (3TSR) domain of thrombospondin-1 is known to have anti-angiogenic effects by targeting tumor-associated endothelial cells, but its effect on tumor cells is unknown. This study explored the potential of 3TSR to target glioblastoma (GBM) cells in vitro and in vivo. We show that 3TSR upregulates death receptor (DR) 4/5 expression in a CD36-dependent manner and primes resistant GBMs to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced caspase-8/3/7 mediated apoptosis. We engineered human mesenchymal stem cells (MSC) for on-site delivery of 3TSR and a potent and secretable variant of TRAIL (S-TRAIL) in an effort to simultaneously target tumor cells and associated endothelial cells and circumvent issues of systemic delivery of drugs across the blood-brain barrier. We show that MSC-3TSR/S-TRAIL inhibits tumor growth in an expanded spectrum of GBMs. In vivo, a single administration of MSC-3TSR/S-TRAIL significantly targets both tumor cells and vascular component of GBMs, inhibits tumor progression, and extends survival of mice bearing highly vascularized GBM. The ability of 3TSR/S-TRAIL to simultaneously act on tumor cells and tumor-associated endothelial cells offers a great potential to target a broad spectrum of cancers and translate 3TSR/TRAIL therapies into clinics.


Subject(s)
Glioblastoma/genetics , Glioblastoma/pathology , Neovascularization, Pathologic/genetics , Protein Interaction Domains and Motifs/genetics , Thrombospondin 1/genetics , Animals , Apoptosis , CD36 Antigens/metabolism , Caspases/metabolism , Cell Line, Tumor , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Lentivirus/genetics , Mesenchymal Stem Cells/metabolism , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thrombospondin 1/chemistry , Transduction, Genetic
11.
Blood ; 121(11): 2154-64, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23315169

ABSTRACT

Angiogenesis plays an important role in cancer and in many other human diseases. Vascular endothelial growth factor-A (VEGF-A), the best known angiogenic factor, was originally discovered as a potent vascular permeability factor (VPF), suggesting that other vascular permeabilizing agents, such as histamine and serotonin, might also have angiogenic activity. We recently demonstrated that, like VEGF-A, histamine and serotonin up-regulate the orphan nuclear receptor and transcription factor TR3 (mouse homolog Nur77) and that TR3/Nur77 is essential for their vascular permeabilizing activities. We now report that histamine and serotonin are also angiogenic factors that, at low micromolar concentrations, induce endothelial cell proliferation, migration and tube formation in vitro, and angiogenesis in vivo. All of these responses are mediated through specific histamine and serotonin receptors, are independent of VEGF-A, and are directly dependent on TR3/Nur77. Initially, the angiogenic response closely resembled that induced by VEGF-A, with generation of "mother" vessels. However, after ~10 days, mother vessels began to regress as histamine and serotonin, unlike VEGF-A, up-regulated the potent angiogenesis inhibitor thrombospondin-1, thereby triggering a negative feedback loop. Thus, histamine and serotonin induce an angiogenic response that fits the time scale of acute inflammation.


Subject(s)
Histamine/pharmacology , Neovascularization, Physiologic/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Serotonin/pharmacology , Thrombospondin 1/physiology , Animals , Capillary Permeability/drug effects , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neovascularization, Physiologic/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Thrombospondin 1/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
12.
Proc Natl Acad Sci U S A ; 108(24): 9939-44, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21622854

ABSTRACT

Whereas the roles of proangiogenic factors in carcinogenesis are well established, those of endogenous angiogenesis inhibitors (EAIs) remain to be fully elaborated. We investigated the roles of three EAIs during de novo tumorigenesis to further test the angiogenic balance hypothesis, which suggests that blood vessel development in the tumor microenvironment can be governed by a net loss of negative regulators of angiogenesis in addition to the well-established principle of up-regulated angiogenesis inducers. In a mouse model of pancreatic neuroendocrine cancer, administration of endostatin, thrombospondin-1, and tumstatin peptides, as well as deletion of their genes, reveal neoplastic stage-specific effects on angiogenesis, tumor progression, and survival, correlating with endothelial expression of their receptors. Deletion of tumstatin and thrombospondin-1 in mice lacking the p53 tumor suppressor gene leads to increased incidence and reduced latency of angiogenic lymphomas associated with diminished overall survival. The results demonstrate that EAIs are part of a balance mechanism regulating tumor angiogenesis, serving as intrinsic microenvironmental barriers to tumorigenesis.


Subject(s)
Autoantigens/metabolism , Collagen Type IV/metabolism , Endostatins/metabolism , Neoplasms/metabolism , Thrombospondin 1/metabolism , Amino Acid Sequence , Animals , Autoantigens/chemistry , Autoantigens/genetics , Cell Line , Collagen Type IV/chemistry , Collagen Type IV/genetics , Disease Progression , Endostatins/chemistry , Endostatins/genetics , Female , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Neoplasm Staging , Neoplasms/genetics , Neoplasms/prevention & control , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/prevention & control , Peptides/pharmacology , Propionates/pharmacology , Survival Analysis , Thrombospondin 1/chemistry , Thrombospondin 1/genetics , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
13.
Blood ; 128(12): 1543-4, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27658700

Subject(s)
Thrombosis , Humans
14.
Blood ; 117(17): 4658-66, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21378271

ABSTRACT

CD36 plays a critical role in the inhibition of angiogenesis through binding to the type 1 repeats of thrombospondin-1 (TSP-1) and activating Fyn tyrosine kinase and MAPK pathways. Here, we reveal a novel association of CD36 with VEGFR-2 and spleen tyrosine kinase (Syk). We also address the correlation between the expression of CD36 and Syk by demonstrating that overexpression of CD36 in HUVECs up-regulates endogenous Syk expression. We also define a new role for TSP-1 and CD36 in the activation of the VEGFR-2 signaling pathway that requires Syk. Our findings also identify a role for Syk as a stimulator of VEGF-A-induced angiogenesis by increasing phosphorylation of Y1175 in VEGFR-2, which is a major tyrosine for promoting VEGF-A-induced endothelial cell migration. Together, these studies introduce a new signaling pathway for TSP-1, CD36, and Syk, and address the role of these proteins in regulating the angiogenic switch.


Subject(s)
CD36 Antigens/metabolism , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Thrombospondin 1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Humans , Neovascularization, Physiologic/physiology , Phosphorylation/physiology , Syk Kinase , Umbilical Veins/cytology , Up-Regulation/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Cancer Cell ; 7(1): 101-11, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15652753

ABSTRACT

Development of antiangiogenic therapies would be significantly facilitated by quantitative surrogate pharmacodynamic markers. Circulating peripheral blood endothelial cells (CECs) and/or their putative progenitor subset (CEPs) have been proposed but not yet fully validated for this purpose. Herein, we provide such validation by showing a striking correlation between highly genetically heterogeneous bFGF- or VEGF-induced angiogenesis and intrinsic CEC or CEP levels measured by flow cytometry, among eight different inbred mouse strains. Moreover, studies using genetically altered mice showed that levels of these cells are affected by regulators of angiogenesis, including VEGF, Tie-2, and thrombospondin-1. Finally, treatment with a targeted VEGFR-2 antibody caused a dose-dependent reduction in viable CEPs that precisely paralleled its previously and empirically determined antitumor activity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Biomarkers , Endothelial Cells/physiology , Neovascularization, Pathologic , Stem Cells/physiology , Angiogenesis Inhibitors/metabolism , Animals , Biological Assay/methods , Cell Survival , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Flow Cytometry , Humans , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Phenotype , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Stem Cells/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Proc Natl Acad Sci U S A ; 107(23): 10649-54, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20498063

ABSTRACT

Although B-Raf(V600E) is the most common somatic mutation in papillary thyroid carcinoma (PTC), how it induces tumor aggressiveness is not fully understood. Using gene set enrichment analysis and in vitro and in vivo functional studies, we identified and validated a B-Raf(V600E) gene set signature associated with tumor progression in PTCs. An independent cohort of B-Raf(V600E)-positive PTCs showed significantly higher expression levels of many extracellular matrix genes compared with controls. We performed extensive in vitro and in vivo validations on thrombospondin-1 (TSP-1), because it has been previously shown to be important in the regulation of tumor angiogenesis and metastasis and is present in abundance in tumor stroma. Knockdown of B-Raf(V600E) resulted in TSP-1 down-regulation and a reduction of adhesion and migration/invasion of human thyroid cancer cells. Knockdown of TSP-1 resulted in a similar phenotype. B-Raf(V600E) cells in which either B-Raf(V600E) or TSP-1 were knocked down were implanted orthotopically into the thyroids of immunocompromised mice, resulting in significant reduction in tumor size and fewer pulmonary metastases from the primary carcinoma as compared with the control cells. Treatment of orthotopic thyroid tumors, initiated 1 week after tumor cell implantation with PLX4720, an orally available selective inhibitor of B-Raf(V600E), caused a significant tumor growth delay and decreased distant metastases, without evidence of toxicity. In conclusion, B-Raf(V600E) plays an important role in PTC progression through genes (i.e., TSP-1) important in tumor invasion and metastasis. Testing of a patient's thyroid cancer for B-Raf(V600E) will yield important information about potential tumor aggressiveness and also allow for future use of targeted therapies with selective B-Raf(V600E) inhibitors, such as PLX4720.


Subject(s)
Disease Progression , Mutation , Proto-Oncogene Proteins B-raf/metabolism , Thrombospondin 1/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Indoles/therapeutic use , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, SCID , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , Signal Transduction , Sulfonamides/therapeutic use , Thrombospondin 1/genetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics
17.
Thyroid ; 33(7): 835-848, 2023 07.
Article in English | MEDLINE | ID: mdl-37171127

ABSTRACT

Background: Anaplastic thyroid carcinoma (ATC) is a rapidly fatal cancer with a median survival of a few months. Enhanced therapeutic options for durable management of ATC will rely on an understanding of genetics and the role of the tumor microenvironment. The prognosis for patients with ATC has not improved despite more detailed scrutiny of underlying tumor genetics. Pericytes in the microenvironment play a key evasive role for thyroid carcinoma (TC) cells. Lenvatinib improves outcomes in patients with radioiodine-refractory well-differentiated TC. In addition to the unclear role of pericytes in ATC, the effect and mechanism of lenvatinib efficacy on ATC have not been sufficiently elucidated. Design: We assessed pericyte enrichment in ATC. We determined the effect of lenvatinib on ATC cell growth cocultured with pericytes and in a xenograft mouse model from human BRAFWT/V600E-ATC-derived cells coimplanted with pericytes. Results: ATC samples were significantly enriched in pericytes compared with normal thyroid samples. BRAFWT/V600E-ATC-derived cells were resistant to lenvatinib treatment shown by a lack of suppression of MAPK and Akt pathways. Moreover, lenvatinib increased CD47 protein (thrombospondin-1 [TSP-1] receptor) levels over time vs. vehicle. TSP-1 levels were downregulated upon lenvatinib at late vs. early time points. Critically, ATC cells, when cocultured with pericytes, showed increased sensitivity to this therapy and ultimately decreased number of cells. The coimplantation in vivo of ATC cells with pericytes increased ATC growth and did not downregulate TSP-1 in the microenvironment in vivo. Conclusions and Implications: Pericytes are enriched in ATC samples. Lenvatinib showed inhibitory effects on BRAFWT/V600E-ATC cells in the presence of pericytes. The presence of pericytes could be crucial for effective lenvatinib treatment in patients with ATC. Degree of pericyte abundance may be an attractive prognostic marker in assessing pharmacotherapeutic options. Effective durable management of ATC will rely on an understanding not only of genetics but also on the role of the tumor microenvironment.


Subject(s)
Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Humans , Animals , Mice , Thyroid Carcinoma, Anaplastic/pathology , Pericytes/metabolism , Pericytes/pathology , Thrombospondin 1/therapeutic use , Tumor Microenvironment , Proto-Oncogene Proteins B-raf , Iodine Radioisotopes/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Disease Models, Animal
18.
Am J Respir Cell Mol Biol ; 44(4): 556-61, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20581099

ABSTRACT

Thrombospondin-1 (TSP-1) is an extracellular protein critical to normal lung homeostasis, and is reported to activate latent transforming growth factor-ß (TGF-ß). Because active TGF-ß is causally involved in lung fibrosis after bleomycin challenge, alterations in TSP-1 may be relevant to pulmonary fibrosis. We sought to determine the effects of TSP-1 deficiency on the susceptibility to bleomycin-induced pulmonary fibrosis in a murine model. Age-matched and sex-matched C57BL/6 wild-type (WT) and TSP-1-deficient mice were treated twice weekly for 4 weeks with intraperitoneal bleomycin (0.035 U/g) or PBS, and were allowed to rest 1 week before being killed. Their lungs were inflated with PBS, fixed in formalin, paraffin-embedded, and sectioned. A certified veterinary pathologist blindly scored each slide for inflammation and fibrosis. Lungs were homogenized to obtain RNA and protein for the real-time RT-PCR analysis of connective tissue growth factor (CTGF) and collagen I, and for Western blotting to detect phospho-Smad2, or total Smad2/3, respectively. In response to bleomycin treatment, measures of fibrosis and inflammation, along with CTGF and collagen I mRNA concentrations, were increased in TSP-1-deficient mice compared with WT mice. Notably, Smad 2/3 signaling was of equal strength in WT and TSP-1 knockout mice treated with bleomycin, suggesting that TSP-1 is not required for the activation of TGF-ß. These results demonstrate that TSP-1 deficiency does not protect mice from systemic bleomycin challenge, and that TSP-1 deficiency is associated with increased expression of lung collagen and CTGF.


Subject(s)
Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/prevention & control , Thrombospondin 1/deficiency , Animals , Bleomycin , Collagen/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Pneumonia/complications , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad2 Protein/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism
19.
Oncologist ; 16(3): 296-309, 2011.
Article in English | MEDLINE | ID: mdl-21355020

ABSTRACT

PURPOSE: B-Raf(V600E) may play a role in the progression from papillary thyroid cancer to anaplastic thyroid cancer (ATC). We tested the effects of a highly selective B-Raf(V600E) inhibitor, PLX4720, on proliferation, migration, and invasion both in human thyroid cancer cell lines (8505c(B-RafV600E) and TPC-1(RET/PTC-1 and wild-type B-Raf)) and in primary human normal thyroid (NT) follicular cells engineered with or without B-Raf(V600E). EXPERIMENTAL DESIGN: Large-scale genotyping analysis by mass spectrometry was performed in order to analyze >900 gene mutations. Cell proliferation and migration/invasion were performed upon PLX4720 treatment in 8505c, TPC-1, and NT cells. Orthotopic implantation of either 8505c or TPC-1 cells into the thyroid of severe combined immunodeficient mice was performed. Gene validations were performed by quantitative polymerase chain reaction and immunohistochemistry. RESULTS: We found that PLX4720 reduced in vitro cell proliferation and migration and invasion of 8505c cells, causing early downregulation of genes involved in tumor progression. PLX4720-treated NT cells overexpressing B-Raf(V600E) (heterozygous wild-type B-Raf/B-Raf(V600E)) showed significantly lower cell proliferation, migration, and invasion. PLX4720 treatment did not block cell invasion in TPC-1 cells with wild-type B-Raf, which showed very low and delayed in vivo tumor growth. In vivo, PLX4720 treatment of 8505c orthotopic thyroid tumors inhibited tumor aggressiveness and significantly upregulated the thyroid differentiation markers thyroid transcription factor 1 and paired box gene 8. CONCLUSIONS: Here, we have shown that PLX4720 preferentially inhibits migration and invasion of B-Raf(V600E) thyroid cancer cells and tumor aggressiveness. Normal thyroid cells were generated to be heterozygous for wild-type B-Raf/B-Raf(V600E), mimicking the condition found in most human thyroid cancers. PLX4720 was effective in reducing cell proliferation, migration, and invasion in this heterozygous model. PLX4720 therapy should be tested and considered for a phase I study for the treatment of patients with B-Raf(V600E) ATC.


Subject(s)
Indoles/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Genotype , Humans , Male , Mass Spectrometry , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/biosynthesis , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Transfection , Xenograft Model Antitumor Assays
20.
BMC Struct Biol ; 11: 22, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21569239

ABSTRACT

BACKGROUND: F-spondin is a multi-domain extracellular matrix (ECM) protein and a contact-repellent molecule that directs axon outgrowth and cell migration during development. The reelin_N domain and the F-spondin domain (FS domain) comprise a proteolytic fragment that interacts with the cell membrane and guides the projection of commissural axons to floor plate. The FS domain is found in F-spondins, mindins, M-spondin and amphiF-spondin. RESULTS: We present the crystal structure of human F-spondin FS domain at 1.95Å resolution. The structure reveals a Ca2+-binding C2 domain variant with an 8-stranded antiparallel ß-sandwich fold. Though the primary sequences of the FS domains of F-spondin and mindin are less than 36% identical, their overall structures are very similar. The unique feature of F-spondin FS domain is the presence of three disulfide bonds associated with the N- and C-termini of the domain and a highly conserved N-linked glycosylation site. The integrin-binding motif found in mindin is not conserved in the F-spondin FS domain. CONCLUSION: The structure of the F-spondin FS domain completes the structural studies of the multiple-domain ECM molecule. The homology of its core structure to a common Ca2+- and lipid-binding C2 domain suggests that the F-spondin FS domain may be responsible for part of the membrane targeting of F-spondin in its regulation of axon development. The structural properties of the FS domain revealed in this study pave the way for further exploration into the functions of F-spondin.


Subject(s)
Calcium/chemistry , Extracellular Matrix Proteins/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Disulfides , Glycosylation , Humans , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Reelin Protein , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL