1.Pathomechanisms of pericyte-myofibroblast transition in kidney and interventional effects of Chinese herbal medicine.
Ying-Lu LIU ; Ge SHI ; Dong-Wei CAO ; Yi-Gang WAN ; Wei WU ; Yue TU ; Bu-Hui LIU ; Wen-Bei HAN ; Jian YAO
China Journal of Chinese Materia Medica 2018;43(21):4192-4197
In the kidney, pericyte is the major source of myofibroblast (MyoF) in renal interstitium. It is reported that pericyte-myofibroblast transition(PMT)is one of the important pathomechanisms of renal interstitial fibrosis(RIF). Among them, the main reasons for promoting RIF formation include pericyte recruitment, activation and isolation, as well as the lack of pericyte-derived erythropoietin. During the PMT startup process, pericyte activation and its separation from microvessels are controlled by multiple signal transduction pathways, such as transforming growth factor-β(TGF-β)pathway, vascular endothelial growth factor receptor (VEGFR) pathway and platelet derived growth factor receptor (PDGFR) pathway;Blocking of these signaling pathways can not only inhibit PMT, but also suppress renal capillaries reduction and further alleviate RIF. In clinic, many traditional Chinese medicine compound prescriptions, single traditional Chinese herbal medicine (CHM) and their extracts have the clear effects in alleviating RIF, and some of their intervention actions may be related to pericyte and its PMT. Therefore, the studies on PMT and its drug intervention will become the main development direction in the research field of anti-organ fibrosis by CHM.
Drugs, Chinese Herbal
;
pharmacology
;
Fibrosis
;
Humans
;
Kidney
;
cytology
;
drug effects
;
pathology
;
Myofibroblasts
;
cytology
;
Pericytes
;
cytology
;
Receptors, Platelet-Derived Growth Factor
;
metabolism
;
Signal Transduction
;
Vascular Endothelial Growth Factor A
;
metabolism
2.Phase II Study of Dovitinib in Patients with Castration-Resistant Prostate Cancer (KCSG-GU11-05).
Yoon Ji CHOI ; Hye Sook KIM ; Se Hoon PARK ; Bong Seog KIM ; Kyoung Ha KIM ; Hyo Jin LEE ; Hong Suk SONG ; Dong Yeop SHIN ; Ha Young LEE ; Hoon Gu KIM ; Kyung Hee LEE ; Jae Lyun LEE ; Kyong Hwa PARK
Cancer Research and Treatment 2018;50(4):1252-1259
PURPOSE: Fibroblast growth factor (FGF) signals are important in carcinogenesis and progression of prostate cancer. Dovitinib is an oral, pan-class inhibitor of vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor, and fibroblast growth factor receptor (FGFR). We evaluated the efficacy and toxicity of dovitinib in men with metastatic castration resistant prostate cancer (mCRPC). MATERIALS AND METHODS: This study was a single-arm, phase II, open-label, multicenter trial of dovitinib 500 mg/day (5-days-on/2-days-off schedule). The primary endpoint was 16-week progression-free survival (PFS). Secondary endpoints were overall survival (OS), toxicity and prostate-specific antigen (PSA) response rate. Biomarker analyses for VEGFR2, FGF23, and FGFR2 using multiplex enzyme-linked immunosorbent assay was performed. RESULTS: Forty-four men were accrued from 11 hospitals. Eighty percent were post-docetaxel. Median PSA was 100 ng/dL, median age was 69, 82% had bone metastases, and 23% had liver metastases. Median cycles of dovitinib was 2 (range, 0 to 33). Median PFS was 3.67 months (95% confidence interval [CI], 1.36 to 5.98) and median OS was 13.70 months (95% CI, 0 to 27.41). Chemotherapy-naïve patients had longer PFS (17.90 months; 95% CI, 9.23 to 28.57) compared with docetaxel-treated patients (2.07 months; 95% CI, 1.73 to 2.41; p=0.001) and the patients with high serum VEGFR2 level over median level (7,800 pg/mL) showed longer PFS compared with others (6.03 months [95% CI, 4.26 to 7.80] vs. 1.97 months [95% CI, 1.79 to 2.15], p=0.023). Grade 3 related adverse events were seen in 40.9% of patients. Grade 1-2 nausea, diarrhea, fatigue, anorexia, and all grade thrombocytopenia are common. CONCLUSION: Dovitinib showed modest antitumor activity with manageable toxicities in men with mCRPC. Especially, patients who were chemo-naïve benefitted from dovitinib.
Anorexia
;
Biomarkers
;
Carcinogenesis
;
Castration
;
Diarrhea
;
Disease-Free Survival
;
Enzyme-Linked Immunosorbent Assay
;
Fatigue
;
Fibroblast Growth Factors
;
Humans
;
Liver
;
Male
;
Multicenter Studies as Topic
;
Nausea
;
Neoplasm Metastasis
;
Prostate*
;
Prostate-Specific Antigen
;
Prostatic Neoplasms*
;
Prostatic Neoplasms, Castration-Resistant
;
Receptors, Fibroblast Growth Factor
;
Receptors, Platelet-Derived Growth Factor
;
Receptors, Vascular Endothelial Growth Factor
;
Thrombocytopenia
3.A Case of Myeloid Neoplasm with a PDGFRB Rearrangement and Eosinophilia.
Nam Gil CHO ; Daejin KIM ; Minjung SHIM ; So Young CHONG
Korean Journal of Medicine 2017;92(1):79-83
Myeloid neoplasia with eosinophilia and platelet-derived growth factor receptor beta (PDGFRB) rearrangements is an uncommon Philadelphia-negative myeloproliferative neoplasm. Their most common morphological diagnosis is chronic myelomonocytic leukemia with eosinophilia, which is associated with t(5;12)(q33;p13) and results in the formation of the ETV6-PDGFRB fusion gene. Here, we report a 49-year-old man with a myeloid neoplasm with a PDGFRB rearrangement, who was incidentally diagnosed with hyperleukocytosis and eosinophilia during a health screening. A chromosome analysis of a bone marrow sample revealed 46, XY, t(5;12)(q33;p13), and fluorescence in situ hybridization analysis revealed the PDGFRB gene rearrangement. The patient was treated with imatinib and subsequently achieved complete hematological and molecular remission.
Bone Marrow
;
Diagnosis
;
Eosinophilia*
;
Fluorescence
;
Gene Rearrangement
;
Humans
;
Imatinib Mesylate
;
In Situ Hybridization
;
Leukemia, Myelomonocytic, Chronic
;
Mass Screening
;
Middle Aged
;
Myeloproliferative Disorders
;
Receptor, Platelet-Derived Growth Factor beta*
;
Receptors, Platelet-Derived Growth Factor
4.Platelet-Derived Growth Factor Receptor-Positive Pericytic Cells of White Adipose Tissue from Critical Limb Ischemia Patients Display Mesenchymal Stem Cell-Like Properties.
Eo Jin KIM ; Sang Gyo SEO ; Hyuk Soo SHIN ; Doo Jae LEE ; Ji Hye KIM ; Dong Yeon LEE
Clinics in Orthopedic Surgery 2017;9(2):239-248
BACKGROUND: The pericytes in the blood vessel wall have recently been identified to be important in regulating vascular formation, stabilization, remodeling, and function. We isolated and identified pericyte-like platelet-derived growth factor receptor beta-positive (PDGFRβ+) cells from the stromal vascular fraction (SVF) of adipose tissue from critical limb ischemia (CLI) patients and investigated their potential as a reliable source of stem cells for cell-based therapy. METHODS: De-identified subcutaneous fat tissues were harvested after amputation in CLI patients. Freshly isolated SVF cells and culture-expanded adipose-derived stem cells (ADSCs) were quantified using flow cytometry. A matrigel tube formation assay and multi-lineage differentiation were performed to assess pericytic and mesenchymal stem cell (MSC)-like characteristics of PDGFRβ+ ADSCs. RESULTS: PDGFRβ+ cells were located in the pericytic area of various sizes of blood vessels and coexpressed mesenchymal stem cell markers. PDGFRβ+ cells in freshly isolated SVF cells expressed a higher level of stem cell markers (CD34 and CXCR4) and mesenchymal markers (CD13, CD44, CD54, and CD90) than PDGFRβ– cells. In vitro expansion of PDGFRβ+ cells resulted in enrichment of the perivascular mesenchymal stem-like (PDGFRβ+/CD90+/CD45–/CD31–) cell fractions. The Matrigel tube formation assay revealed that PDGFRβ+ cells were located in the peritubular area. CONCLUSIONS: PDGFRβ+ ADSCs cells demonstrated a good multilineage differentiation potential. Pericyte-like PDGFRβ+ cells from the SVF of adipose tissue from CLI patients had MSC-like characteristics and could be amplified by in vitro culture with preservation of their cell characteristics. We believe PDGFRβ+ cells in the SVF of adipose tissue can be used as a reliable source of stem cells even in CLI patients.
Adipose Tissue
;
Adipose Tissue, White*
;
Adult Stem Cells
;
Amputation
;
Blood Vessels
;
Extremities*
;
Flow Cytometry
;
Humans
;
In Vitro Techniques
;
Ischemia*
;
Mesenchymal Stromal Cells
;
Pericytes
;
Platelet-Derived Growth Factor*
;
Receptors, Platelet-Derived Growth Factor
;
Stem Cells
;
Subcutaneous Fat
5.Impaired angiogenesis in the enalapril-treated neonatal rat kidney.
Hyung Eun YIM ; Kee Hwan YOO ; Eun Soo BAE ; Young Sook HONG ; Joo Won LEE
Korean Journal of Pediatrics 2016;59(1):8-15
PURPOSE: Nephrogenesis is normally accompanied by a tightly regulated and efficient vascularization. We investigated the effect of angiotensin II inhibition on angiogenesis in the developing rat kidney. METHODS: Newborn rat pups were treated with enalapril (30 mg/kg/day) or vehicle (control) for 7 days after birth. Renal histological changes were checked using Hematoxylin & Eosin staining. We also investigated the intrarenal expression of vascular endothelial growth factor (VEGF)-A, VEGF receptor 1 (VEGFR1), VEGFR2, platelet-derived growth factor (PDGF)-B, and PDGF receptor-beta with Western blotting and immunohistochemical staining at postnatal day 8. Expression of the endothelial cell marker CD31 was examined to determine glomerular and peritubular capillary density. RESULTS: Enalapril-treated rat kidneys showed disrupted tubules and vessels when compared with the control rat kidneys. In the enalapril-treated group, intrarenal VEGF-A protein expression was significantly higher, whereas VEGFR1 protein expression was lower than that in the control group (P<0.05). The expression of VEGFR2, PDGF-B, and PDGF receptor-beta was not different between the 2 groups. The increased capillary CD31 expression on the western blots of enalapril-treated rat kidneys indicated that the total endothelial cell protein level was increased, while the cortical capillary density, assessed using CD31 immunohistochemical staining, was decreased. CONCLUSION: Impaired VEGF-VEGFR signaling and altered capillary repair may play a role in the deterioration of the kidney vasculature after blocking of angiotensin II during renal development.
Angiotensin II
;
Animals
;
Blotting, Western
;
Capillaries
;
Enalapril
;
Endothelial Cells
;
Eosine Yellowish-(YS)
;
Growth and Development
;
Hematoxylin
;
Humans
;
Infant, Newborn
;
Kidney*
;
Parturition
;
Platelet-Derived Growth Factor
;
Rats*
;
Receptors, Vascular Endothelial Growth Factor
;
Vascular Endothelial Growth Factor A
6.Efficacy of Imatinib in Patients with Platelet-Derived Growth Factor Receptor Alpha-Mutated Gastrointestinal Stromal Tumors.
Changhoon YOO ; Min Hee RYU ; Jungmin JO ; Inkeun PARK ; Baek Yeol RYOO ; Yoon Koo KANG
Cancer Research and Treatment 2016;48(2):546-552
PURPOSE: The incidence of gastrointestinal stromal tumors (GISTs) harboring platelet-derived growth factor receptor alpha (PDGFRA) mutations is low, therefore further investigation of the efficacy of imatinib in this subgroup was needed. MATERIALS AND METHODS: Patients with PDGFRA-mutant GISTs who received imatinib as primary therapy for advanced disease between January 2000 and June 2012 were identified from the GIST registry of Asan Medical Center, Seoul, Korea. RESULTS: KIT and PDGFRA genotyping in 823 patients identified 18 patients (2%) with PDGFRA mutations who were treated with first-line imatinib. Exon 18 D842V substitution, non-D842V exon 18 mutations, and exon 12 mutations were detected in nine (50%), four (22%), and five (28%) patients, respectively. Objective response rate differed significantly between patients with the D842V mutation and those with non-D842V mutations (0% [0/5] vs. 71% [5/7], p=0.03). In all patients, median progression-free survival (PFS) and overall survival (OS) was 24.8 months (95% confidence interval [CI], 0.0 to 57.2) and 51.2 months (95% CI, 37.1 to 65.3), respectively. Significantly, poorer PFS was observed for patients with D842V-mutant GISTs than those with non-D842V PDGFRA-mutant GISTs: median 3.8 months (95% CI, 1.4 to 6.3) versus 29.5 months (95% CI, 18.3 to 40.7) (p < 0.001). Patients with the D842V mutation had poorer OS than those with non-D842V PDGFRA mutations: median 25.2 months (95% CI, 12.7 to 37.8) versus 59.8 months (95% CI, 43.0 to 76.5) (p=0.02). CONCLUSION: Imatinib is active against non-D842V PDGFRA-mutant GISTs, whereas GISTs harboring the D842V mutation are primarily resistant to imatinib.
Chungcheongnam-do
;
Disease-Free Survival
;
Exons
;
Gastrointestinal Stromal Tumors
;
Humans
;
Incidence
;
Korea
;
Platelet-Derived Growth Factor*
;
Receptors, Platelet-Derived Growth Factor*
;
Seoul
7.VEGF and Ki-67 Overexpression in Predicting Poor Overall Survival in Adenoid Cystic Carcinoma.
Seongyeol PARK ; Soo Jeong NAM ; Bhumsuk KEAM ; Tae Min KIM ; Yoon Kyung JEON ; Se Hoon LEE ; J Hun HAH ; Tack Kyun KWON ; Dong Wan KIM ; Myung Whun SUNG ; Dae Seog HEO ; Yung Jue BANG
Cancer Research and Treatment 2016;48(2):518-526
PURPOSE: The purpose of this study was to evaluate potential prognostic factors in patients with adenoid cystic carcinoma (ACC). MATERIALS AND METHODS: A total of 68 patients who underwent curative surgery and had available tissue were enrolled in this study. Their medical records and pathologic slides were reviewed and immunohistochemistry for basic fibroblast growth factor, fibroblast growth factor receptor (FGFR) 2, FGFR3, c-kit, Myb proto-oncogene protein, platelet-derived growth factor receptor beta, vascular endothelial growth factor (VEGF), and Ki-67 was performed. Univariate and multivariate analysis was performed for determination of disease-free survival (DFS) and overall survival (OS). RESULTS: In univariate analyses, primary site of nasal cavity and paranasal sinus (p=0.022) and Ki-67 expression of more than 7% (p=0.001) were statistically significant factors for poor DFS. Regarding OS, perineural invasion (p=0.032), high expression of VEGF (p=0.033), and high expression of Ki-67 (p=0.007) were poor prognostic factors. In multivariate analyses, primary site of nasal cavity and paranasal sinus (p=0.028) and high expression of Ki-67 (p=0.004) were independent risk factors for poor DFS, and high expression of VEGF (p=0.011) and Ki-67 (p=0.011) showed independent association with poor OS. CONCLUSION: High expression of VEGF and Ki-67 were independent poor prognostic factors for OS in ACC.
Adenoids*
;
Carcinoma, Adenoid Cystic*
;
Disease-Free Survival
;
Fibroblast Growth Factor 2
;
Humans
;
Immunohistochemistry
;
Medical Records
;
Multivariate Analysis
;
Nasal Cavity
;
Prognosis
;
Proto-Oncogenes
;
Receptors, Fibroblast Growth Factor
;
Receptors, Platelet-Derived Growth Factor
;
Risk Factors
;
Vascular Endothelial Growth Factor A*
8.Different anti-remodeling effect of nilotinib and fluticasone in a chronic asthma model.
Hye Seon KANG ; Chin Kook RHEE ; Hea Yon LEE ; Hyoung Kyu YOON ; Soon Seok KWON ; Sook Young LEE
The Korean Journal of Internal Medicine 2016;31(6):1150-1158
BACKGROUND/AIMS: Inhaled corticosteroids are the most effective treatment currently available for asthma, but their beneficial effect against airway remodeling is limited. The tyrosine kinase inhibitor nilotinib has inhibitory activity against c-kit and the platelet-derived growth factor receptor. We compared the effects of fluticasone and nilotinib on airway remodeling in a chronic asthma model. We also examined whether co-treatment with nilotinib and fluticasone had any synergistic effect in preventing airway remodeling. METHODS: We developed a mouse model of airway remodeling, including smooth muscle thickening, in which ovalbumin (OVA)-sensitized female BALB/c-mice were repeatedly exposed to intranasal OVA administration twice per week for 3 months. Mice were treated with fluticasone and/or nilotinib intranasally during the OVA challenge. RESULTS: Mice chronically exposed to OVA developed eosinophilic airway inflammation and showed features of airway remodeling, including thickening of the peribronchial smooth muscle layer. Both fluticasone and nilotinib attenuated airway smooth muscle thickening. However, only nilotinib suppressed fibrotic changes, demonstrating inhibition of collagen deposition. Fluticasone reduced pro-inflammatory cells, such as eosinophils, and several cytokines, such as interleukin 4 (IL-4), IL-5, and IL-13, induced by repeated OVA challenges. On the other hand, nilotinib reduced transforming growth factor β1 levels in bronchoalveolar lavage fluid and inhibited fibroblast proliferation significantly. CONCLUSIONS: These results suggest that fluticasone and nilotinib suppressed airway remodeling in this chronic asthma model through anti-inflammatory and anti-fibrotic pathways, respectively.
Adrenal Cortex Hormones
;
Airway Remodeling
;
Animals
;
Asthma*
;
Bronchoalveolar Lavage Fluid
;
Collagen
;
Cytokines
;
Eosinophils
;
Female
;
Fibroblasts
;
Fluticasone*
;
Hand
;
Humans
;
Inflammation
;
Interleukin-13
;
Interleukin-4
;
Interleukin-5
;
Mice
;
Muscle, Smooth
;
Ovalbumin
;
Ovum
;
Protein-Tyrosine Kinases
;
Receptors, Platelet-Derived Growth Factor
;
Transforming Growth Factors
9.Nuclear factor I-C inhibits platelet-derived growth factor-induced enhancement of dermal fibroblast sensitivity to TGF-β.
Liangping ZHANG ; Yang WANG ; Rui LEI ; Hui SHEN ; Yichen SHEN ; Zhinan WU ; Jinghong XU
Journal of Southern Medical University 2015;35(9):1245-1250
OBJECTIVETo investigate the effect of nuclear factor I-C (NFI-C) on platelet-derived growth factor (PDGF)-induced up-regulation of TGF-β receptor II (TβRII) in dermal fibroblasts.
METHODSA lentiviral vector containing NFI-C sequence (Lenti-GFP-NFI-C) was transfected into a human foreskin fibroblast cell line (HFF-1). Cultured HFF-1 cells, cells transfected with Lenti-GFP-NFI-C, and cells transfected with a negative virus were stimulated with PDGF-BB, and Western blotting and RT-qPCR were used to detect the expression levels of TβRII in the treated cells.
RESULTSPDGF treatment significantly increased the expression level of TβRII in HFF-1 cells (P<0.05). The cells transfected with Lenti-GFP-NFI-C expressed a significantly lower level of TβRII than non-transfected cells in response to PDGF stimulation (P<0.05), but the negative virus showed no such inhibitory effect (P>0.05). No significant difference was found in the expression level of TβRII protein between cells transfected with Lenti-GFP-NFI-C-transfection before PDGF stimulation and the blank control cells.
CONCLUSIONNFI-C can inhibit PDGF-induced up-regulation of TβRII and thus reduce the sensitivity of the dermal fibroblasts to TGF-β.
Cell Line ; Fibroblasts ; drug effects ; Genetic Vectors ; Humans ; Lentivirus ; NFI Transcription Factors ; genetics ; Platelet-Derived Growth Factor ; pharmacology ; Protein-Serine-Threonine Kinases ; metabolism ; Proto-Oncogene Proteins c-sis ; Receptors, Transforming Growth Factor beta ; metabolism ; Transfection ; Transforming Growth Factor beta ; pharmacology ; Up-Regulation
10.Serum Response Factor Is Essential for Prenatal Gastrointestinal Smooth Muscle Development and Maintenance of Differentiated Phenotype.
Chanjae PARK ; Moon Young LEE ; Paul J PARK ; Se Eun HA ; Robyn M BERENT ; Robert FUCHS ; Joseph M MIANO ; Laren S BECKER ; Kenton M SANDERS ; Seungil RO
Journal of Neurogastroenterology and Motility 2015;21(4):589-602
BACKGROUND/AIMS: Smooth muscle cells (SMCs) characteristically express serum response factor (SRF), which regulates their development. The role of SRF in SMC plasticity in the pathophysiological conditions of gastrointestinal (GI) tract is less characterized. METHODS: We generated SMC-specific Srf knockout mice and characterized the prenatally lethal phenotype using ultrasound biomicroscopy and histological analysis. We used small bowel partial obstruction surgeries and primary cell culture using cell-specific enhanced green fluorescent protein (EGFP) mouse lines to study phenotypic and molecular changes of SMCs by immunofluorescence, Western blotting, and quantitative polymerase chain reaction. Finally we examined SRF change in human rectal prolapse tissue by immunofluorescence. RESULTS: Congenital SMC-specific Srf knockout mice died before birth and displayed severe GI and cardiac defects. Partial obstruction resulted in an overall increase in SRF protein expression. However, individual SMCs appeared to gradually lose SRF in the hypertrophic muscle. Cells expressing low levels of SRF also expressed low levels of platelet-derived growth factor receptor alpha (PDGFRalphalow) and Ki67. SMCs grown in culture recaptured the phenotypic switch from differentiated SMCs to proliferative PDGFRalphalow cells. The immediate and dramatic reduction of Srf and Myh11 mRNA expression confirmed the phenotypic change. Human rectal prolapse tissue also demonstrated significant loss of SRF expression. CONCLUSIONS: SRF expression in SMCs is essential for prenatal development of the GI tract and heart. Following partial obstruction, SMCs down-regulate SRF to transition into proliferative PDGFRalphalow cells that may represent a phenotype responsible for their plasticity. These findings demonstrate that SRF also plays a critical role in the remodeling process following GI injury.
Animals
;
Blotting, Western
;
Fluorescent Antibody Technique
;
Gastrointestinal Tract
;
Heart
;
Humans
;
Mice
;
Mice, Knockout
;
Microscopy, Acoustic
;
Muscle Cells
;
Muscle, Smooth*
;
Myocytes, Smooth Muscle
;
Parturition
;
Phenotype*
;
Plastics
;
Polymerase Chain Reaction
;
Primary Cell Culture
;
Receptors, Platelet-Derived Growth Factor
;
Rectal Prolapse
;
RNA, Messenger
;
Serum Response Factor*

Result Analysis
Print
Save
E-mail