Review Article PDGF Signaling in Cancer Progression

Total Page:16

File Type:pdf, Size:1020Kb

Review Article PDGF Signaling in Cancer Progression Int J Clin Exp Med 2017;10(7):9918-9929 www.ijcem.com /ISSN:1940-5901/IJCEM0055300 Review Article PDGF signaling in cancer progression Feng Huang1, Dong Wang2, Yongliang Yao1, Mei Wang3 1Department of Clinical Laboratory, The First People’s Hospital of Kunshan, Affiliated to Jiangsu University, Suzhou, Jiangsu Province, China; 2Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong Univer- sity, Nantong, China; 3School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China Received April 12, 2017; Accepted June 7, 2017; Epub July 15, 2017; Published July 30, 2017 Abstract: Platelet-derived growth factor (PDGF) is a pro-angiogenic factor that was isolated from human platelets. PDGF family is comprised of four different polypeptide chains encoded by different genes, which have been iden- tified: PDGF-A, PDGF-B, and recently discovered PDGF-C and PDGF-D. PDGF is a variety of strong mesenchymal cell mitogenic agents and growth chemokines. PDGF, acting as a vascular endothelial growth factor, is closely as- sociated with tumor development. PDGF signaling pathway has been extensively studied and well characterized because PDGF can regulate many cellular processes, including cell proliferation, migration, invasion, angiogenesis and metastasis. According to the available data, PDGF plays an important role in the development of breast cancer, stomach cancer, gastric cancer, prostate cancer, lung cancer, colon cancer, and other cancers. Effects of PDGF on cancer are complex and diverse, and further analysis is needed to confirm the predictive and therapeutic values of PDGF and its receptor. This article reviews the recent progress of PDGF in tumor progression. Keywords: Platelet-derived growth factor, signal pathway, proliferation, migration, invasion, angiogenesis, metas- tasis, tumor progression Introduction PDGF domain, which can induce the phosphory- lation of β-PDGF receptor and stimulate LNCaP Platelet-derived growth factor (PDGF) is a pro- cell proliferation in an autocrine manner. angiogenic factor that was isolated from human Additionally, LNCaP-PDGFD-conditioned medi- platelets [1-3]. PDGFs are a variety of strong um induces migration of the prostate fibroblast mesenchymal cell mitogenic agents and growth cell line 1532-FTX, indicating LNCaP-processed chemokines; they are also important modifiers PDGFD acts in a paracrine manner as well. The for the normal and pathological vascular devel- PDGF signaling pathway combines with a vari- opment [4-6]. PDGF, acting as vascular endo- ety of substrate protein signaling molecules, thelial growth factor, plays a pivotal role in the including SRC tyrosine kinase subfamily, development and progression of human malig- phospholipase C (PLC-γ), phosphatidylinositol nancies. The latest findings show that PDGFs 3-kinase (PI3-K), GTPase activation protein regulate tumor growth and metastasis by tar- (RAS-gap), growth factor binding protein (Grb2), geting malignant cells, vascular cells, and stro- tyrosine specific phosphatase (SYP), Src homol- mal cells [7-10]. ogy and cross-linked protein (SHC) and adapta- PDGF and cancer cells tion (Crk) protein, and non-receptor tyrosine kinase family (SRC) and so on, and forms a vari- Cancer cell proliferation ety of signaling pathways, such as RAS-MAPK pathway, PI3-K pathway, the PLC pathway, and PDGF stimulates cancer cell proliferation STAT pathway [13]. These signal molecules through autocrine and paracrine manners, mediate cytoplasmic complex signaling net- since some cancer cells express the PDGF works, and play a role in cell membrane serine receptor and some do not. For example, Ustach threonine residues, resulting in a variety of et al. [11, 12] demonstrated that LNCaP cells gene regulation and protein phosphorylation, auto-activate latent PDGFD into the active thus promoting cancer cell growth and division PDGF signaling in cancer progression [14]. For example, PDGF has been proved to ing number of researchers believe that cancer promote the proliferation of human meningio- growth, metastasis, and invasion depend not mas and mesothelioma cell through PI3-K only on the tumor cells themselves, but also on pathway [15, 16]. Lu Y. demonstrated that the growth of tumor microenvironment, which hypoxia-induced PDGF-BB secretion by HCC supports cancer behavior. Studies have shown cells stimulates HSCs to accumulate and prolif- that PDGF-C in breast cancer activates the erate in the tumor stroma [17]. Whereas angio- receptor by binding to the receptor, and activa- genesis inhibitor sorafenib downregulates the tion of the receptor leads to tyrosine kinase expression of PDGF-BB and TGF-β1 in the HSCs pathway activation, which mediates fibroblast supernatant, and restrains the viability of the activation and is involved in the secretion of a HSCs, resulting in suppressed proliferation variety of cytokines and the formation of cellu- and invasion in HepG2 cells [18]. Meanwhile, lar microenvironment [26]. Activated fibro- Platelet-derived growth factor receptor (PDGFR) blasts, known as cancer-associated fibroblasts signaling participates in different processes in (CAFs), promote the formation of tumor metas- solid tumors, including autocrine stimulation of tasis and directional vessel by secreting SDF-1/ tumor cell growth, recruitment of tumor stroma CXCR4. CAFs also can promote tumor growth fibroblasts, and stimulation of tumor angio- and metastasis by secreting other factors such genesis [19]. Moreover, malignant melanoma as hepatocyte growth factor (HGF), epidermal upregulates hyaluronan synthesis in fibroblasts growth factor (EGF), basic fibroblast growth fac- by releasing PDGF-AA and PDGF-CC, which in tor (b FGF), and insulin-like growth factor (IGF). turn stimulates the malignant melanoma cell PDGF-DD secreted by gastric cancer-derived proliferation in a paracrine manner [20]. PDGF mesenchymal stem cells (GC-MSCs) is capable also increases proliferation of Luminal breast of promoting gastric cancer cell proliferation cancer cells in the absence of estrogens [21]. and migration in vitro and in vivo. Jieqiong Liu et In conclusion, PDGF induces proliferation and al. [27] reported that overexpression of PDGF-D migration of tumor cells and inhibits apoptosis promoted tumor growth and lymph node [21]. metastasis through increased proliferation, decreased apoptosis, and induction of CXCR4 Cancer cells metastasis expression [28]. A study reports a mechanism of the interaction between perivascular cells Cancer cell metastasis begins with detachment and tumor-associated macrophages (TAMs) of metastatic cells from the primary tumor, trav- that promotes metastasis through the IL-33- eling of the cells to different sites through ST2-dependent pathway in xenograft mouse blood/lymphatic vessels, settlement and models of cancer. While PDGF-BB upregulates growth of the cells at a distal site [22]. During IL-33 gene though stimulating the activation of the process, metastatic cells go through pericytes SOX7 transcription factor. Gain- and detachment, migration, invasion, and adhesion loss-of-function experiments validated that [22]. For tumor metastasis, cardiovascular gen- IL-33 promotes metastasis through recruit- eration is required, which provides oxygen and ment of TAMs. Pharmacological inhibition of nutrients for tumor growth and metastasis. the IL-33-ST2 signaling by a soluble ST2 PDGF can promote connective tissue to pro- significantly inhibits TAMs and metastasis [29]. duce extracellular matrix proteins, which direct- Genetic deletion of host IL-33 in mice blocks ly or indirectly induces tumor angiogenesis, PDGF-BB-induced TAM recruitment and metas- significantly affecting tumor metastasis. PDGF- tasis [29]. Yasuhiko Kitadai discovered that D-overexpressing tumors express higher levels expression and phosphorylation of PDGF-Rb by of MMP-9 and PDGFD to induce renal cancer stromal cells and pericytes was higher in ortho- cells proliferation and migration, thus providing topic tumors than in ectopic tumors and, there- a molecular mechanism for the higher inci- fore, was associated with the metastatic poten- dence of lung metastasis and pericyte cover- tial of the neoplasms [30]. PDGFs may also age observed in PDGF-D-overexpressing tu- have a role in determining the preferential mors [23, 24]. PDGFD plays an important role organ of metastatization. Indeed, PDGFs in breast tumor aggressiveness and this pro- released by the tumor cells are potent che- cess is mechanistically linked with the activa- moattractants and mitogens for host mesen- tion of Notch and NF-κB signaling [25]. A grow- chymal cells and could mediate the interac- 9919 Int J Clin Exp Med 2017;10(7):9918-9929 PDGF signaling in cancer progression tions between cancer cells and the host mote the cell recruitment in peripheral blood environment of the preferred metastatic site vessel wall and increase the stability of the [31]. PDGF plays an important role in epithelial structure [38]. The recruitment of peripheral mesenchymal transition. For example, it has blood cells increased blood vessel coverage been recently demonstrated that autocrine which may lead to the resistance to anti-angio- PDGF signaling maintains EMT and promotes genic drugs. In the study of Zhao Z et al., PDGFD metastatization in mouse mammary carcinoma is one of the key candidate genes which may and tumor dissemination [32]. PDGF promotes influence chemo sensitivity of glioblastoma EMT via activation of STAT3 or PI3K pathway (GBM) to Semustine (Me-CCNU) [39]. and PDGF-D over-expression was positively
Recommended publications
  • Role and Regulation of Pdgfra Signaling in Liver Development and Regeneration
    The American Journal of Pathology, Vol. 182, No. 5, May 2013 ajp.amjpathol.org GROWTH FACTORS, CYTOKINES, AND CELL CYCLE MOLECULES Role and Regulation of PDGFRa Signaling in Liver Development and Regeneration Prince K. Awuah,* Kari N. Nejak-Bowen,* and Satdarshan P.S. Monga*y From the Division of Experimental Pathology,* Department of Pathology, and the Department of Medicine,y University of Pittsburgh, Pittsburgh, Pennsylvania Accepted for publication January 22, 2013. Aberrant platelet-derived growth factor receptor-a (PDGFRa) signaling is evident in a subset of hepato- cellular cancers (HCCs). However, its role and regulation in hepatic physiology remains elusive. In the Address correspondence to a fi Satdarshan P.S. Monga, M.D., current study, we examined PDGFR signaling in liver development and regeneration. We identi ed a a Divisions of Experimental notable PDGFR activation in hepatic morphogenesis that, when interrupted by PDGFR -blocking anti- Pathology, Pathology and body, led to decreased hepatoblast proliferation and survival in embryonic liver cultures. We also identified Medicine, University of Pitts- temporal PDGFRa overexpression, which is regulated by epidermal growth factor (EGF) and tumor necrosis burgh School of Medicine, 200 factor a, and its activation at 3 to 24 hours after partial hepatectomy. Through generation of hepatocyte- Lothrop St., S-422 BST, Pitts- specific PDGFRA knockout (KO) mice that lack an overt phenotype, we show that absent PDGFRa burgh, PA 15261. E-mail: compromises extracelluar signal-regulated kinases and AKT activation 3 hours after partial hepatectomy, [email protected]. which, however, is alleviated by temporal compensatory increases in the EGF receptor (EGFR) and the hepatocyte growth factor receptor (Met) expression and activation along with rebound activation of extracellular signal-regulated kinases and AKT at 24 hours.
    [Show full text]
  • Differential Expression of PDGFD in Cancers of the Breast
    Differential expression of platelet-derived growth factor D in cancers of the breast. Shahan Mamoor, MS1 [email protected] East Islip, NY 11730 Breast cancer affects women at relatively high frequency1. We mined published microarray datasets2,3 to determine in an unbiased fashion and at the systems level genes most differentially expressed in the primary tumors of patients with breast cancer. We report here significant differential expression of the gene encoding platelet-derived growth factor D, PDGFD, when comparing primary tumors of the breast to the tissue of origin, the normal breast. PDGFD was also differentially expressed in the tumor cells of patients with triple negative breast cancer. PDGFD mRNA was present at significantly lower quantities in tumors of the breast as compared to normal breast tissue. Analysis of human survival data revealed that expression of PDGFD in primary tumors of the breast was correlated with overall survival in patients with basal and luminal A subtype cancer, demonstrating a relationship between primary tumor expression of a differentially expressed gene and patient survival outcomes influenced by molecular subtype. PDGFD may be of relevance to initiation, maintenance or progression of cancers of the female breast. Keywords: breast cancer, PDGFD, platelet-derived growth factor D, systems biology of breast cancer, targeted therapeutics in breast cancer. 1 Invasive breast cancer is diagnosed in over a quarter of a million women in the United States each year1 and in 2018, breast cancer was the leading cause of cancer death in women worldwide4. While patients with localized breast cancer are provided a 99% 5-year survival rate, patients with regional breast cancer, cancer that has spread to lymph nodes or nearby structures, are provided an 86% 5-year survival rate5,6.
    [Show full text]
  • PDGF-C and PDGF-D Signaling in Vascular Diseases and Animal Models
    Molecular Aspects of Medicine 62 (2018) 1e11 Contents lists available at ScienceDirect Molecular Aspects of Medicine journal homepage: www.elsevier.com/locate/mam PDGF-C and PDGF-D signaling in vascular diseases and animal models * Erika Folestad a, Anne Kunath b, Dick Wågsater€ b, a Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden b Division of Drug Research, Department of Medical and Health Sciences, Linkoping€ University, Linkoping,€ Sweden article info abstract Article history: Members of the platelet-derived growth factor (PDGF) family are well known to be involved in different Received 31 August 2017 pathological conditions. The cellular and molecular mechanisms induced by the PDGF signaling have Received in revised form been well studied. Nevertheless, there is much more to discover about their functions and some 14 November 2017 important questions to be answered. This review summarizes the known roles of two of the PDGFs, Accepted 22 January 2018 PDGF-C and PDGF-D, in vascular diseases. There are clear implications for these growth factors in several Available online 14 February 2018 vascular diseases, such as atherosclerosis and stroke. The PDGF receptors are broadly expressed in the cardiovascular system in cells such as fibroblasts, smooth muscle cells and pericytes. Altered expression Keywords: Aneurysm of the receptors and the ligands have been found in various cardiovascular diseases and current studies fi Atherosclerosis have shown important implications of PDGF-C and PDGF-D signaling in brosis, neovascularization, Growth factor atherosclerosis and restenosis. Myocardial infarction © 2018 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND Smooth muscle cells license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
    [Show full text]
  • Ep 3217179 A1
    (19) TZZ¥ ___T (11) EP 3 217 179 A1 (12) EUROPEAN PATENT APPLICATION (43) Date of publication: (51) Int Cl.: 13.09.2017 Bulletin 2017/37 G01N 33/68 (2006.01) (21) Application number: 17167637.2 (22) Date of filing: 02.10.2013 (84) Designated Contracting States: • LIU, Xinjun AL AT BE BG CH CY CZ DE DK EE ES FI FR GB San Diego, CA 92130 (US) GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO • HAUENSTEIN, Scott PL PT RO RS SE SI SK SM TR San Diego, CA 92130 (US) • KIRKLAND, Richard (30) Priority: 05.10.2012 US 201261710491 P San Diego, CA 92111 (US) 17.05.2013 US 201361824959 P (74) Representative: Krishnan, Sri (62) Document number(s) of the earlier application(s) in Nestec S.A. accordance with Art. 76 EPC: Centre de Recherche Nestlé 13779638.9 / 2 904 405 Vers-chez-les-Blanc Case Postale 44 (71) Applicant: Nestec S.A. 1000 Lausanne 26 (CH) 1800 Vevey (CH) Remarks: (72) Inventors: This application was filed on 21-04-2017 as a • SINGH, Sharat divisional application to the application mentioned Rancho Santa Fe, CA 92127 (US) under INID code 62. (54) METHODS FOR PREDICTING AND MONITORING MUCOSAL HEALING (57) The present invention provides methods for pre- an individual with a disease such as IBD. Information on dicting the likelihood of mucosal healing in an individual mucosal healing status derived from the use of the with a disease such as inflammatory bowel disease present invention can also aid in optimizing therapy (IBD).
    [Show full text]
  • Regulation of PDGFC Signalling and Extracellular Matrix Composition by FREM1 in Mice
    RESEARCH ARTICLE Disease Models & Mechanisms 6, 1426-1433 (2013) doi:10.1242/dmm.013748 Regulation of PDGFC signalling and extracellular matrix composition by FREM1 in mice Fenny Wiradjaja1, Denny L. Cottle1, Lynelle Jones1 and Ian Smyth1,2,* SUMMARY Fras1-related extracellular matrix protein 1 (FREM1) is required for epidermal adhesion during embryogenesis, and mice lacking the gene develop fetal skin blisters and a range of other developmental defects. Mutations in members of the FRAS/FREM gene family cause diseases of the Fraser syndrome spectrum. Embryonic epidermal blistering is also observed in mice lacking PdgfC and its receptor, PDGFRα. In this article, we show that FREM1 binds to PDGFC and that this interaction regulates signalling downstream of PDGFRα. Fibroblasts from Frem1-mutant mice respond to PDGFC stimulation, but with a shorter duration and amplitude than do wild-type cells. Significantly, PDGFC-stimulated expression of the metalloproteinase inhibitor Timp1 is reduced in cells with Frem1 mutations, leading to reduced basement membrane collagen I deposition. These results show that the physical interaction of FREM1 with PDGFC can regulate remodelling of the extracellular matrix downstream of PDGFRα. We propose that loss of FREM1 function promotes epidermal blistering in Fraser syndrome as a consequence of reduced PDGFC activity, in addition to its stabilising role in the basement membrane. DMM INTRODUCTION The FRAS/FREM family of proteins share characteristic The FRAS/FREM extracellular matrix (ECM) proteins (FRAS1, chondroitin sulphate proteoglycan (CSPG) core repeats similar FREM1 and FREM2) mediate adhesion between the epidermal to those found in the NG2 proteoglycan (Stallcup, 2002). In this basement membrane and the underlying dermis during embryonic protein, they directly bind platelet-derived growth factor A development (reviewed in Short et al., 2007; Petrou et al., 2008).
    [Show full text]
  • PDGFC (Human) Recombinant Protein
    PDGFC (Human) Recombinant pH 3.0. protein Storage Instruction: Store at -20°C. Reconstitute in water to a concentration of 0.1-1.0 Catalog Number: P6130 mg/mL. Do not vortex. Regulation Status: For research use only (RUO) For extended storage, it is recommended to further dilute in a buffer containing a carrier protein (example 0.1% Product Description: Human PDGFC (Q9NRA1) partial BSA) and store in working aliquots at -20°C to -80°C. recombinant protein expressed in Escherichia coli. Entrez GeneID: 56034 Sequence: MVVDLNLLTEEVRLYSCTPRNFSVSIREELKRTDTIFW Gene Symbol: PDGFC PGCLLVKRCGGNCACCLHNCNECQCVPSKVTKKYHE Gene Alias: FALLOTEIN, SCDGF VLQLRPKTGVRGLHKSLTDVALEHHEECDCVCRGSTG G Gene Summary: The protein encoded by this gene is a member of the platelet-derived growth factor family. The Host: Escherichia coli four members of this family are mitogenic factors for Theoretical MW (kDa): 25.0 cells of mesenchymal origin and are characterized by a core motif of eight cysteines. This gene product appears Reactivity: Human to form only homodimers. It differs from the platelet-derived growth factor alpha and beta Applications: Func, SDS-PAGE polypeptides in having an unusual N-terminal domain, (See our web site product page for detailed applications the CUB domain. [provided by RefSeq] information) Protocols: See our web site at http://www.abnova.com/support/protocols.asp or product page for detailed protocols Form: Lyophilized Preparation Method: Escherichia coli expression system Purity: 98% Endotoxin Level: Endotoxin level is <0.1 ng/ug of protein (<1 EU/ug). Activity: Determined by the dose-dependent stimulation of the proliferation of Balb/c 3T3 cells. The expected The ED50 for this effect is 15-20 ng/mL.
    [Show full text]
  • WO 2014/054013 Al 10 April 2014 (10.04.2014) P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2014/054013 Al 10 April 2014 (10.04.2014) P O P C T (51) International Patent Classification: (81) Designated States (unless otherwise indicated, for every G01N 33/68 (2006.01) kind of national protection available): AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, (21) International Application Number: BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, PCT/IB2013/059077 DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, (22) International Filing Date: HN, HR, HU, ID, IL, IN, IS, JP, KE, KG, KN, KP, KR, 2 October 2013 (02. 10.2013) KZ, LA, LC, LK, LR, LS, LT, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, (25) Filing Language: English OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, (26) Publication Language: English SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, (30) Priority Data: ZW. 61/710,491 5 October 2012 (05. 10.2012) 61/824,959 17 May 2013 (17.05.2013) (84) Designated States (unless otherwise indicated, for every kind of regional protection available): ARIPO (BW, GH, (71) Applicant: NESTEC S.A. [CH/CH]; Ave. Nestle 55, CH- GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, SZ, TZ, 1800 Vevey (CH).
    [Show full text]
  • Identification of Shared and Unique Gene Families Associated with Oral
    International Journal of Oral Science (2017) 9, 104–109 OPEN www.nature.com/ijos ORIGINAL ARTICLE Identification of shared and unique gene families associated with oral clefts Noriko Funato and Masataka Nakamura Oral clefts, the most frequent congenital birth defects in humans, are multifactorial disorders caused by genetic and environmental factors. Epidemiological studies point to different etiologies underlying the oral cleft phenotypes, cleft lip (CL), CL and/or palate (CL/P) and cleft palate (CP). More than 350 genes have syndromic and/or nonsyndromic oral cleft associations in humans. Although genes related to genetic disorders associated with oral cleft phenotypes are known, a gap between detecting these associations and interpretation of their biological importance has remained. Here, using a gene ontology analysis approach, we grouped these candidate genes on the basis of different functional categories to gain insight into the genetic etiology of oral clefts. We identified different genetic profiles and found correlations between the functions of gene products and oral cleft phenotypes. Our results indicate inherent differences in the genetic etiologies that underlie oral cleft phenotypes and support epidemiological evidence that genes associated with CL/P are both developmentally and genetically different from CP only, incomplete CP, and submucous CP. The epidemiological differences among cleft phenotypes may reflect differences in the underlying genetic causes. Understanding the different causative etiologies of oral clefts is
    [Show full text]
  • Xo PANEL DNA GENE LIST
    xO PANEL DNA GENE LIST ~1700 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes (at 400 -500x average coverage on tumor) Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11
    [Show full text]
  • PDGFC Antibody Cat
    PDGFC Antibody Cat. No.: 59-045 PDGFC Antibody PDGFC antibody immunohistochemistry analysis in formalin fixed and paraffin embedded human kidney tissue followed by peroxidase conjugation of the secondary antibody and DAB staining. Specifications HOST SPECIES: Rabbit SPECIES REACTIVITY: Human HOMOLOGY: Predicted species reactivity based on immunogen sequence: Chicken, Mouse This PDGFC antibody is generated from rabbits immunized with a KLH conjugated IMMUNOGEN: synthetic peptide between 74-103 amino acids from the N-terminal region of human PDGFC. TESTED APPLICATIONS: IHC-P, WB For WB starting dilution is: 1:1000 APPLICATIONS: For IHC-P starting dilution is: 1:10~50 September 28, 2021 1 https://www.prosci-inc.com/pdgfc-antibody-59-045.html PREDICTED MOLECULAR 39 kDa WEIGHT: Properties This antibody is prepared by Saturated Ammonium Sulfate (SAS) precipitation followed by PURIFICATION: dialysis CLONALITY: Polyclonal ISOTYPE: Rabbit Ig CONJUGATE: Unconjugated PHYSICAL STATE: Liquid BUFFER: Supplied in PBS with 0.09% (W/V) sodium azide. CONCENTRATION: batch dependent Store at 4˚C for three months and -20˚C, stable for up to one year. As with all antibodies STORAGE CONDITIONS: care should be taken to avoid repeated freeze thaw cycles. Antibodies should not be exposed to prolonged high temperatures. Additional Info OFFICIAL SYMBOL: PDGFC Platelet-derived growth factor C, PDGF-C, Fallotein, Spinal cord-derived growth factor, SCDGF, VEGF-E, Platelet-derived growth factor C, latent form, PDGFC latent form, Platelet- ALTERNATE NAMES: derived growth factor C, receptor-binding form, PDGFC receptor-binding form, PDGFC, SCDGF ACCESSION NO.: Q9NRA1 PROTEIN GI NO.: 205830662 GENE ID: 56034 USER NOTE: Optimal dilutions for each application to be determined by the researcher.
    [Show full text]
  • Differences in Gene Expression Profiles and Carcinogenesis Pathways Involved in Cisplatin Resistance of Four Types of Cancer
    596 ONCOLOGY REPORTS 30: 596-614, 2013 Differences in gene expression profiles and carcinogenesis pathways involved in cisplatin resistance of four types of cancer YONG YANG1,2, HUI LI1,2, SHENGCAI HOU1,2, BIN HU1,2, JIE LIU1,3 and JUN WANG1,3 1Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Capital Medical University, Beijing 100069; 2Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020; 3Department of Physiology, Capital Medical University, Beijing 100069, P.R. China Received December 23, 2012; Accepted March 4, 2013 DOI: 10.3892/or.2013.2514 Abstract. Cisplatin-based chemotherapy is the standard Introduction therapy used for the treatment of several types of cancer. However, its efficacy is largely limited by the acquired drug Cisplatin is primarily effective through DNA damage and is resistance. To date, little is known about the RNA expression widely used for the treatment of several types of cancer, such changes in cisplatin-resistant cancers. Identification of the as testicular, lung and ovarian cancer. However, the ability RNAs related to cisplatin resistance may provide specific of cancer cells to become resistant to cisplatin remains a insight into cancer therapy. In the present study, expression significant impediment to successful chemotherapy. Although profiling of 7 cancer cell lines was performed using oligo- previous studies have identified numerous mechanisms in nucleotide microarray analysis data obtained from the GEO cisplatin resistance, it remains a major problem that severely database. Bioinformatic analyses such as the Gene Ontology limits the usefulness of this chemotherapeutic agent. Therefore, (GO) and KEGG pathway were used to identify genes and it is crucial to examine more elaborate mechanisms of cisplatin pathways specifically associated with cisplatin resistance.
    [Show full text]
  • Release of Stem Cells from Quiescence Reveals Multiple Gliogenic Domains in the Adult Brain
    bioRxiv preprint doi: https://doi.org/10.1101/738013; this version posted August 16, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Release of stem cells from quiescence reveals multiple gliogenic domains in the adult brain Ana C. Delgado1*, Angel R. Maldonado-Soto2*, Violeta Silva-Vargas1, Dogukan Mizrak3,4, Thomas von Känel1, Alex Paul5,9, Aviv Madar7, Henar Cuervo8, Jan Kitajewski8, Chyuan- Sheng Lin3,6 and Fiona Doetsch1 # 1Biozentrum, University of Basel, Basel, Switzerland, 2Departments of Neurology, 3Pathology and Cell Biology, 4Systems Biology, 5Genetics and Development, 6Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, 7Department of Biology, Center for Genomics and Systems Biology, New York University, 8Department of Physiology and Biophysics, University of Illinois at Chicago. 9Current Address: Laboratory of Social Evolution and Behavior, Rockefeller University, NYC, USA *equal contribution #Corresponding author Fiona Doetsch Biozentrum, University of Basel Klingelbergstrasse 50/70 CH 4056 Basel Switzerland Telephone: +41 61 267 22 30 Email: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/738013; this version posted August 16, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract Quiescent neural stem cells (NSCs) in the adult ventricular-subventricular zone (V- SVZ) have a regional identity and undergo activation to generate neurons. The domains for gliogenesis are less explored. Here we show that Platelet-Derived Growth Factor Receptor beta (PDGFRβ) is expressed by adult V-SVZ NSCs that generate olfactory bulb interneurons and glia with slow baseline kinetics.
    [Show full text]