Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
Development ; 148(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34751773

ABSTRACT

Pericytes reside in capillary beds where they share a basement membrane with endothelial cells and regulate their function. However, little is known about embryonic pericyte development, in part, due to lack of specific molecular markers and genetic tools. Here, we applied single cell RNA-sequencing (scRNA-seq) of platelet derived growth factor beta (pdgfrb)-positive cells to molecularly characterize pericytes in zebrafish larvae. scRNA-seq revealed zebrafish cells expressing mouse pericyte gene orthologs, and comparison with bulk RNA-seq from wild-type and pdgfrb mutant larvae further refined a pericyte gene set. Subsequent integration with mouse pericyte scRNA-seq profiles revealed a core set of conserved pericyte genes. Using transgenic reporter lines, we validated pericyte expression of two genes identified in our analysis: NDUFA4 mitochondrial complex associated like 2a (ndufa4l2a), and potassium voltage-gated channel, Isk-related family, member 4 (kcne4). Both reporter lines exhibited pericyte expression in multiple anatomical locations, and kcne4 was also detected in a subset of vascular smooth muscle cells. Thus, our integrated molecular analysis revealed a molecular profile for zebrafish pericytes and allowed us to develop new tools to observe these cells in vivo.


Subject(s)
Gene Expression Regulation, Developmental , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pericytes/metabolism , Zebrafish Proteins/biosynthesis , Zebrafish/embryology , Animals , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/genetics , Mutation , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
2.
Brain Res Bull ; 175: 99-106, 2021 10.
Article in English | MEDLINE | ID: mdl-34303767

ABSTRACT

BACKGROUND: Cognitive dysfunction is increasingly recognized as an important complication of diabetes mellitus (DM). Accumulating evidence indicates that the abnormality of cerebrovascular structure and function plays an essential role in diabetic cognitive impairment (DCI), however, changes in cerebrovascular factors have been blurred during the development of diabetes. OBJECTIVE: To evaluate the changes in the structure and function of cerebrovascular in DCI mice and to investigate the changes of cerebral angiogenesis and stability factors during the development of DM. METHODS: Diabetes was induced by feeding with high-fat diet combined with intraperitoneal injection of streptozotocin (STZ,120 mg/kg). Cognitive function was evaluated at different stages of DM, cerebral neovascularization, blood-brain barrier (BBB) permeability and hippocampal neurons were measured of DCI mice, and the expression of vascular endothelial growth factor (VEGF) and platelet-derived growth factor receptor ß (PDGFRß) in hippocampus was detected during the development of DM. RESULTS: With the progress of diabetes, the learning and memory ability of mice gradually decreased, and DCI mice showed neuronal degeneration, increased BBB permeability and pathological cerebral neovascularization. Moreover, the expression of VEGF in the hippocampus increased first and then decreased at DM+8week, PDGFRß decreased continuously with the development of diabetes. CONCLUSIONS: Our results demonstrate that DCI may be attributed to the dynamic expression of VEGF/PDGFRß in diabetic hippocampus, and pathological cerebral neovascularization, increased BBB permeability and neuronal degeneration are the key links.


Subject(s)
Brain Chemistry/genetics , Cognition Disorders/genetics , Cognition Disorders/psychology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/psychology , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Blood-Brain Barrier , Diet, High-Fat , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Psychomotor Performance
3.
Exp Eye Res ; 197: 108116, 2020 08.
Article in English | MEDLINE | ID: mdl-32561481

ABSTRACT

Platelet-derived growth factor (PDGF) is associated with clinical proliferative vitreoretinopathy (PVR), which is characterized by formation of sub- or epi-retinal membranes that consist of cells including retinal pigment epithelial (RPE) cells and extracellular matrix. RPE cells play an important role in PVR pathogenesis. Previous findings indicated that PDGF receptor (PDGFR)α was essential in experimental PVR induced by fibroblasts. In RPE cells derived from epiretinal membranes from patients with PVR (RPEMs), Akt was activated by PDGF-B but not PDGF-A, which suggested that PDGFRß was the predominant PDGFR isoform expressed in RPEMs. Indeed, CRISPR/Cas9-mediated depletion of PDGFRß in RPEMs attenuated patient vitreous-induced Akt activation and cellular responses intrinsic to PVR including cell proliferation, migration, and contraction. We conclude that PDGFRß appears to be the PVR relevant PDGFR isoform in RPEMs.


Subject(s)
DNA/genetics , Epiretinal Membrane/genetics , Gene Expression Regulation , Receptor, Platelet-Derived Growth Factor beta/genetics , Retinal Pigment Epithelium/metabolism , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , DNA/metabolism , Epiretinal Membrane/metabolism , Epiretinal Membrane/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Retinal Pigment Epithelium/pathology
4.
J Clin Invest ; 129(11): 4609-4628, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31287804

ABSTRACT

Cancer-associated fibroblasts (CAFs) are key actors in modulating the progression of many solid tumors such as breast cancer (BC). Herein, we identify an integrin α11/PDGFRß+ CAF subset displaying tumor-promoting features in BC. In the preclinical MMTV-PyMT mouse model, integrin α11-deficiency led to a drastic reduction of tumor progression and metastasis. A clear association between integrin α11 and PDGFRß was found at both transcriptional and histological levels in BC specimens. High stromal integrin α11/PDGFRß expression was associated with high grades and poorer clinical outcome in human BC patients. Functional assays using five CAF subpopulations (one murine, four human) revealed that integrin α11 promotes CAF invasion and CAF-induced tumor cell invasion upon PDGF-BB stimulation. Mechanistically, integrin α11 pro-invasive activity relies on its ability to interact with PDGFRß in a ligand-dependent manner and to promote its downstream JNK activation, leading to the production of tenascin C, a pro-invasive matricellular protein. Pharmacological inhibition of PDGFRß and JNK impaired tumor cell invasion induced by integrin α11-positive CAFs. Collectively, our study uncovers an integrin α11-positive subset of pro-tumoral CAFs that exploits PDGFRß/JNK signalling axis to promote tumor invasiveness in BC.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Integrin alpha Chains/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/metabolism , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Female , Humans , Integrin alpha Chains/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics
5.
Hum Gene Ther ; 30(2): 179-196, 2019 02.
Article in English | MEDLINE | ID: mdl-30024280

ABSTRACT

Hepatic stellate cells (HSCs) are the primary cell type responsible for liver fibrogenesis. Transforming growth factor beta 1 (TGF-ß1) and platelet-derived growth factor (PDGF) are key profibrotic cytokines that regulate HSC activation and proliferation with functional convergence. Dual RNA interference against their receptors may achieve therapeutic effects. A novel RNAi strategy based on HSC-specific GFAP promoter-driven and lentiviral-expressed artificial microRNAs (amiRNAs) was devised that consists of an microRNA-30a backbone and effective shRNAs against mouse Pdgfrß and Tgfbr2. Then, its antifibrotic efficacy was tested in primary and cultured HSCs and in mice affected with carbon tetrachloride-induced hepatic fibrosis. The study shows that amiRNA-mediated Pdgfrß and Tgfbr2 co-silencing inhibits HSC activation and proliferation. After recombinant lentiviral particles were delivered into the liver via tail-vein injection, therapeutic amiRNAs were preferentially expressed in HSCs and efficiently co-knocked down in situ Tgfbr2 and Pdgfrß expression, which correlates with downregulated expression of target or effector genes of their signaling, which include Pai-1, P70S6K, and D-cyclins. amiRNA-based HSC-specific co-silencing of Tgfbr2 and Pdgfrß significantly suppressed hepatic expression of fibrotic markers α-Sma and Col1a1, extracellular matrix regulators Mmps and Timp1, and phenotypically ameliorated liver fibrosis, as indicated by reductions in serum alanine aminotransferase activity, collagen deposition, and α-Sma-positive staining. The findings provide proof of concept for the use of amiRNA-mediated co-silencing of two profibrogenic pathways in liver fibrosis treatment and highlight the therapeutic potential of concatenated amiRNAs for gene therapy.


Subject(s)
Carbon Tetrachloride Poisoning , Gene Silencing , Liver Cirrhosis , MicroRNAs , Receptor, Platelet-Derived Growth Factor beta , Receptor, Transforming Growth Factor-beta Type II , Animals , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/therapy , Cell Line , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Transforming Growth Factor-beta Type II/biosynthesis , Receptor, Transforming Growth Factor-beta Type II/genetics
6.
Neuron ; 100(1): 183-200.e8, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30269986

ABSTRACT

Acute infection, if not kept in check, can lead to systemic inflammatory responses in the brain. Here, we show that within 2 hr of systemic inflammation, PDGFRß mural cells of blood vessels rapidly secrete chemokine CCL2, which in turn increases total neuronal excitability by promoting excitatory synaptic transmission in glutamatergic neurons of multiple brain regions. By single-cell RNA sequencing, we identified Col1a1 and Rgs5 subgroups of PDGFRß cells as the main source of CCL2. Lipopolysaccharide (LPS)- or Poly(I:C)-treated pericyte culture medium induced similar effects in a CCL2-dependent manner. Importantly, in Pdgfrb-Cre;Ccl2fl/fl mice, LPS-induced increase in excitatory synaptic transmission was significantly attenuated. These results demonstrate in vivo that PDGFRß cells function as initial sensors of external insults by secreting CCL2, which relays the signal to the central nervous system. Through their gateway position in the brain, PDGFRß cells are ideally positioned to respond rapidly to environmental changes and to coordinate responses.


Subject(s)
Chemokine CCL2/metabolism , Inflammation/metabolism , Neuroimmunomodulation/physiology , Pericytes/metabolism , Animals , Collagen Type I/biosynthesis , Collagen Type I, alpha 1 Chain , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/physiology , Pericytes/cytology , RGS Proteins/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Synaptic Transmission/physiology
7.
PLoS One ; 13(8): e0201395, 2018.
Article in English | MEDLINE | ID: mdl-30125282

ABSTRACT

Various angiogenic factors have been shown to play important roles in intraplaque angiogenesis, while little is known about the dynamic expression change and interplay between various angiogenic factors and intraplaque angiogenesis under high cholesterol conditions. New Zealand rabbits underwent balloon injury of the abdominal artery and then were assigned to a control group (n = 15, normal chow) or high cholesterol group (n = 25, 1% high cholesterol diet). At weeks 4, 6, 8, 10, and 12 after acclimation, rabbits (high cholesterol group, n = 5; control group, n = 3) were euthanized. No lesions were observed in the control group. From week 4 to week 12, the expression of vascular endothelial growth factor A (VEGF-A), VEGF receptor 2 (VEGFR-2), fibroblast growth factor 2 (FGF-2), FGF receptor 1 (FGFR-1), platelet-derived growth factor-BB (PDGF-BB), and tumor necrosis factor alpha (TNF-α), the vulnerability index (VI) and the microvessel density (MVD) were significantly elevated in the high cholesterol group; however, PDGF receptor ß (PDGFR-ß) expression showed little change. Analysis by double-label immunofluorescence (CD31 and Ng2) and FITC-dextran indicated that the neovessels within the plaque were leaky due to a lack of pericytes. As indicated by Pearson's correlation analysis, there was a highly positive correlation between the VI, MVD, macrophage content, and TNF-α level, and the levels of VEGF-A/VEGFR-2 and FGF-2/FGFR-1. However, no correlations were observed between PDGFR-ß levels and the VI or MVD. High expression of VEGF-A/VEGFR-2 and FGF-2/FGFR-1 but not of PDGF-BB/PDGFR-ß may contribute to immature and inflammatory intraplaque angiogenesis and plaque instability in a rabbit model of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Fibroblast Growth Factor 2/biosynthesis , Neovascularization, Pathologic/metabolism , Plaque, Atherosclerotic/metabolism , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Animals , Atherosclerosis/pathology , Becaplermin/biosynthesis , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Male , Neovascularization, Pathologic/pathology , Pericytes/metabolism , Pericytes/pathology , Plaque, Atherosclerotic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rabbits , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
8.
J Cereb Blood Flow Metab ; 38(1): 103-115, 2018 01.
Article in English | MEDLINE | ID: mdl-28151041

ABSTRACT

Recent studies implicate loss of pericytes in hypoperfusion and blood-brain barrier (BBB) leakage in Alzheimer's disease (AD). In this study, we have measured levels of the pericyte marker, platelet-derived growth factor receptor-ß (PDGFRB), and fibrinogen (to assess blood-brain barrier leakage), and analyzed their relationship to indicators of microvessel density (von Willebrand factor level), ante-mortem oxygenation (myelin-associated glycoprotein:proteolipid protein-1 ratio and vascular endothelial growth factor level), Aß level and plaque load, in precuneus and underlying white matter from 49 AD to 37 control brains. There was reduction in PDGFRB and increased fibrinogen in the precuneus in AD. These changes correlated with reduction in oxygenation and with plaque load. In the underlying white matter, increased fibrinogen correlated with reduced oxygenation, but PDGFRB level was unchanged. The level of platelet-derived growth factor-ßß (PDGF-BB), important for pericyte maintenance, was increased in AD but mainly in the insoluble tissue fraction, correlating with insoluble Aß level. Loss of the PDGFRB within the precuneus in AD is associated with fibrinogen leakage and reduced oxygenation, and related to fibrillar Aß accumulation. In contrast, fibrinogen leakage and reduced oxygenation of underlying white matter occur independently of loss of PDGFRB, perhaps secondary to reduced transcortical perfusion.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/pathology , Brain/pathology , Pericytes/pathology , White Matter/pathology , Aged , Aged, 80 and over , Biomarkers/analysis , Capillary Permeability , Cerebrovascular Circulation/physiology , Female , Humans , Male , Receptor, Platelet-Derived Growth Factor beta/analysis , Receptor, Platelet-Derived Growth Factor beta/biosynthesis
9.
Curr Protoc Stem Cell Biol ; 39: 1H.10.1-1H.10.23, 2017 Nov 08.
Article in English | MEDLINE | ID: mdl-29081882

ABSTRACT

Oligodendrocytes are the main myelinating cell of the adult CNS and are vulnerable to injury in diverse disorders such as spinal cord injury, stroke, trauma, pharmacological and radiation toxicity, as well as neuroinflammation. Human pluripotent stem cells are attractive sources of oligodendrocyte lineage cells and provide a promising treatment strategy for exogenous myelin repair through transplantation. This unit describes a protocol for the step-wise differentiation of forebrain late oligodendrocyte progenitor cells (OPCs) from human pluripotent stem cells in defined chemical in vitro culture conditions. It involves a stepwise progression of oligodendrocyte progenitors through their known developmental phases, starting with the expression of appropriate transcription factors (Olig2, Nkx2.2), the upregulation of PDGFRA, followed by the appearance of O4-expressing cells, then O1 expression and finally mature myelin-binding protein (MBP) expressing cells. Validation of cell fate is performed by extensive transcriptomal profiling, as well in vitro myelination essays with hESCs derived neuronal cells. Recapitulating forebrain oligodendrocyte development may generate cells more suitable for transplantation strategies for disorders primarily involving the telencephalon.


Subject(s)
Cell Differentiation , Oligodendroglia/metabolism , Pluripotent Stem Cells/metabolism , Telencephalon/metabolism , Gene Expression Regulation , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/biosynthesis , Humans , Nuclear Proteins , Oligodendrocyte Transcription Factor 2/biosynthesis , Oligodendroglia/cytology , Pluripotent Stem Cells/cytology , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Telencephalon/cytology , Transcription Factors/biosynthesis , Transcription, Genetic , Zebrafish Proteins
10.
Eur Rev Med Pharmacol Sci ; 21(17): 3857-3865, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28975979

ABSTRACT

OBJECTIVE: In this study, we aimed to investigate the downstream effector of GLI2 in gastric cancer (GC) and their regulative effect on cancer stem cell (CSC) properties of GC. MATERIALS AND METHODS: Bioinformatic data mining was performed in TCGA-Stomach Adenocarcinoma (STAD), as well as in Kaplan-Meier plotter. Moderate-differentiated GC cell line SGC-7901 and poor-differentiated GC cell line MKN-45 were used as in-vitro model to investigate the regulative effect of GLI2 on PDGFRB expression. MKN-45 cells were further used to explore the effect of GLI2 shRNA or PDGFRB shRNA on CSC properties of the cells. RESULTS: Bioinformatic results showed that GLI2 is usually upregulated in GC tissues than in normal tissues, and high GLI2 expression is associated with unfavorable first progression free survival (PFS) and also worse overall survival (OS) in patients with GC. PDGFRB is co-upregulated with GLI2 in GC and its promoter region contains a putative GLI2 binding site. The results of dual luciferase assay confirmed this binding site. Enforced GLI2 expression elevated PDGFRB expression at both mRNA and protein level. GLI2 or PDGFRB knockdown showed similar effect on reducing spheroid colony formation and on reducing the expression of CSC related genes, including CD44, Nanog, and Oct4 in MKN-45 cells. CONCLUSIONS: High GLI2 or PDGFRB expression is associated with unfavorable survival in GC patients. GLI2 can induce PDGFRB expression in GC cells via directly binding to its promoter. In addition, the GLI2-PDGFRB axis might be an important signaling pathway modulating CSC properties of GC cells.


Subject(s)
Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Zinc Finger Protein Gli2/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Proliferation , Disease-Free Survival , Female , Gene Knockdown Techniques , Humans , Hyaluronan Receptors/biosynthesis , Nanog Homeobox Protein/biosynthesis , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/biosynthesis , Signal Transduction , Survival Analysis , Tumor Stem Cell Assay , Up-Regulation
11.
Oncotarget ; 8(11): 17771-17784, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28147311

ABSTRACT

Esophageal cancer is a highly aggressive malignancy with very poor overall prognosis. Given the strong clinical relevance of SATB1 in esophagus cancer and other cancers suggested by previous studies, the exact function of SATB1 in esophagus cancer development is still unknown. Here we showed that the knockdown of SATB1 in esophageal cancer cell lines diminished the cell proliferation, survival and invasion. Whole genome transcriptome analysis of SATB1 knockdown cells revealed the different gene expression profiles between TE-1 cells and MDA-MB-231 cells. Network analysis and functional experiments further identified FN1 and PDGFRB to be key downstream genes regulated by SATB1 in esophageal cancer cells. Importantly, FN1 and PDGFRB were found to be highly expressed in human esophageal cancer. In summary, we provided the first molecular evidence that SATB1 played an oncogenic role in esophageal cancer by up-regulation of FN1 and PDGFRB.


Subject(s)
Cell Proliferation/genetics , Esophageal Neoplasms/genetics , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Matrix Attachment Region Binding Proteins/genetics , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Disease Progression , Esophageal Neoplasms/pathology , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Matrix Attachment Region Binding Proteins/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , RNA Interference , RNA, Small Interfering/genetics , Transcriptional Activation , Up-Regulation
12.
J Recept Signal Transduct Res ; 37(2): 167-173, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27400779

ABSTRACT

Hyperuricemia and angiotensin II (Ang II) may have a pathogenetic role in the development of hypertension and atherosclerosis as well as cardiovascular disease (CVD) and its prognosis. The purpose of this study was to investigate whether uric acid can induce proliferative pathways of vascular smooth muscle cell (VSMC) that are thought to be responsible for the development of CVD. The phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK), p44/42 mitogen-activated protein kinase (p44/42 MAPK) and platelet-derived growth factor receptor ß (PDGFRß) was measured by Elisa and Western blot techniques to determine the activation of proliferative pathways in primary cultured VSMCs from rat aorta. Results demonstrated that uric acid can stimulate p38 MAPK, p44/42 MAPK and PDGFRß phosphorylation in a time- and concentration-dependent manner. Furthermore, treatment of VSMCs with the angiotensin II type I receptor (AT1R) inhibitor losartan suppressed p38 MAPK and p44/42 MAPK induction by uric acid. The stimulatory effect of uric acid on p38 MAPK was higher compared to that of Ang II. The results of this study show for the first time that uric acid-induced PDGFRß phosphorylation plays a crucial role in the development of CVDs and that elevated uric acid levels could be a potential therapeutical target in CVD patients.


Subject(s)
Cardiovascular Diseases/drug therapy , Mitogen-Activated Protein Kinase 3/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , p38 Mitogen-Activated Protein Kinases/biosynthesis , Angiotensin II/metabolism , Animals , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/pathology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Humans , Hypertension/drug therapy , Hypertension/genetics , Hypertension/pathology , Hyperuricemia/drug therapy , Hyperuricemia/genetics , Losartan/administration & dosage , Mitogen-Activated Protein Kinase 3/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phosphorylation/drug effects , Rats , Receptor, Angiotensin, Type 1/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Uric Acid/administration & dosage , p38 Mitogen-Activated Protein Kinases/genetics
13.
Vet Comp Oncol ; 15(1): 65-77, 2017 Mar.
Article in English | MEDLINE | ID: mdl-25665137

ABSTRACT

Information about histiocytic disease in cats is limited. The aim of this study was to document clinical findings and outcome in feline histiocytic disorders, and characterize the expression of PDGFRß and KIT in order to identify potential treatment targets. Morphologically diagnosed feline histiocytic tumours were reviewed and characterized by immunohistochemistry (IHC). Five cases of feline progressive histiocytosis (FPH), eight histiocytic sarcomas (HS) and two haemophagocytic histiocytic sarcomas (HaeHS) were confirmed. PDGFRß was variably positive in most histiocytic cases, while KIT was negative in all. Clinical presentation, treatment and outcome were also evaluated. Partial responses were recorded in measurable disease with tyrosine kinase inhibitors and lomustine, and radiotherapy achieved long-term control in some cases. Survival times were shortest in HaeHS and disseminated disease. PDGFRß, but not KIT, may represent a therapeutic target in feline histiocytic disorders but more studies are needed to investigate other potential treatment targets.


Subject(s)
Cat Diseases/metabolism , Histiocytic Disorders, Malignant/veterinary , Proto-Oncogene Proteins c-kit/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Animals , Biomarkers, Tumor , Cat Diseases/diagnosis , Cat Diseases/pathology , Cats , Databases as Topic , Female , Histiocytic Disorders, Malignant/metabolism , Histiocytic Disorders, Malignant/pathology , Histiocytic Disorders, Malignant/therapy , Immunohistochemistry/veterinary , Male , Neoplasm Staging , Treatment Outcome , United Kingdom
14.
World Neurosurg ; 98: 288-295, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27506406

ABSTRACT

OBJECTIVE: We sought to compare the prognosis of clival chordomas with different dural penetration and establish the relationship between dural penetration and platelet-derived growth factor receptor (PDGFR)-ß signaling pathway. METHODS: Tumors in Type I (33 cases) showed limited dural penetration, while those in Type II (34 cases) had more serious dural penetration. Cox multivariate regression analysis was used to analyze risk factors affecting survival. Kaplan-Meier analysis measured overall survival (OS) and progression-free survival (PFS). To determine the relationship between dural penetration and PDGFR-ß signaling, expression of PDGFR-ß, Akt, mammalian target of rapamycin (mTOR), and phosphatase and tensin homolog (PTEN) expression was compared using immunohistochemistry, quantitative reverse transcription polymerase chain reaction, and Western blotting. RESULTS: Total resection was achieved in 9 cases in Type I and 11 in Type II. There were significant correlations between OS and dural penetration (P = 0.032) and age (P = 0.034). PFS correlated significantly with dural penetration (P = 0.022), gender (P = 0.001), and degree of resection (P = 0.001). Mean OS in Type I was significantly longer than in Type II (P = 0.046). Patients aged <55 years had longer OS than those aged ≥55 years (P = 0.004). Total resection was correlated with longer PFS (P = 0.011). Among patients with tumors totally resected, mean PFS in Type I was significantly longer than in Type II (P = 0.007). Expression of PDGFR-ß in Type II was higher than in Type I. CONCLUSIONS: Clival chordomas have different degrees of dural penetration. Patients with chordomas with serious dural penetration have poorer prognosis. Higher expression of PDGFR-ß is related to more serious dural penetration of clival chordomas.


Subject(s)
Chordoma/metabolism , Dura Mater/metabolism , Gene Expression Regulation, Neoplastic , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Skull Base Neoplasms/metabolism , Adult , Chordoma/diagnostic imaging , Chordoma/surgery , Cranial Fossa, Posterior/diagnostic imaging , Cranial Fossa, Posterior/surgery , Dura Mater/diagnostic imaging , Dura Mater/surgery , Female , Humans , Male , Middle Aged , Prognosis , Receptor, Platelet-Derived Growth Factor beta/genetics , Skull Base Neoplasms/diagnostic imaging , Skull Base Neoplasms/surgery
15.
BMC Cardiovasc Disord ; 16(1): 179, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27619504

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPCs) play important roles in the regeneration of the vascular endothelial cells (ECs). Platelet-derived growth factor receptor (PDGFR)-ß is known to contribute to proliferation, migration, and angiogenesis of EPCs, this study aims to investigate effects of transplantation of EPCs overexpressing PDGFR-ß on vascular regeneration. METHODS: We transplanted genetically modified EPCs overexpressing PDGFR-ß into a mouse model with carotid artery injury. After 3 days of EPCs transplantation, the enhanced green fluorescent protein (EGFP)-expressing cells were found at the injury site and the lining of the lumen by laser scanning confocal microscope (LSCM). At 4, 7, and 14 days of the carotid artery injury, reendothelialization was evaluated by Evans Blue staining. Neointima formation was evaluated at day 14 with hematoxylin and eosin (HE) staining by calculating the neointimal area, medial area, and neointimal/media (NI/M) ratio. Intimal cell apoptosis was evaluated using TUNEL assay. Then we tested whether PDGF-BB-induced VSMC migration and PDGF-BB's function in reducing VSMC apoptosis can be attenuated by EPCs overexpressing PDGFR-ß in a transwell co-culture system. RESULTS: Our results showed that EPCs overexpressing PDGFR-ß accelerates reendothelialization and mitigates neointimal formation at 14 days after injury. Moreover, we found that there is great possibility that EPCs overexpressing PDGFR-ß enhanc VSMC apoptosis and suppress VSMC migration by competitive consumption of PDGF-BB in the early phase after carotid artery injury in mice. CONCLUSIONS: We report the first in vivo and in vitro evidence that transplantation of genetically modified EPC can have a combined effect of both amplifying the reendothelialization capacity of EPCs and inhibiting neointima formation so as to facilitate better inhibition of adverse remodeling after vascular injury.


Subject(s)
Carotid Artery Injuries/surgery , Endothelial Progenitor Cells/transplantation , Endothelium, Vascular/pathology , Gene Expression Regulation , Receptor, Platelet-Derived Growth Factor beta/genetics , Regeneration/genetics , Stem Cell Transplantation/methods , Animals , Carotid Artery Injuries/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Endothelium, Vascular/metabolism , Male , Mice , Mice, Inbred C57BL , Neointima/pathology , RNA/genetics , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
16.
Cancer Res ; 76(18): 5562-72, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27402080

ABSTRACT

Organization of cancer cells into endothelial-like cell-lined structures to support neovascularization and to fuel solid tumors is a hallmark of progression and poor outcome. In triple-negative breast cancer (TNBC), PDGFRß has been identified as a key player of this process and is considered a promising target for breast cancer therapy. Thus, we aimed at investigating the role of miRNAs as a therapeutic approach to inhibit PDGFRß-mediated vasculogenic properties of TNBC, focusing on miR-9 and miR-200. In MDA-MB-231 and MDA-MB-157 TNBC cell lines, miR-9 and miR-200 promoted and inhibited, respectively, the formation of vascular-like structures in vitro Induction of endogenous miR-9 expression, upon ligand-dependent stimulation of PDGFRß signaling, promoted significant vascular sprouting of TNBC cells, in part, by direct repression of STARD13. Conversely, ectopic expression of miR-200 inhibited this sprouting by indirectly reducing the protein levels of PDGFRß through the direct suppression of ZEB1. Notably, in vivo miR-9 inhibition or miR-200c restoration, through either the generation of MDA-MB-231-stable clones or peritumoral delivery in MDA-MB-231 xenografted mice, strongly decreased the number of vascular lacunae. Finally, IHC and immunofluorescence analyses in TNBC specimens indicated that PDGFRß expression marked tumor cells engaged in vascular lacunae. In conclusion, our results demonstrate that miR-9 and miR-200 play opposite roles in the regulation of the vasculogenic ability of TNBC, acting as facilitator and suppressor of PDGFRß, respectively. Moreover, our data support the possibility to therapeutically exploit miR-9 and miR-200 to inhibit the process of vascular lacunae formation in TNBC. Cancer Res; 76(18); 5562-72. ©2016 AACR.


Subject(s)
MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Triple Negative Breast Neoplasms/pathology , Animals , Blotting, Western , Cell Differentiation , Endothelial Cells/pathology , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Heterografts , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, SCID , Neovascularization, Pathologic/pathology , Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor beta/genetics , Triple Negative Breast Neoplasms/genetics
17.
Stem Cells Transl Med ; 5(6): 745-53, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27112178

ABSTRACT

UNLABELLED: : By using surgical mouse models, this study investigated how the tissue environment influences the osteogenic potential of muscle progenitors (m-progenitors) and potentially contributes to heterotopic ossification (HO). Injury was induced by clamping the gluteus maximus and medius (group M) or osteotomy of greater trochanter (group O) on the right hip, as well as combined muscle injury and osteotomy of greater trochanter (group M+O). The gluteus maximus and medius of the operated hips were harvested at days 1, 3, 5, and 10 for isolation of m-progenitors. The cells were cultured in an osteogenic medium for 3 weeks, and osteogenesis was evaluated by matrix mineralization and the expression of osteogenesis-related genes. The expression of type I collagen, RUNX2 (runt-related transcription factor 2), and osteocalcin by the m-progenitors of group M+O was significantly increased, compared with groups M and O. Osteogenic m-progenitors in group O increased the expression of bone morphogenetic protein 2 and also bone morphogenetic protein antagonist differential screening-selected gene aberrative in neuroblastoma. On histology, there was calcium deposition mostly in the muscles of group M+O harvested at day 10. CD56, representing myogenic progenitors, was highly expressed in the m-progenitors isolated from group M (day 10), but m-progenitors of group M+O (day 10) exhibited the highest expression of platelet-derived growth factor receptor α (PDGFR-α), a marker of muscle-derived mesenchymal stem cells (M-MSCs). The expressions of PDGFR-α and RUNX2 were colocalized in osteogenic m-progenitors. The data indicate that the tissue environment simulated in the M+O model is a favorable condition for HO formation. Most likely, M-MSCs, rather than myogenic progenitors, in the m-progenitors participate in HO formation. SIGNIFICANCE: The prevalence of traumatic heterotopic ossification (HO) is high in war injury. The pathogenesis of HO is still unknown. This study clarified the contribution of a tissue environment created by bone or muscle injury to the formation of HO. The study also found that muscle-derived mesenchymal stem cells, but not myogenic progenitors, are involved in the formation of HO. The findings of this study could be used to strategize the prevention and treatment of HO.


Subject(s)
Mesenchymal Stem Cells , Muscle, Skeletal/growth & development , Ossification, Heterotopic/physiopathology , Osteogenesis/genetics , Animals , Bone Development/genetics , Bone and Bones/injuries , Bone and Bones/physiopathology , Bone and Bones/surgery , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Humans , Mice , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , Muscle, Skeletal/surgery , Ossification, Heterotopic/surgery , Ossification, Heterotopic/therapy , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics
18.
Tumour Biol ; 37(8): 10577-86, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26857280

ABSTRACT

Glioblastoma (GBM) is the highly malignant glioma and exhibits microvascular proliferation. PCR mRNA arrays and immunohistochemical stains on tissue microarray demonstrated that the expression level of PDGFRB in GBM microvascular proliferation was significantly higher than that in GBM tumor cells while the expression level of EGFR was lower in microvascular proliferation than in GBM tumor cells. PDGFRB protein was selectively expressed in pericytes in GBM microvascular proliferation. By analyzing The Cancer Genome Atlas (TCGA) datasets for GBM, it was found that genomic DNA alterations were the main reason for the high expression of EGFR in GBM tumor cells. Our miRNA microarray data showed that microRNAs (miRNAs) (miR-193b-3p, miR-518b, miR-520f-3p, and miR-506-5p) targeting PDGFRB were downregulated in microvascular proliferation, which might be the most likely reason for the high expression of PDGFRB in GBM microvascular proliferation. The increase of several miRNAs (miR-133b, miR-30b-3p, miR-145-5p, and miR-146a-5p) targeting EGFR in GBM microvascular proliferation was one of the reasons for the lack of expression of EGFR in GBM microvascular proliferation. These findings implicated that miRNAs, such as miR-506, miR-133b, miR-145, and miR-146a, that target PDGFRB or EGFR, might be potential therapeutic agents for GBM. A new generation of targeted therapeutic agents against both EGFR and PDGFRB might be developed in the future.


Subject(s)
Brain Neoplasms/blood supply , ErbB Receptors/biosynthesis , Gene Expression Regulation, Neoplastic , Glioblastoma/blood supply , Neoplasm Proteins/biosynthesis , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Female , Genes, erbB-1 , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Tissue Array Analysis
19.
Exp Hematol ; 44(3): 177-88.e5, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26703895

ABSTRACT

ATF7IP-PDGFRB is a novel PDGFRB-related fusion gene identified in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) with a signature similar to that of Ph1 ALL, so-called Ph-like ALL. When we introduced ATF7IP-PDGFRB, murine Ba/F3 cells acquired the ability to proliferate in an interleukin (IL)-3-independent manner. On the contrary, the expression of wild-type PDGFRB is not sufficient to acquire the ability for IL-3-independent proliferation in Ba/F3 cells. The introduction of ATF7IP-PDGFRB also induces a typical gene expression profile for Ph1-ALL in Ba/F3 cells. A series of biochemical and cell biological experiments revealed the constitutive activation of ATF7IP-PDGFRB as well as downstream signaling molecules, including AKT and MAPK. Although the phosphoinositide 3-kinase inhibitor led to cell death in both cells into which ATF7IP-PDGFRB had been introduced and IL-3-maintained Mock cells, MEK inhibitor selectively led to cell death into which ATF7IP-PDGFRB had been introduced. The introduction of tyrosine to phenylalanine mutations at binding sites of adaptor molecules important in the MAPK pathway located in the PDGFRB portion abolished ATF7IP-PDGFRB-mediated cell transformation, suggesting that MAPK-mediated signals are critical in ATF7IP-PDGFRB-mediated cell transformation. On treatment with tyrosine kinase inhibitors, ATF7IP-PDGFRB-expressing, but not Mock, Ba/F3 cells underwent rapid apoptosis accompanied by reduced phosphorylation of MAPK. Importantly, the sensitivity of ATF7IP-PDGFRB-expressing Ba/F3 cells to imatinib is significantly higher than that of BCR-ABL1-transformed Ba/F3 cells, as assessed by the IC50. Taken together, ATF7IP-PDGFRB has transforming potential via the constitutive activation of MAPK and participates in the pathogenesis of Ph-like ALL. Our observations suggest the therapeutic importance of tyrosine kinase inhibitors and possibly MEK inhibitor for a subset of BCP-ALL harboring PDGFRB-related fusion kinases.


Subject(s)
Gene Expression Regulation, Leukemic/drug effects , Oncogene Proteins, Fusion , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Protein Kinase Inhibitors/pharmacology , Receptor, Platelet-Derived Growth Factor beta , Repressor Proteins , Animals , Cell Line, Tumor , Humans , Mice , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/biosynthesis , Repressor Proteins/genetics
20.
PLoS One ; 10(11): e0141462, 2015.
Article in English | MEDLINE | ID: mdl-26556479

ABSTRACT

Pancreatic fibrosis, a prominent feature of chronic pancreatitis (CP), induces persistent and permanent damage in the pancreas. Pancreatic stellate cells (PSCs) provide a major source of extracellular matrix (ECM) deposition during pancreatic injury, and persistent activation of PSCs plays a vital role in the progression of pancreatic fibrosis. Retinoic acid (RA), a retinoid, has a broad range of biological functions, including regulation of cell differentiation and proliferation, attenuating progressive fibrosis of multiple organs. In the present study, we investigated the effects of RA on fibrosis in experimental CP and cultured PSCs. CP was induced in mice by repetitive cerulein injection in vivo, and mouse PSCs were isolated and activated in vitro. Suppression of pancreatic fibrosis upon administration of RA was confirmed based on reduction of histological damage, α-smooth muscle actin (α-SMA) expression and mRNA levels of ß-catenin, platelet-derived growth factor (PDGF)-Rß transforming growth factor (TGF)-ßRII and collagen 1α1 in vivo. Wnt 2 and ß-catenin protein levels were markedly down-regulated, while Axin 2 expression level was up-regulated in the presence of RA, both in vivo and in vitro. Nuclear translation of ß-catenin was significantly decreased following RA treatment, compared with cerulein-induced CP in mice and activated PSCs. Furthermore, RA induced significant PSC apoptosis, inhibited proliferation, suppressed TCF/LEF-dependent transcriptional activity and ECM production of PSC via down-regulation of TGFßRII, PDGFRß and collagen 1α1 in vitro. These results indicate a critical role of the Wnt/ß-catenin signaling pathway in RA-induced effects on CP and PSC regulation and support the potential of RA as a suppressor of pancreatic fibrosis in mice.


Subject(s)
Pancreatic Stellate Cells/drug effects , Pancreatitis, Chronic/drug therapy , Tretinoin/therapeutic use , Wnt Signaling Pathway/drug effects , Actins/biosynthesis , Actins/genetics , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Axin Protein/biosynthesis , Axin Protein/genetics , Cells, Cultured , Ceruletide/toxicity , Collagen Type I/biosynthesis , Collagen Type I/genetics , Disease Progression , Drug Evaluation, Preclinical , Fibrosis/prevention & control , Gene Expression Regulation/drug effects , Lipase/blood , Male , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Pancreas/drug effects , Pancreas/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic alpha-Amylases/blood , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Proteoglycans/biosynthesis , Proteoglycans/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...