1
|
Zhang L, Liu X, Liu J, Zhou Z, Song Y, Cao B, An X. miR-182 aids in receptive endometrium development in dairy goats by down-regulating PTN expression. PLoS One 2017; 12:e0179783. [PMID: 28678802 PMCID: PMC5497977 DOI: 10.1371/journal.pone.0179783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/04/2017] [Indexed: 02/04/2023] Open
Abstract
Increasing evidence has shown that miRNAs play important roles in endometrium development during the menstrual cycle in humans and many other animals. Our previous data indicated that miR-182 levels increase 15.55-fold and pleiotrophin (PTN) levels decrease 20.97-fold in the receptive endometrium (RE, D15) compared with the pre-receptive endometrium (PE, D5) in dairy goats. The present study shows that miR-182 is widely expressed in different tissues of dairy goats and that its expression levels are regulated by E2 and P4 in endometrial epithelium cells (EECs). We confirmed that PTN is a target of miR-182 and that miR-182 regulates the protein levels of AKT, Bcl-2, FAS, MAPK, Caspase-3 and SP1 in EECs. Furthermore, miR-182 up-regulates or maintains the expression levels of osteopontin (OPN), cyclooxygenase-2 (COX-2) and prolactin receptor (PRLR) in EECs, suggesting that miR-182 is an important regulatory factor in the construction of endometrial receptivity in dairy goats. In conclusion, miR-182 participates in the development of endometrial receptivity by down-regulating PTN and affecting the expression of select apoptosis-related genes and increasing or maintaining the expression levels of OPN, COX-2 and PRLR in the EECs of dairy goats.
Collapse
Affiliation(s)
- Lei Zhang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Xiaorui Liu
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Junze Liu
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Zhanqin Zhou
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Binyun Cao
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| |
Collapse
|
2
|
Michelotti GA, Tucker A, Swiderska-Syn M, Machado MV, Choi SS, Kruger L, Soderblom E, Thompson JW, Mayer-Salman M, Himburg HA, Moylan CA, Guy CD, Garman KS, Premont RT, Chute JP, Diehl AM. Pleiotrophin regulates the ductular reaction by controlling the migration of cells in liver progenitor niches. Gut 2016; 65:683-92. [PMID: 25596181 PMCID: PMC4504836 DOI: 10.1136/gutjnl-2014-308176] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/22/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The ductular reaction (DR) involves mobilisation of reactive-appearing duct-like cells (RDC) along canals of Hering, and myofibroblastic (MF) differentiation of hepatic stellate cells (HSC) in the space of Disse. Perivascular cells in stem cell niches produce pleiotrophin (PTN) to inactivate the PTN receptor, protein tyrosine phosphatase receptor zeta-1 (PTPRZ1), thereby augmenting phosphoprotein-dependent signalling. We hypothesised that the DR is regulated by PTN/PTPRZ1 signalling. DESIGN PTN-GFP, PTN-knockout (KO), PTPRZ1-KO, and wild type (WT) mice were examined before and after bile duct ligation (BDL) for PTN, PTPRZ1 and the DR. RDC and HSC from WT, PTN-KO, and PTPRZ1-KO mice were also treated with PTN to determine effects on downstream signaling phosphoproteins, gene expression, growth, and migration. Liver biopsies from patients with DRs were also interrogated. RESULTS Although quiescent HSC and RDC lines expressed PTN and PTPRZ1 mRNAs, neither PTN nor PTPRZ1 protein was demonstrated in healthy liver. BDL induced PTN in MF-HSC and increased PTPRZ1 in MF-HSC and RDC. In WT mice, BDL triggered a DR characterised by periportal accumulation of collagen, RDC and MF-HSC. All aspects of this DR were increased in PTN-KO mice and suppressed in PTPRZ1-KO mice. In vitro studies revealed PTN-dependent accumulation of phosphoproteins that control cell-cell adhesion and migration, with resultant inhibition of cell migration. PTPRZ1-positive cells were prominent in the DRs of patients with ductal plate defects and adult cholestatic diseases. CONCLUSIONS PTN, and its receptor, PTPRZ1, regulate the DR to liver injury by controlling the migration of resident cells in adult liver progenitor niches.
Collapse
Affiliation(s)
| | - Anikia Tucker
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | | | | | - Steve S Choi
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Leandi Kruger
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | - Erik Soderblom
- Proteomics Center, Duke University, Durham, North Carolina, USA
| | - J Will Thompson
- Proteomics Center, Duke University, Durham, North Carolina, USA
| | | | - Heather A Himburg
- Division of Hematology and Oncology, UCLA, Los Angeles, California, USA
| | - Cynthia A Moylan
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Cynthia D Guy
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Katherine S Garman
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA Section of Gastroenterology, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Richard T Premont
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | - John P Chute
- Division of Hematology and Oncology, UCLA, Los Angeles, California, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
3
|
Hyperactivation of Akt/mTOR and deficiency in tuberin increased the oxidative DNA damage in kidney cancer patients with diabetes. Oncotarget 2015; 5:2542-50. [PMID: 24797175 PMCID: PMC4058025 DOI: 10.18632/oncotarget.1833] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recent study from our laboratory showed that patients with diabetes are at a higher risk of developing kidney cancer. In the current study, we have explored one of the mechanisms by which diabetes accelerates tumorigenesis in the kidney. Kidney cancer tissue from patients with diabetes showed a higher activity of Akt and decreased in total protein of tuberin compared to kidney cancer patient without diabetes or diabetes alone. In addition, a significant increase in phospho-Akt/tuberin expression was associated with an increase in Ki67 expression and activation of mTOR in kidney tumor with or without diabetes compared to diabetes alone. In addition, decrease in tuberin expression resulted in a significant decrease in protein expression of OGG1 and increased in oxidative DNA damage, 8-oxodG in kidney tissues from patients with cancer or cancer+diabetes. Importantly, these data showed that the majority of the staining of Akt/tuberin/p70S6K phosphorylation was more prominently in the tubular cells. In addition, accumulation of oxidative DNA damage is localized only in the nucleus of tubular cells within the cortex region. These data suggest that Akt/tuberin/mTOR pathway plays an important role in the regulation DNA damage and repair pathways that may predispose diabetic kidneys to pathogenesis of renal cell carcinoma.
Collapse
|
4
|
Sethi G, Kwon Y, Burkhalter RJ, Pathak HB, Madan R, McHugh S, Atay S, Murthy S, Tawfik OW, Godwin AK. PTN signaling: Components and mechanistic insights in human ovarian cancer. Mol Carcinog 2014; 54:1772-85. [PMID: 25418856 DOI: 10.1002/mc.22249] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/30/2014] [Accepted: 10/10/2014] [Indexed: 12/13/2022]
Abstract
Molecular vulnerabilities represent promising candidates for the development of targeted therapies that hold the promise to overcome the challenges encountered with non-targeted chemotherapy for the treatment of ovarian cancer. Through a synthetic lethality screen, we previously identified pleiotrophin (PTN) as a molecular vulnerability in ovarian cancer and showed that siRNA-mediated PTN knockdown induced apoptotic cell death in epithelial ovarian cancer (EOC) cells. Although, it is well known that PTN elicits its pro-tumorigenic effects through its receptor, protein tyrosine phosphatase receptor Z1 (PTPRZ1), little is known about the potential importance of this pathway in the pathogenesis of ovarian cancer. In this study, we show that PTN is expressed, produced, and secreted in a panel of EOC cell lines. PTN levels in serous ovarian tumor tissues are on average 3.5-fold higher relative to normal tissue and PTN is detectable in serum samples of patients with EOC. PTPRZ1 is also expressed and produced by EOC cells and is found to be up-regulated in serous ovarian tumor tissue relative to normal ovarian surface epithelial tissue (P < 0.05). Gene silencing of PTPRZ1 in EOC cell lines using siRNA-mediated knockdown shows that PTPRZ1 is essential for viability and results in significant apoptosis with no effect on the cell cycle phase distribution. In order to determine how PTN mediates survival, we silenced the gene using siRNA mediated knockdown and performed expression profiling of 36 survival-related genes. Through computational mapping of the differentially expressed genes, members of the MAPK (mitogen-activated protein kinase) family were found to be likely effectors of PTN signaling in EOC cells. Our results provide the first experimental evidence that PTN and its signaling components may be of significance in the pathogenesis of epithelial ovarian cancer and provide a rationale for clinical evaluation of MAPK inhibitors in PTN and/or PTPRZ1 expressing ovarian tumors.
Collapse
Affiliation(s)
- Geetika Sethi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Biochemistry, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Youngjoo Kwon
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Rebecca J Burkhalter
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Harsh B Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sarah McHugh
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Safinur Atay
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Smruthi Murthy
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Ossama W Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,University of Kansas Cancer Center, Kansas City, Kansas
| |
Collapse
|
5
|
Choi WJ, Kim M, Park JY, Park TJ, Kang HY. Pleiotrophin inhibits melanogenesisviaErk1/2-MITF signaling in normal human melanocytes. Pigment Cell Melanoma Res 2014; 28:51-60. [DOI: 10.1111/pcmr.12309] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/16/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Woo Jong Choi
- Department of Dermatology; Ajou University School of Medicine; Suwon Korea
- Department of Biochemistry and Molecular Biology; Ajou University School of Medicine; Suwon Korea
| | - Misun Kim
- Department of Dermatology; Ajou University School of Medicine; Suwon Korea
| | - Ji-Youn Park
- Department of Dermatology; Ajou University School of Medicine; Suwon Korea
| | - Tae Jun Park
- Department of Biochemistry and Molecular Biology; Ajou University School of Medicine; Suwon Korea
| | - Hee Young Kang
- Department of Dermatology; Ajou University School of Medicine; Suwon Korea
| |
Collapse
|
6
|
Wellstein A. ALK receptor activation, ligands and therapeutic targeting in glioblastoma and in other cancers. Front Oncol 2012; 2:192. [PMID: 23267434 PMCID: PMC3525999 DOI: 10.3389/fonc.2012.00192] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 11/27/2012] [Indexed: 11/13/2022] Open
Abstract
The intracellular anaplastic lymphoma kinase (ALK) fragment shows striking homology with members of the insulin receptor family and was initially identified as an oncogenic fusion protein resulting from a translocation in lymphoma and more recently in a range of cancers. The full-length ALK transmembrane receptor of ~220 kDa was identified based on this initial work. This tyrosine kinase receptor and its ligands, the growth factors pleiotrophin (PTN) and midkine (MK) are highly expressed during development of the nervous system and other organs. Each of these genes has been implicated in malignant progression of different tumor types and shown to alter phenotypes as well as signal transduction in cultured normal and tumor cells. Beyond its role in cancer, the ALK receptor pathway is thought to contribute to nervous system development, function, and repair, as well as metabolic homeostasis and the maintenance of tissue regeneration. ALK receptor activity in cancer can be up-regulated by amplification, overexpression, ligand binding, mutations in the intracellular domain of the receptor and by activity of the receptor tyrosine phosphatase PTPRz. Here we discuss the evidence for ligand control of ALK activity as well as the potential prognostic and therapeutic implications from gene expression and functional studies. An analysis of 18 published gene expression data sets from different cancers shows that overexpression of ALK, its smaller homolog LTK (leukocyte tyrosine kinase) and the ligands PTN and MK in cancer tissues from patients correlate significantly with worse course and outcome of the disease. This observation together with preclinical functional studies suggests that this pathway could be a valid therapeutic target for which complementary targeting strategies with small molecule kinase inhibitors as well as antibodies to ligands or the receptors may be used.
Collapse
Affiliation(s)
- Anton Wellstein
- Lombardi Cancer Center, Georgetown UniversityWashington, DC, USA
| |
Collapse
|
7
|
Rosenfield SM, Bowden ET, Cohen-Missner S, Gibby KA, Ory V, Henke RT, Riegel AT, Wellstein A. Pleiotrophin (PTN) expression and function and in the mouse mammary gland and mammary epithelial cells. PLoS One 2012; 7:e47876. [PMID: 23077670 PMCID: PMC3471873 DOI: 10.1371/journal.pone.0047876] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 09/24/2012] [Indexed: 11/19/2022] Open
Abstract
Expression of the heparin-binding growth factor, pleiotrophin (PTN) in the mammary gland has been reported but its function during mammary gland development is not known. We examined the expression of PTN and its receptor ALK (Anaplastic Lymphoma Kinase) at various stages of mouse mammary gland development and found that their expression in epithelial cells is regulated in parallel during pregnancy. A 30-fold downregulation of PTN mRNA expression was observed during mid-pregnancy when the mammary gland undergoes lobular-alveolar differentiation. After weaning of pups, PTN expression was restored although baseline expression of PTN was reduced significantly in mammary glands of mice that had undergone multiple pregnancies. We found PTN expressed in epithelial cells of the mammary gland and thus used a monoclonal anti-PTN blocking antibody to elucidate its function in cultured mammary epithelial cells (MECs) as well as during gland development. Real-time impedance monitoring of MECs growth, migration and invasion during anti-PTN blocking antibody treatment showed that MECs motility and invasion but not proliferation depend on the activity of endogenous PTN. Increased number of mammospheres with laminin deposition after anti-PTN blocking antibody treatment of MECs in 3D culture and expression of progenitor markers suggest that the endogenously expressed PTN inhibits the expansion and differentiation of epithelial progenitor cells by disrupting cell-matrix adhesion. In vivo, PTN activity was found to inhibit ductal outgrowth and branching via the inhibition of phospho ERK1/2 signaling in the mammary epithelial cells. We conclude that PTN delays the maturation of the mammary gland by maintaining mammary epithelial cells in a progenitor phenotype and by inhibiting their differentiation during mammary gland development.
Collapse
Affiliation(s)
- Sonia M. Rosenfield
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Emma T. Bowden
- MedImmune, Gaithersburg, Maryland, United States of America
| | - Shani Cohen-Missner
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Krissa A. Gibby
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Virginie Ory
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Ralf T. Henke
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Anna T. Riegel
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
| | - Anton Wellstein
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, United States of America
- * E-mail:
| |
Collapse
|
8
|
Koutsioumpa M, Drosou G, Mikelis C, Theochari K, Vourtsis D, Katsoris P, Giannopoulou E, Courty J, Petrou C, Magafa V, Cordopatis P, Papadimitriou E. Pleiotrophin expression and role in physiological angiogenesis in vivo: potential involvement of nucleolin. Vasc Cell 2012; 4:4. [PMID: 22423616 PMCID: PMC3379939 DOI: 10.1186/2045-824x-4-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 03/16/2012] [Indexed: 11/20/2022] Open
Abstract
Background Pleiotrophin (PTN) is a heparin-binding growth factor with significant role(s) in tumour growth and angiogenesis. Although implication of endogenous PTN has been studied in several in vivo models of tumour angiogenesis, its role in physiological angiogenesis has not been addressed. In the present work, we studied expression and functional significance of endogenous PTN during angiogenesis in the chicken embryo chorioallantoic membrane (CAM). Methods Using molecular, cellular and biochemical assays, we studied the expression pattern of PTN in CAM and human endothelial cells and its possible interaction with nucleolin (NCL). CAM cells were transfected with a pCDNA3.1 vector, empty (PC) or containing full length cDNA for PTN in antisense orientation (AS-PTN). Angiogenesis was estimated by measuring total vessel length. In vitro, human endothelial cells migration was studied by using a transwell assay, and down-regulation of NCL was performed by using a proper siRNA. Results Endogenous PTN mRNA and protein levels, as well as protein levels of its receptor protein tyrosine phosphatase beta/zeta (RPTPβ/ζ) were maximal at early stages, when CAM angiogenesis is active. Application of AS-PTN onto CAM at days of active angiogenesis was not toxic to the tissue and led to dose-dependent decreased expression of endogenous PTN, ERK1/2 activity and angiogenesis. Interestingly, endogenous PTN was also immunolocalized at the endothelial cell nucleus, possibly through interaction with NCL, a protein that has a significant role in the nuclear translocation of many proteins. Down-regulation of NCL by siRNA in human endothelial cells significantly decreased nuclear PTN, verifying this hypothesis. Moreover, it led to abolishment of PTN-induced endothelial cell migration, suggesting, for the first time, that PTN-NCL interaction has a functional significance. Conclusions Expression of endogenous PTN correlates with and seems to be involved in angiogenesis of the chicken embryo CAM. Our data suggest that NCL may have a role, increasing the number of growth factors whose angiogenic/tumorigenic activities are mediated by NCL.
Collapse
Affiliation(s)
- Marina Koutsioumpa
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Georgia Drosou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Constantinos Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece.,Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Building 30, Room 203, Bethesda, MD 20892-4340, USA
| | - Katerina Theochari
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Dionussios Vourtsis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| | - Panagiotis Katsoris
- Division of Genetics, Cell & Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Efstathia Giannopoulou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece.,Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University Hospital of Patras, Patras Medical School, 26504 Rio, Greece
| | - Jose Courty
- Laboratoire CRRET, Université Paris Est Créteil, Val de Marne, 61 avenue du Général de Gaulle, 94010 Créteil, Cedex, France
| | - Christos Petrou
- Laboratory of Pharmacognocy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Athens, Greece
| | - Vassiliki Magafa
- Laboratory of Pharmacognocy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Athens, Greece
| | - Paul Cordopatis
- Laboratory of Pharmacognocy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Athens, Greece
| | - Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras GR 26504, Greece
| |
Collapse
|
9
|
Hamma-Kourbali Y, Bermek O, Bernard-Pierrot I, Karaky R, Martel-Renoir D, Frechault S, Courty J, Delbé J. The synthetic peptide P111-136 derived from the C-terminal domain of heparin affin regulatory peptide inhibits tumour growth of prostate cancer PC-3 cells. BMC Cancer 2011; 11:212. [PMID: 21624116 PMCID: PMC3118947 DOI: 10.1186/1471-2407-11-212] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 05/30/2011] [Indexed: 01/19/2023] Open
Abstract
Background Heparin affin regulatory peptide (HARP), also called pleiotrophin, is a heparin-binding, secreted factor that is overexpressed in several tumours and associated to tumour growth, angiogenesis and metastasis. The C-terminus part of HARP composed of amino acids 111 to 136 is particularly involved in its biological activities and we previously established that a synthetic peptide composed of the same amino acids (P111-136) was capable of inhibiting the biological activities of HARP. Here we evaluate the ability of P111-136 to inhibit in vitro and in vivo the growth of a human tumour cell line PC-3 which possess an HARP autocrine loop. Methods A total lysate of PC-3 cells was incubated with biotinylated P111-136 and pulled down for the presence of the HARP receptors in Western blot. In vitro, the P111-136 effect on HARP autocrine loop in PC-3 cells was determined by colony formation in soft agar. In vivo, PC-3 cells were inoculated in the flank of athymic nude mice. Animals were treated with P111-136 (5 mg/kg/day) for 25 days. Tumour volume was evaluated during the treatment. After the animal sacrifice, the tumour apoptosis and associated angiogenesis were evaluated by immunohistochemistry. In vivo anti-angiogenic effect was confirmed using a mouse Matrigel™ plug assay. Results Using pull down experiments, we identified the HARP receptors RPTPβ/ζ, ALK and nucleolin as P111-136 binding proteins. In vitro, P111-136 inhibits dose-dependently PC-3 cell colony formation. Treatment with P111-136 inhibits significantly the PC-3 tumour growth in the xenograft model as well as tumour angiogenesis. The angiostatic effect of P111-136 on HARP was also confirmed using an in vivo Matrigel™ plug assay in mice Conclusions Our results demonstrate that P111-136 strongly inhibits the mitogenic effect of HARP on in vitro and in vivo growth of PC-3 cells. This inhibition could be linked to a direct or indirect binding of this peptide to the HARP receptors (ALK, RPTPβ/ζ, nucleolin). In vivo, the P111-136 treatment significantly inhibits both the PC-3 tumour growth and the associated angiogenesis. Thus, P111-136 may be considered as an interesting pharmacological tool to interfere with tumour growth that has now to be evaluated in other cancer types.
Collapse
Affiliation(s)
- Yamina Hamma-Kourbali
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires, Université Paris Est Créteil, CNRS, avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Himburg HA, Muramoto GG, Daher P, Meadows SK, Russell JL, Doan P, Chi JT, Salter AB, Lento WE, Reya T, Chao NJ, Chute JP. Pleiotrophin regulates the expansion and regeneration of hematopoietic stem cells. Nat Med 2010; 16:475-82. [PMID: 20305662 DOI: 10.1038/nm.2119] [Citation(s) in RCA: 219] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 02/11/2010] [Indexed: 11/09/2022]
Abstract
Hematopoietic stem cell (HSC) self-renewal is regulated by both intrinsic and extrinsic signals. Although some of the pathways that regulate HSC self-renewal have been uncovered, it remains largely unknown whether these pathways can be triggered by deliverable growth factors to induce HSC growth or regeneration. Here we show that pleiotrophin, a neurite outgrowth factor with no known function in hematopoiesis, efficiently promotes HSC expansion in vitro and HSC regeneration in vivo. Treatment of mouse bone marrow HSCs with pleiotrophin caused a marked increase in long-term repopulating HSC numbers in culture, as measured in competitive repopulating assays. Treatment of human cord blood CD34(+)CDCD38(-)Lin(-) cells with pleiotrophin also substantially increased severe combined immunodeficient (SCID)-repopulating cell counts in culture, compared to input and cytokine-treated cultures. Systemic administration of pleiotrophin to irradiated mice caused a pronounced expansion of bone marrow stem and progenitor cells in vivo, indicating that pleiotrophin is a regenerative growth factor for HSCs. Mechanistically, pleiotrophin activated phosphoinositide 3-kinase (PI3K) signaling in HSCs; antagonism of PI3K or Notch signaling inhibited pleiotrophin-mediated expansion of HSCs in culture. We identify the secreted growth factor pleiotrophin as a new regulator of both HSC expansion and regeneration.
Collapse
Affiliation(s)
- Heather A Himburg
- Division of Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Saravanamuthu SS, Gao CY, Zelenka PS. Notch signaling is required for lateral induction of Jagged1 during FGF-induced lens fiber differentiation. Dev Biol 2009; 332:166-76. [PMID: 19481073 DOI: 10.1016/j.ydbio.2009.05.566] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/19/2009] [Accepted: 05/20/2009] [Indexed: 01/08/2023]
Abstract
Previous studies of the developing lens have shown that Notch signaling regulates differentiation of lens fiber cells by maintaining a proliferating precursor pool in the anterior epithelium. However, whether Notch signaling is further required after the onset of fiber cell differentiation is not clear. This work investigates the role of Notch2 and Jagged1 (Jag1) in secondary fiber cell differentiation using rat lens epithelial explants undergoing FGF-2 dependent differentiation in vitro. FGF induced Jag1 expression and Notch2 signaling (as judged by the appearance of activated Notch2 Intracellular Domain (N2ICD)) within 12-24 h. These changes were correlated with induction of the Notch effector, Hes5, upregulation of N-cadherin (N-cad), and downregulation of E-cadherin (E-cad), a cadherin switch characteristic of fiber cell differentiation. Induction of Jag1 was efficiently blocked by U0126, a specific inhibitor of MAPK/ERK signaling, indicating a requirement for signaling through this pathway downstream of the FGF receptor. Other growth factors that activate MAPK/ERK signaling (EGF, PDGF, IGF) did not induce Jag1. Inhibition of Notch signaling using gamma secretase inhibitors DAPT and L-685,458 or anti-Jag1 antibody markedly decreased FGF-dependent expression of Jag1 demonstrating Notch-dependent lateral induction. In addition, inhibition of Notch signaling reduced expression of N-cad, and the cyclin dependent kinase inhibitor, p57Kip2, indicating a direct role for Notch signaling in secondary fiber cell differentiation. These results demonstrate that Notch-mediated lateral induction of Jag1 is an essential component of FGF-dependent lens fiber cell differentiation.
Collapse
Affiliation(s)
- Senthil S Saravanamuthu
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
12
|
Albini A, Mirisola V, Pfeffer U. Metastasis signatures: genes regulating tumor-microenvironment interactions predict metastatic behavior. Cancer Metastasis Rev 2008; 27:75-83. [PMID: 18046511 DOI: 10.1007/s10555-007-9111-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The possibility of predicting clinical outcome of cancer patients through the analysis of gene expression profiles in the primary tumor is a kind of ideological revolution as the multistep carcinogenesis model postulates that the proportion of cells within the primary tumor that actually acquire metastasis driving mutation(s) is small; too small to leave its imprint on the gene expression profile. The data collected to date have brought a new paradigm to reality in the metastasis field: metastasis must at least in part rely on mutations and/or gene regulation events present in the majority of cells which constitute the primary tumor mass. By analyses of differential expression of primary tumors versus metastases or by functional analyses of putative metastasis genes in experimental metastasis, many metastasis-associated gene expression events have been identified that correlate with the development of metastases. Among genes "favoring" metastasis, we find many molecules that are expressed not by the tumor cell itself but by the cells of the microenvironment, as well as genes over-expressed in the primary tumor that have a principle role in mediating tumor-host interactions. Here we review these concepts and advance hypotheses on how gene expression of the primary tumor and the microenvironment can favor the spread of the metastasis seeds and how this knowledge can provide tools to secondary prevention.
Collapse
Affiliation(s)
- Adriana Albini
- IRCCS MultiMedica, Scientific and Technological Pole, Via Fantoli 16/15, 20138, Milan, Italy.
| | | | | |
Collapse
|
13
|
Li J, Wei H, Chesley A, Moon C, Krawczyk M, Volkova M, Ziman B, Margulies KB, Talan M, Crow MT, Boheler KR. The Pro-angiogenic Cytokine Pleiotrophin Potentiates Cardiomyocyte Apoptosis through Inhibition of Endogenous AKT/PKB Activity. J Biol Chem 2007; 282:34984-93. [DOI: 10.1074/jbc.m703513200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
14
|
Mathivet T, Mazot P, Vigny M. In contrast to agonist monoclonal antibodies, both C-terminal truncated form and full length form of Pleiotrophin failed to activate vertebrate ALK (anaplastic lymphoma kinase)? Cell Signal 2007; 19:2434-43. [PMID: 17904822 DOI: 10.1016/j.cellsig.2007.07.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 07/12/2007] [Accepted: 07/18/2007] [Indexed: 02/04/2023]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase essentially and transiently expressed during development in specific regions of the central and peripheral nervous system. ALK expression persists at a lower level in the adult brain. Thus, it might play an important role in both the normal development and function of the nervous system. The nature of the cognate ligand of this receptor in vertebrates is still a matter of debate. Pleiotrophin and midkine have been proposed as ligands of ALK but several independent studies do not confirm this hypothesis. Interestingly, a recent study proposed that a C-terminal truncated form of Pleiotrophin (Pleiotrophin.15) and not the full length form (Pleiotrophin.18) promotes glioblastoma proliferation in an ALK-dependent fashion. These data were obviously a strong basis to conciliate the conflicting results so far reported in the literature. In the present study, we first purified to homogeneity the two forms of Pleiotrophin secreted by HEK 293 cells. In contrast to agonist monoclonal antibodies, both Pleiotrophin.15 and Pleiotrophin.18 failed to activate ALK in neuroblastoma and glioblastoma cells expressing this receptor. Thus, for our point of view, ALK is still an orphan receptor in vertebrates.
Collapse
Affiliation(s)
- Thomas Mathivet
- INSERM, UMR-S 839, Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris, F-75005, France
| | | | | |
Collapse
|
15
|
Abstract
Pleiotropin (PTN) is a secreted heparin-binding peptide expressed in mesodermal and neuroectodermal cells during development, but rarely in adult tissues. Although PTN is abundant in fetal or juvenile cartilage, it is undectable in mature cartilage. However, PTN is re-expressed in chondrocytes in early stages of osteoarthritis where it is detectable in situ and in synovial fluids from patients. PTN enhances chondrogenesis by stimulation of extra-cellular matrix synthesis, reduction of degrading matrix metalloproteases and induction of their inhibitors; PTN also slightly reduces pro-inflammatory factors, such as nitric oxide and vascular endothelial growth factor. Furthermore, PTN stimulates chondrocyte clustering and proliferation. Thus, PTN appears to mediate repair and protective processes in osteoarthritic cartilage and appears to be a promising factor to treat osteoarthritis.
Collapse
Affiliation(s)
- Rolf Mentlein
- University of Kiel, Department of Anatomy, Olshausenstrasse 40, 24098 Kiel, Germany.
| |
Collapse
|
16
|
Chen H, Gordon MS, Campbell RA, Li M, Wang CS, Lee HJ, Sanchez E, Manyak SJ, Gui D, Shalitin D, Said J, Chang Y, Deuel TF, Baritaki S, Bonavida B, Berenson JR. Pleiotrophin is highly expressed by myeloma cells and promotes myeloma tumor growth. Blood 2007; 110:287-95. [PMID: 17369488 DOI: 10.1182/blood-2006-08-042374] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Pleiotrophin (PTN) is an important developmental cytokine that is highly expressed during embryogenesis but shows very limited expression in adult tissues, where it is largely restricted to the brain. High PTN serum levels are associated with a variety of solid tumors. We recently showed that patients with multiple myeloma (MM) also have elevated serum levels of this protein and the amount of PTN correlated with the patients' disease status and response to treatment. In this study, we demonstrate that MM cell lines and the malignant cells from MM patients' bone marrow produced PTN and secreted PTN protein into the supernatants during short-term culture. Moreover, Ptn gene expression correlated with the patients' disease status. Inhibition of PTN with a polyclonal anti-PTN antibody reduced growth and enhanced apoptosis of MM cell lines and freshly isolated bone marrow tumor cells from MM patients in vitro. Importantly, this antibody also markedly suppressed the growth of MM in vivo using a severe combined immunodeficiency (SCID)-hu murine model. This represents the first study showing the importance of PTN in the growth of any hematological disorder. Because the expression of this protein is very limited in normal adult tissues, PTN may represent a new target for the treatment of MM.
Collapse
Affiliation(s)
- Haiming Chen
- Institute for Myeloma & Bone Cancer Research, West Hollywood, CA 90069, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Marchionini DM, Lehrmann E, Chu Y, He B, Sortwell CE, Becker KG, Freed WJ, Kordower JH, Collier TJ. Role of heparin binding growth factors in nigrostriatal dopamine system development and Parkinson's disease. Brain Res 2007; 1147:77-88. [PMID: 17368428 DOI: 10.1016/j.brainres.2007.02.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 11/11/2006] [Accepted: 02/06/2007] [Indexed: 11/17/2022]
Abstract
The developmental biology of the dopamine (DA) system may hold important clues to its reconstruction. We hypothesized that factors highly expressed during nigrostriatal development and re-expressed after injury and disease may play a role in protection and reconstruction of the nigrostriatal system. Examination of gene expression in the developing striatum suggested an important role for the heparin binding growth factor family at time points relevant to establishment of dopaminergic innervation. Midkine, pleiotrophin (PTN), and their receptors syndecan-3 and receptor protein tyrosine phosphatase beta/zeta, were highly expressed in the striatum during development. Furthermore, PTN was up-regulated in the degenerating substantia nigra of Parkinson's patients. The addition of PTN to ventral mesencephalic cultures augmented DA neuron survival and neurite outgrowth. Thus, PTN was identified as a factor that plays a role in the nigrostriatal system during development and in response to disease, and may therefore be useful for neuroprotection or reconstruction of the DA system.
Collapse
Affiliation(s)
- Deanna M Marchionini
- Dept. Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Pufe T, Groth G, Goldring MB, Tillmann B, Mentlein R. Effects of pleiotrophin, a heparin-binding growth factor, on human primary and immortalized chondrocytes. Osteoarthritis Cartilage 2007; 15:155-62. [PMID: 16949312 DOI: 10.1016/j.joca.2006.07.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Accepted: 07/13/2006] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Pleiotrophin (PTN) is a secreted heparin-binding peptide expressed in mesodermal and neuroectodermal cells during development, but rarely in adult tissues. In fetal and juvenile, but not in mature cartilage, PTN is abundant. Furthermore, PTN is re-expressed in chondrocytes in early stages of osteoarthritis (OA). Since little is known about the functions of PTN in cartilage, we investigated the occurrence of PTN receptors in human articular cartilage in situ and PTN effects on human primary and immortalized chondrocytes in vitro. METHODS Receptor expression and regulation was monitored by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry. PTN effects and signal transduction were studied by electrophoretic mobility shift, Boyden chamber cell migration and proliferation assays, effects on gene expression by real time RT-PCR and that on nitric oxide (NO) by the Griess reaction. RESULTS Of the putative PTN signaling receptors, immortalized and primary chondrocytes (pc) expressed the anaplastic lymphoma kinase (ALK), less the receptor-type protein tyrosine phosphatase zeta/beta (PTPzeta). ALK expression was upregulated upon ligand exposure. PTN stimulation activated the AP-1 (activator protein-1) transcription factor and altered gene expression. Prolonged stimulation induced PTN mRNA expression slightly, reduced vascular endothelial growth factor (VEGF) mRNA as well as NO production. Whereas mRNA expression of matrix metalloproteinases (MMPs) MMP-1 and MMP-13 was reduced, their inhibitors TIMP-1 and TIMP-2 were induced. Furthermore, PTN stimulated chondrocyte migration and proliferation. CONCLUSIONS These results show that PTN is an autocrine growth factor in cartilage. We suggest that PTN may be involved in the clustering and proliferation of chondrocytes observed in the early stages of OA.
Collapse
Affiliation(s)
- T Pufe
- Department of Anatomy, University of Kiel, D-24098 Kiel, Germany
| | | | | | | | | |
Collapse
|
19
|
Gu D, Yu B, Zhao C, Ye W, Lv Q, Hua Z, Ma J, Zhang Y. The effect of pleiotrophin signaling on adipogenesis. FEBS Lett 2007; 581:382-8. [PMID: 17239862 DOI: 10.1016/j.febslet.2006.12.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Revised: 12/15/2006] [Accepted: 12/19/2006] [Indexed: 11/30/2022]
Abstract
Pleiotrophin (PTN) plays diverse roles in cell growth and differentiation. In this investigation, we demonstrate that PTN plays a negative role in adipogensis and that glycogen synthase kinase 3beta (GSK-3beta) and beta-catenin are involved in the regulation of PTN-mediated preadipocyte differentiation. Knocking down the expression of PTN using siRNA resulted in an increase in phospho-GSK-3beta expression, and the accumulation of nuclear beta-catenin, which are critical downstream signaling proteins for both the PTN and Wnt signaling pathways. Our investigation suggests that there is a PTN/PI3K/AKT/GSK-3beta/beta-catenin signaling pathway, which cross-talks with the Wnt/Fz/GSK-3beta/beta-catenin pathway and negatively regulates adipogenesis.
Collapse
Affiliation(s)
- Dayong Gu
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Room 407, Building L, Tsinghua Campus, University Town, Shenzhen, Guangdong 518055, PR China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Li G, Hu Y, Huo Y, Liu M, Freeman D, Gao J, Liu X, Wu DC, Wu H. PTEN deletion leads to up-regulation of a secreted growth factor pleiotrophin. J Biol Chem 2006; 281:10663-8. [PMID: 16507572 DOI: 10.1074/jbc.m512509200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Tumor suppressor gene PTEN is highly mutated in a wide variety of human tumors. To identify unknown targets or signal transduction pathways that are regulated by PTEN, microarray analysis was performed to compare the gene expression profiles of Pten null mouse embryonic fibroblasts (MEFs) cell lines and their isogenic counterparts. Expression of a heparin binding growth factor, pleiotrophin (Ptn), was found to be up-regulated in Pten-/- MEFs as well as Pten null mammary tumors. Further experiments revealed that Ptn expression is regulated by the PTEN-PI3K-AKT pathway. Knocking down the expression of Ptn by small interfering RNA resulted in the reduction of Akt and GSK-3beta phosphorylation and suppression of the growth and the tumorigenicity of Pten null MEFs. Our results suggest that PTN participates in tumorigenesis caused by PTEN loss and PTN may be a potential target for anticancer therapy, especially for those tumors with PTEN deficiencies.
Collapse
Affiliation(s)
- Gang Li
- Department of Molecular and Medical Pharmacology, UCLA School of Medicine, Los Angeles, California 90095-1735, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Antoine M, Tag CG, Wirz W, Borkham-Kamphorst E, Sawitza I, Gressner AM, Kiefer P. Upregulation of pleiotrophin expression in rat hepatic stellate cells by PDGF and hypoxia: Implications for its role in experimental biliary liver fibrogenesis. Biochem Biophys Res Commun 2005; 337:1153-64. [PMID: 16226713 DOI: 10.1016/j.bbrc.2005.09.173] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Accepted: 09/27/2005] [Indexed: 01/16/2023]
Abstract
Pleiotrophin (PTN) is a secretory heparin binding protein with various biological activities including mitogenesis, angiogenesis, and tissue repair after injury. Recent studies have shown that PTN is a strong mitogen of hepatocytes and involved in liver regeneration. In adult liver cells Ptn gene is mainly expressed by quiescent hepatic stellate cells (HSCs). Although we have been able to demonstrate mRNA and protein expression of the anaplastic lymphoma kinase-the receptor tyrosine kinase for PTN-on HSCs, PTN did not act as a mitogen of HSCs in contrast to hepatocytes. PTN immunoreactivity was markedly increased in experimental fibrogenesis by common bile duct ligation and observed in sinusoidal HSCs. In primary HSC cultures, Ptn transcription was significantly increased by PDGF-BB, and under hypoxic atmosphere. Mechanistically, hypoxia and PDGF mediated induction of PTN expression in sinusoidal HSCs may provide a strong mitogenic signal for hepatocytes to limit the damage to the parenchymal cells in biliary-type liver fibrogenesis.
Collapse
Affiliation(s)
- M Antoine
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH Aachen, Germany
| | | | | | | | | | | | | |
Collapse
|
22
|
Galindo CL, Fadl AA, Sha J, Pillai L, Gutierrez C, Chopra AK. Microarray and proteomics analyses of human intestinal epithelial cells treated with the Aeromonas hydrophila cytotoxic enterotoxin. Infect Immun 2005; 73:2628-43. [PMID: 15845465 PMCID: PMC1087361 DOI: 10.1128/iai.73.5.2628-2643.2005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We performed microarray analyses on RNA from human intestinal epithelial (HT-29) cells treated with the cytotoxic enterotoxin (Act) of Aeromonas hydrophila to examine global cellular transcriptional responses. Based on three independent experiments, Act upregulated the expression of 34 genes involved in cell growth, adhesion, signaling, immune responses (including interleukin-8 [IL-8] production), and apoptosis. We verified the upregulation of 14 genes by real-time reverse transcriptase-PCR and confirmed Act-induced production of IL-8 by enzyme-linked immunosorbent assay on supernatants from nonpolarized and polarized HT-29 cells. Maximal production of IL-8 in response to Act required the presence of intracellular calcium, since chelation of calcium with BAPTA-AM significantly reduced Act-induced IL-8 production in HT-29 cells. We also examined activation of mitogen-activated protein kinases and, as demonstrated by Western blot analysis of apical side-treated polarized HT-29 cells, Act induced phosphorylation of p38, c-Jun NH(2)-terminal kinase, and extracellular signal-regulated kinase 1/2. In addition, KinetWorks proteomics screening of whole-cell lysates revealed Act-induced phosphorylation of cyclic AMP-response element binding protein (CREB), c-Jun, adducin, protein kinase C, and signal transducer and activator of transcription 3 (STAT3) and decreased phosphorylation of protein kinase Balpha, v-raf-1 murine leukemia viral oncogene homolog 1 (i.e., Raf1), and STAT1. We verified activation of CREB and activator protein 1 in polarized cells by gel shift assay. This is the first description of human intestinal epithelial cell transcriptional alterations, phosphorylation or activation of signaling molecules, cytokine production, and calcium mobilization in response to this toxin.
Collapse
Affiliation(s)
- C L Galindo
- Department of Microbiology and Immunology, Medical Research Building, 301 University Blvd., University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | | | | | | | | | |
Collapse
|
23
|
Polykratis A, Katsoris P, Courty J, Papadimitriou E. Characterization of Heparin Affin Regulatory Peptide Signaling in Human Endothelial Cells. J Biol Chem 2005; 280:22454-61. [PMID: 15797857 DOI: 10.1074/jbc.m414407200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Heparin affin regulatory peptide (HARP) is an 18-kDa secreted growth factor that has a high affinity for heparin and a potent role on tumor growth and angiogenesis. We have previously reported that HARP is mitogenic for different types of endothelial cells and also affects cell migration and differentiation (12). In this study we examined the signaling pathways involved in the migration and tube formation on matrigel of human umbilical vein endothelial cells (HUVEC) induced by HARP. We report for the first time that receptor-type protein-tyrosine phosphatase beta/zeta (RPTPbeta/zeta), which is a receptor for HARP in neuronal cell types, is also expressed in HUVEC. We also document that HARP signaling through RPTPbeta/zeta leads to activation of Src kinase, focal adhesion kinase, phosphatidylinositol 3-kinase, and Erk1/2. Sodium orthovanadate, chondroitin sulfate-C, PP1, wortmannin, LY294002, and U0126 inhibit HARP-mediated signaling and HUVEC migration and tube formation. In addition, RPTPbeta/zeta suppression using small interfering RNA technology interrupts intracellular signals and HUVEC migration and tube formation induced by HARP. These results establish the role of RPTPbeta/zeta as a receptor of HARP in HUVEC and elucidate the HARP signaling pathway in endothelial cells.
Collapse
MESH Headings
- Blotting, Western
- CSK Tyrosine-Protein Kinase
- Carrier Proteins/chemistry
- Carrier Proteins/metabolism
- Cell Movement
- Cells, Cultured
- Collagen/pharmacology
- Cytokines/chemistry
- Cytokines/metabolism
- Dose-Response Relationship, Drug
- Drug Combinations
- Electrophoresis, Polyacrylamide Gel
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Focal Adhesion Kinase 1
- Focal Adhesion Protein-Tyrosine Kinases
- Humans
- Immunoprecipitation
- Laminin/pharmacology
- Neovascularization, Pathologic
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation
- Protein Structure, Tertiary
- Protein-Tyrosine Kinases/metabolism
- Proteoglycans/pharmacology
- RNA/metabolism
- RNA Interference
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Time Factors
- src-Family Kinases
Collapse
Affiliation(s)
- Apostolos Polykratis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | | | | | | |
Collapse
|
24
|
Lu KV, Jong KA, Kim GY, Singh J, Dia EQ, Yoshimoto K, Wang MY, Cloughesy TF, Nelson SF, Mischel PS. Differential induction of glioblastoma migration and growth by two forms of pleiotrophin. J Biol Chem 2005; 280:26953-64. [PMID: 15908427 DOI: 10.1074/jbc.m502614200] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma is the most common malignant brain tumor of adults and one of the most lethal cancers. The secreted growth factor pleiotrophin (PTN) promotes glioblastoma migration and proliferation, initiating its oncogenic activities through two cell surface receptors, the protein tyrosine phosphatase receptor zeta (PTPRZ1) and the anaplastic lymphoma kinase (ALK), respectively. Here, we report on the presence and purification of two naturally occurring forms of PTN (18 and 15 kDa) that differentially promote glioblastoma migration and proliferation. Using a panel of glioblastoma cell lines, including low passage patient-derived cultures, we demonstrate that PTN15 promotes glioblastoma proliferation in an ALK-dependent fashion, whereas immobilized PTN18 promotes haptotactic migration of glioblastoma cells in a PTPRZ1-dependent fashion. Mass spectrometric analysis indicated that PTN15 differs from PTN18 by processing of 12 C-terminal amino acids. To demonstrate clinical relevance, we show that PTN15, PTN18, and PTPRZ1 are significantly overexpressed in glioblastoma relative to normal brain at both mRNA and protein levels using microarray, Western blot, and tissue microarray analyses on human tumors. These results indicate that the PTN18-PTPRZ1 and the PTN15-ALK signaling pathways represent potentially important therapeutic targets for glioblastoma invasion and growth.
Collapse
Affiliation(s)
- Kan V Lu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Moog-Lutz C, Degoutin J, Gouzi JY, Frobert Y, Brunet-de Carvalho N, Bureau J, Créminon C, Vigny M. Activation and inhibition of anaplastic lymphoma kinase receptor tyrosine kinase by monoclonal antibodies and absence of agonist activity of pleiotrophin. J Biol Chem 2005; 280:26039-48. [PMID: 15886198 DOI: 10.1074/jbc.m501972200] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that is transiently expressed in specific regions of the central and peripheral nervous systems, suggesting a role in its normal development and function. The nature of the cognate ligands of ALK in vertebrate is still a matter of debate. We produced a panel of monoclonal antibodies (mAbs) directed against the extracellular domain of the human receptor. Two major species of ALK (220 and 140 kDa) were identified in transfected cells, and the use of our mAbs established that the 140-kDa species results from a cleavage of the 220-kDa form. Two mAbs, in the nm range, induced the differentiation of PC12 cells transiently transfected with ALK. In human embryonic kidney 293 cells stably expressing ALK, these two mAbs strongly activated the receptor and subsequently the mitogen-activated protein kinase pathway. We further showed for the first time that activation of ALK also resulted in a specific activation of STAT3. In contrast, other mAbs presented the characteristics of blocking antibodies. Finally, in these cell systems, a mitogenic form of pleiotrophin, a proposed ligand of ALK, failed to activate this receptor. Thus, in the absence of clearly established ligand(s) in vertebrates, the availability of mAbs allowing the activation or the inhibition of the receptor will be essential for a better understanding of the biological roles of ALK.
Collapse
Affiliation(s)
- Christel Moog-Lutz
- INSERM, Unité 706/Université Pierre et Marie Curie, Paris F-75005, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Lovicu FJ, McAvoy JW. Growth factor regulation of lens development. Dev Biol 2005; 280:1-14. [PMID: 15766743 DOI: 10.1016/j.ydbio.2005.01.020] [Citation(s) in RCA: 262] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Revised: 01/05/2005] [Accepted: 01/13/2005] [Indexed: 02/01/2023]
Abstract
Lens arises from ectoderm situated next to the optic vesicles. By thickening and invaginating, the ectoderm forms the lens vesicle. Growth factors are key regulators of cell fate and behavior. Current evidence indicates that FGFs and BMPs are required to induce lens differentiation from ectoderm. In the lens vesicle, posterior cells elongate to form the primary fibers whereas anterior cells differentiate into epithelial cells. The divergent fates of these embryonic cells give the lens its distinctive polarity. There is now compelling evidence that, at least in mammals, FGF is required to initiate fiber differentiation and that progression of this complex process depends on the synchronized and integrated action of a number of distinct growth factor-induced signaling pathways. It is also proposed that an antero-posterior gradient of FGF stimulation in the mammalian eye ensures that the lens attains and maintains its polarity and growth patterns. Less is known about differentiation of the lens epithelium; however, recent studies point to a role for Wnt signaling. Multiple Wnts and their receptors are expressed in the lens epithelium, and mice with impaired Wnt signaling have a deficient epithelium. Recent studies also indicate that other families of molecules, that can modulate growth factor signaling, have a role in regulating the ordered growth and differentiation of the lens.
Collapse
Affiliation(s)
- F J Lovicu
- Save Sight Institute, University of Sydney, NSW, Australia
| | | |
Collapse
|
27
|
Motegi A, Fujimoto J, Kotani M, Sakuraba H, Yamamoto T. ALK receptor tyrosine kinase promotes cell growth and neurite outgrowth. J Cell Sci 2005; 117:3319-29. [PMID: 15226403 DOI: 10.1242/jcs.01183] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor-type protein tyrosine kinase that is expressed preferentially in neurons of the central and peripheral nervous systems at late embryonic stages. To elucidate the role of ALK in neurons, we developed an agonist monoclonal antibody (mAb) against the extracellular domain of ALK. Here we show that mAb16-39 elicits tyrosine phosphorylation of endogenously expressed ALK in human neuroblastoma (SK-N-SH) cells. Stimulation of these cells with mAb16-39 markedly induces the tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1), Shc, and c-Cbl and also their interaction with ALK and activation of ERK1/2. Furthermore, we show that continuous incubation with mAb16-39 induces the cell growth and neurite outgrowth of SK-N-SH cells. These responses are completely blocked by MEK inhibitor PD98059 but not by the phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor wortmannin, indicating an essential role of the mitogen-activated protein kinase (MAP kinase) signaling cascade in ALK-mediated growth and differentiation of neurons.
Collapse
Affiliation(s)
- Akira Motegi
- Division of Oncology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639, Japan
| | | | | | | | | |
Collapse
|
28
|
Abstract
The urinary collecting system is derived from an epithelial protrusion arising from the Wolffian duct called the ureteric bud (UB) by the signal from its inductive tissue, metanephric mesenchyme (MM). Targeted gene mutation studies have shown that several transcription factors and MM-secreted glial cell line-derived neurotrophic factor (GDNF) are critical for initiation of the UB. After initiation, the UB undergoes branching morphogenesis. Results obtained from in vitro culture systems, including an isolated UB culture, together with gene mutation studies suggest that interplay of multiple positive and negative soluble factors as well as extracellular matrix (ECM) and matrix-degrading proteinases regulate branching morphogenesis.
Collapse
Affiliation(s)
- Hiroyuki Sakurai
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, 9500 Gilman Drive 0693, La Jolla, CA 92093-0693, USA.
| |
Collapse
|
29
|
Pufe T, Bartscher M, Petersen W, Tillmann B, Mentlein R. Pleiotrophin, an embryonic differentiation and growth factor, is expressed in osteoarthritis. Osteoarthritis Cartilage 2003; 11:260-4. [PMID: 12681952 DOI: 10.1016/s1063-4584(02)00385-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Pleiotrophin (PTN) is a 15.3 kDa heparin-binding peptide, which is expressed in mesodermal and neuroectodermal cells during development, but rarely in adult tissues. In fetal or juvenile cartilage, PTN is an abundant protein and appears to be involved in chondrocyte differentiation. Since developmentally regulated factors often re-appear in the disease state, we examined PTN expression in cartilage and synovial fluid of patients with osteoarthritis (OA). METHODS PTN mRNA and protein expression was assayed by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot, the protein was localized by immunohistochemistry and quantified by enzyme-linked immunoassay (ELISA). RESULTS PTN was undetectable in normal adult cartilage, but PTN mRNA and protein were found in OA. In cartilage from the tibial plateaus of OA patients, PTN could be immunostained in clusters of superficial chondrocytes. In the synovial fluids of OA patients, PTN concentrations were elevated in earlier OA stages, but rarely in late OA stages. Chondrosarcomas were PTN-immunonegative. CONCLUSIONS In addition to certain types of cancer, the embryonic growth and differentiation factor PTN is found also in adults in inflammatory diseases. In OA, PTN is especially expressed in early stages, and PTN concentrations in the synovial fluid could serve as a marker for the progress of the disease. PTN might be involved in cartilage repair in OA, in particular, in earlier stages.
Collapse
Affiliation(s)
- T Pufe
- Department of Anatomy, University of Kiel, D-24098, Kiel, Germany
| | | | | | | | | |
Collapse
|
30
|
Moon HS, Park WI, Sung SH, Choi EA, Chung HW, Woo BH. Immunohistochemical and quantitative competitive PCR analyses of midkine and pleiotrophin expression in cervical cancer. Gynecol Oncol 2003; 88:289-97. [PMID: 12648577 DOI: 10.1016/s0090-8258(02)00070-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE The aim of this study was to determine midkine (MK) and pleiotrophin (PTN) expression in cervical cancer. METHODS Prospective study in tertiary teaching hospital. Normal and cancerous cervical tissues were obtained from healthy women (n = 19) and from patients with cervical cancer (n = 42). The expressions of MK and PTN mRNA and protein were examined by quantitative competitive PCR and by immunohistochemistry. MK and PTN mRNA and protein expressions were examined with respect to tumor stage and size. RESULTS The expressions of midkine and pleiotrophin mRNA in cervical cancer were higher than those in the normal cervix (MK, 175.59 +/- 63.3 vs 1.00 +/- 0.18 fmol, respectively; PTN, 3.18 +/- 1.25 vs. 0.86 +/- 0.12 fmol, respectively, P < 0.05), and their expressions were not correlated with cervical cancer stage or size of the tumor. The expressions of MK and PTN protein in cancerous tissue were higher than those in the normal cervix (P < 0.05). Moreover, the protein expression of MK, but not of PTN, correlated with tumor stage and size. The expressions of MK and PTN were not correlated with vascular density. CONCLUSIONS Our results suggest that increased midkine mRNA and protein expressions are associated with the carcinogenesis of cervical cancer.
Collapse
Affiliation(s)
- Hye-Sung Moon
- Department of Obstetrics & Gynecology, Ewha Womans University and Medical Research Center, Seoul, Korea.
| | | | | | | | | | | |
Collapse
|
31
|
Pufe T, Bartscher M, Petersen W, Tillmann B, Mentlein R. Expression of pleiotrophin, an embryonic growth and differentiation factor, in rheumatoid arthritis. ARTHRITIS AND RHEUMATISM 2003; 48:660-7. [PMID: 12632418 DOI: 10.1002/art.10839] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Pleiotrophin (PTN), a 15.3-kd heparin-binding peptide, is expressed in mesodermal and neuroectodermal cells during development, but rarely in adult tissues. Since developmentally regulated factors often reappear during disease, we sought to determine whether there was PTN expression in the synovial membranes of patients with rheumatoid arthritis (RA). METHODS PTN messenger RNA expression was assayed by quantitative reverse transcriptase-polymerase chain reaction. The protein was localized by immunohistochemistry and quantified by enzyme-linked immunosorbent assay (ELISA). Effects of PTN on cell proliferation in vitro were determined by DNA measurements. RESULTS PTN expression in normal adult synovial membranes and cartilage was barely detectable. However, PTN was strongly up-regulated in synovial tissues from patients with RA. In contrast, samples from patients with pyogenic arthritis had moderate PTN levels, and those from patients with osteoarthritis had only a slight increase in PTN, as measured by ELISA. In RA patients, PTN was localized primarily in synoviocytes but was also found in endothelial cells of blood vessels. In cultured mouse fibroblasts used as a model, PTN expression was up-regulated by tumor necrosis factor alpha and was more weakly up-regulated by epidermal growth factor. Recombinant PTN stimulated the proliferation of cultured human synoviocytes and the monocyte cell line THP-1, but not human dermal fibroblasts, in which PTN increased the synthesis of vascular endothelial growth factor. CONCLUSION In addition to certain types of cancer, the embryonic growth and differentiation factor PTN is expressed in adults with inflammatory diseases, in particular, RA. Proinflammatory cytokines enhance the expression of PTN. Thus, we propose that PTN is a further paracrine angiogenesis and growth factor for synovial cells in RA.
Collapse
|
32
|
Deepa SS, Umehara Y, Higashiyama S, Itoh N, Sugahara K. Specific molecular interactions of oversulfated chondroitin sulfate E with various heparin-binding growth factors. Implications as a physiological binding partner in the brain and other tissues. J Biol Chem 2002; 277:43707-16. [PMID: 12221095 DOI: 10.1074/jbc.m207105200] [Citation(s) in RCA: 269] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously observed that the cortical neuronal cell adhesion mediated by midkine (MK), a heparin (Hep)-binding growth factor, is specifically inhibited by oversulfated chondroitin sulfate-E (CS-E) (Ueoka, C., Kaneda, N., Okazaki, I., Nadanaka, S., Muramatsu, T., and Sugahara, K. (2000) J. Biol. Chem. 275, 37407-37413) and that CS-E exhibits neurite outgrowth promoting activities toward embryonic rat hippocampal neurons. We have also shown oversulfated CS chains in embryonic chick and rat brains and demonstrated that the CS disaccharide composition changes during brain development. In view of these findings, here we tested the possibility of CS-E interacting with Hep-binding growth factors during development, using squid cartilage CS-E. The binding ability of Hep-binding growth factors (MK, pleiotrophin (PTN), fibroblast growth factor-1 (FGF-1), FGF-2, Hep-binding epidermal growth factor-like growth factor (HB-EGF), FGF-10, FGF-16, and FGF-18) toward [(3)H]CS-E was first tested by a filter binding assay, which demonstrated direct binding of all growth factors, except FGF-1, to CS-E. The bindings were characterized further in an Interaction Analysis system, where all of the growth factors, except FGF-1, gave concentration-dependent and specific bindings. The kinetic constants k(a), k(d), and K(d) suggested that MK, PTN, FGF-16, FGF-18, and HB-EGF bound strongly to CS-E, in comparable degrees to the binding to Hep, whereas the intensity of binding of FGF-2 and FGF-10 toward CS-E was lower than that for Hep. These findings suggest the possibility of CS-E being a binding partner, a coreceptor, or a genuine receptor for various Hep-binding growth factors in the brain and possibly also in other tissues.
Collapse
|
33
|
Mentlein R, Held-Feindt J. Pleiotrophin, an angiogenic and mitogenic growth factor, is expressed in human gliomas. J Neurochem 2002; 83:747-53. [PMID: 12421346 DOI: 10.1046/j.1471-4159.2002.01179.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pleiotrophin (PTN) is a mitogenic/angiogenic, 15.3 kDa heparin-binding peptide that is found in embryonic or early postnatal, but rarely in adult, tissues. Since developmentally regulated factors often re-appear in malignant cells, we examined PTN expression in human glioma cell lines, cell cultures derived from solid gliomas and glioma sections. PTN mRNA or protein was detected by reverse transcriptase-polymerase chain reaction, immunohistochemistry, western blot or enzyme-linked immunoassay in all WHO III and IV grade gliomas and cells analyzed in vitro or in situ. One WHO II grade glioma investigated was PTN negative. In vitro, PTN was synthesized in perinuclear regions of glioma cells, secreted into the cultivation medium, but its production varied considerably between glioma cells cultivated from different solid gliomas or glioma cell lines. In situ, PTN expression was restricted to distinct parts/cells of the tumour. PTN did not influence the proliferation of glioma cells themselves, but stimulated [3H]thymidine incorporation into DNA of microglial cells. Furthermore, in Boyden chamber assays, PTN showed a strong chemotactic effect on murine BV-2 microglial cells. PTN is supposed to be a paracrine growth/angiogenic factor that is produced by gliomas and contributes to their malignancy by targeting endothelial and microglial cells.
Collapse
Affiliation(s)
- Rolf Mentlein
- Department of Anatomy, University of Kiel, Kiel, Germany.
| | | |
Collapse
|
34
|
Bowden ET, Stoica GE, Wellstein A. Anti-apoptotic signaling of pleiotrophin through its receptor, anaplastic lymphoma kinase. J Biol Chem 2002; 277:35862-8. [PMID: 12107166 DOI: 10.1074/jbc.m203963200] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The secreted growth factor pleiotrophin (PTN) can induce mitogenesis in cells that express the receptor for this growth factor, anaplastic lymphoma kinase (ALK). Here we examine the ability of PTN to produce anti-apoptotic signals. We demonstrate that PTN is a survival factor for SW-13 epithelial cells and show that ribozyme-mediated depletion of ALK from SW-13 cells abolishes this effect of PTN. Furthermore, in serum-starved NIH3T3 fibroblasts PTN prevents apoptosis (measured by annexin V staining) with an EC(50) of 0.2 ng/ml and induces cell growth at higher concentrations of PTN. A polyclonal antibody against the PTN ligand-binding domain of the ALK receptor (alpha-LBD) was a partial agonist for ALK in NIH3T3 cells. This alpha-LBD antibody showed high agonist activity for anti-apoptosis (56 +/- 9% relative to PTN), low agonist activity for cell growth (21 +/- 1% relative to PTN), and was an antagonist of PTN-induced cell growth (61 +/- 2% inhibition). Both MAP kinase and phosphatidylinositol (PI) 3-kinase cascades in NIH3T3 cells were activated by PTN, and this effect persisted for up to 3 h. Surprisingly, the anti-apoptotic effect of PTN was completely blocked by the MAP kinase inhibitor UO126, but was not affected by the PI 3-kinase inhibitor LY294002. In contrast, PTN-dependent cell growth required both MAPK and PI 3-kinase activity. We conclude that anti-apoptotic signaling of PTN through ALK in NIH3T3 fibroblasts is via the MAP kinase pathway.
Collapse
Affiliation(s)
- Emma T Bowden
- Department of Oncology, Lombardi Cancer Center, Georgetown University Medical School, Washington, D. C. 20007, USA
| | | | | |
Collapse
|
35
|
Stoica GE, Kuo A, Powers C, Bowden ET, Sale EB, Riegel AT, Wellstein A. Midkine binds to anaplastic lymphoma kinase (ALK) and acts as a growth factor for different cell types. J Biol Chem 2002; 277:35990-8. [PMID: 12122009 DOI: 10.1074/jbc.m205749200] [Citation(s) in RCA: 254] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Midkine (MK) is a developmentally regulated, secreted growth factor homologous to pleiotrophin (PTN). To investigate the potential role of MK in tumor growth, we expressed MK in human SW-13 cells and studied receptor binding, signal transduction, and activity of MK. The MK protein stimulates soft agar colony formation in vitro and tumor growth of SW-13 cells in athymic nude mice, as well as proliferation of human endothelial cells from brain microvasculature and umbilical vein (HUVEC) in the low ng/ml range. MK binds to anaplastic lymphoma kinase (ALK), the receptor for PTN, with an apparent K(d) of 170 pm in intact cells, and this receptor binding of MK is competed by PTN with an apparent K(d) of approximately 20 pm. Monoclonal antibodies raised against the extracellular ligand-binding domain of ALK inhibit ALK receptor binding of MK as well as MK-stimulated colony formation of SW-13 cells. Furthermore, MK stimulates ALK phosphorylation in WI-38 human fibroblasts and activates PI3-kinase and MAP kinase signal transduction in WI-38, HUVEC, neuroblastoma (SH SY-5Y) and glioblastoma (U87MG) cells that express the ALK protein. We conclude that MK can act as a growth, survival, and angiogenic factor during tumorigenesis and signals through the ALK receptor.
Collapse
Affiliation(s)
- Gerald E Stoica
- Lombardi Cancer Center, Georgetown University, Washington, D. C. 20007, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Bernard-Pierrot I, Delbé J, Rouet V, Vigny M, Kerros ME, Caruelle D, Raulais D, Barritault D, Courty J, Milhiet PE. Dominant negative effectors of heparin affin regulatory peptide (HARP) angiogenic and transforming activities. J Biol Chem 2002; 277:32071-7. [PMID: 12070152 DOI: 10.1074/jbc.m202747200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparin affin regulatory peptide (HARP) is an heparin-binding growth factor, highly expressed in several primary human tumors and considered as a rate-limiting angiogenic factor in tumor growth, invasion, and metastasis. Implication of this protein in carcinogenesis is linked to its mitogenic, angiogenic, and transforming activities. Recently, we have demonstrated that the C-terminal residues 111-136 of HARP are required for its mitogenic and transforming activities (Bernard-Pierrot, I., Delbe, J., Caruelle, D., Barritault, D., Courty, J., and Milhiet, P. E. (2001) J. Biol. Chem. 276, 12228-12234). In this paper, HARP deleted of its last 26 amino acids was shown to act as a dominant negative effector for its mitogenic, angiogenic, transforming, and tumor-formation activities by heterodimerizing with the wild type protein. Similarly, the synthetic corresponding peptide P111-136 displayed in vitro inhibition of wild type HARP activities, but in this case, the inhibition was mainly explained by the competition of the peptide with HARP for the binding to the extracellular domain of the high affinity ALK receptor.
Collapse
Affiliation(s)
- Isabelle Bernard-Pierrot
- Laboratoire de recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), CNRS UPRES-A 7053, Université Paris XII, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Piccinini G, Bacchiocchi R, Serresi M, Vivani C, Rossetti S, Gennaretti C, Carbonari D, Fazioli F. A ligand-inducible epidermal growth factor receptor/anaplastic lymphoma kinase chimera promotes mitogenesis and transforming properties in 3T3 cells. J Biol Chem 2002; 277:22231-9. [PMID: 11919185 DOI: 10.1074/jbc.m111145200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oncogenic rearrangements of the anaplastic lymphoma kinase (ALK) gene, encoding a receptor type tyrosine kinase, are frequently associated with anaplastic large cell lymphomas. Such rearrangements juxtapose the intracellular domain of ALK to 5'-end sequences belonging to different genes and create transforming fusion proteins. To understand how the oncogenic versions of ALK contribute to lymphomagenesis, it is important to analyze the biological effects and the biochemical properties of this receptor under controlled conditions of activation. To this aim, we constructed chimeric receptor molecules in which the extracellular domain of the ALK kinase is replaced by the extracellular, ligand-binding domain of the epidermal growth factor receptor (EGFR). Upon transfection in NIH 3T3 fibroblasts, the EGFR/ALK chimera was correctly synthesized and transported to the cell surface, where it was fully functional in forming high versus low affinity EGF-binding sites and transducing an EGF-dependent signal intracellularly. Overexpression of the EGFR/ALK chimera in NIH 3T3 was sufficient to induce the malignant phenotype; the appearance of the transformed phenotype was, however, conditionally dependent on the administration of EGF. Moreover, the EGFR/ALK chimera was significantly more active in inducing transformation and DNA synthesis than the wild type EGFR when either was expressed at similar levels in NIH 3T3 cells. Comparative analysis of the biochemical pathways implicated in the transduction of mitogenic signals did not show any increased ability of the EGFR/ALK to phosphorylate PLC-gamma and MAPK compared with the EGFR. On the contrary, EGFR/ALK showed to have a consistently greater effect on phosphatidylinositol 3-kinase activity compared with the EGFR, indicating that this enzyme plays a major role in mediating the mitogenic effects of ALK in NIH 3T3 cells.
Collapse
Affiliation(s)
- Gina Piccinini
- Laboratory of Cellular and Molecular Biology, Institute of Internal Medicine, University of Ancona, Via Tronto 10/A, 60020 Ancona, Italy
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Zatechka SD, Lou MF. Studies of the mitogen-activated protein kinases and phosphatidylinositol-3 kinase in the lens. 1. The mitogenic and stress responses. Exp Eye Res 2002; 74:703-17. [PMID: 12126944 DOI: 10.1006/exer.2002.1168] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The current understanding of the cellular signal transduction system is that cells initially respond to outside stimuli, such as growth factors or neurotransmitters, through ligand binding to the respective growth factor receptors or the G-protein-coupled receptors, to initiate transduction of the stimulus. This is followed by a series of association-dissociation and phosphorylation-dephosphorylation processes among the components of a well-defined and intricate infrastructure between the cell membrane and the cytosolic protein kinases to activate and initiate nuclear target genes for cell proliferation, differentiation and other cellular functions. Although some past reports have indicated this signaling machinery is present in the lens, certain pathways, namely the mitogen-response pathway (Raf-MEK-ERK cascade), the stress-response pathways (p38 and SAPK/JNK cascades) and the survival pathway (PI-3K-Akt), have not been thoroughly explored in an intact lens. These pathways were studied using porcine lenses cultured under mitogenic (10 ngml(-1) growth factor) or osmotic stress (30 mM galactose) conditions to examine the cellular response in the epithelial layer, using unstimulated lenses as controls. It was found that all the key members in the Raf-MEK-ERK cascade and PI-3K-Akt cascade were present and that growth factors had a differential stimulatory effect on them. Basic-FGF was the most potent stimulator for ERK followed by EGF and IGF-1, while PDGFab and VEGF were less active. The opposite was true for their stimulatory effect on PI-3K. Hyperglycemic-induced osmotic stress stimulated p38 but not SAPK/JNK, while bFGF could stimulate SAPK/JNK but not p38. Both stimuli activated the Raf-MEK-ERK and PI-3K-Akt pathways. Osmotic-induced activation could be normalized using an aldose reductase inhibitor.
Collapse
Affiliation(s)
- Steven D Zatechka
- Department of Veterinary and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | | |
Collapse
|
39
|
Powers C, Aigner A, Stoica GE, McDonnell K, Wellstein A. Pleiotrophin signaling through anaplastic lymphoma kinase is rate-limiting for glioblastoma growth. J Biol Chem 2002; 277:14153-8. [PMID: 11809760 DOI: 10.1074/jbc.m112354200] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma multiforme is the most common highly aggressive human brain cancer, and receptor tyrosine kinases have been implicated in the progression of this malignancy. We have recently identified anaplastic lymphoma kinase (ALK) as a tyrosine kinase receptor for pleiotrophin, a secreted growth factor that is highly expressed during embryonic brain development and in tumors of the central nervous system. Here we report on the contribution of pleiotrophin-ALK signaling to glioblastoma growth. We found ALK overexpressed in human glioblastoma relative to normal brain and detected ALK mRNA in glioblastoma cell lines. We reduced the endogenous ALK in glioblastoma cells by ribozyme targeting and demonstrated that this prevents pleiotrophin-stimulated phosphorylation of the anti-apoptotic protein Akt. Furthermore, this depletion of ALK reduced tumor growth of xenografts in athymic nude mice and prolonged survival of the animals because of increased apoptosis in the tumors. These findings directly implicate ALK signaling as a rate-limiting factor in the growth of glioblastoma multiforme and suggest potential utility of therapeutic targeting of ALK.
Collapse
Affiliation(s)
- Ciaran Powers
- Lombardi Cancer Center, Department of Oncology, Georgetown University, Washington, DC, 20007, USA
| | | | | | | | | |
Collapse
|
40
|
Haynes L, Rumsby M. The pleiotropin/midkine family of cytokines: role in glial-neuronal signalling. PROGRESS IN BRAIN RESEARCH 2001; 132:313-24. [PMID: 11545000 DOI: 10.1016/s0079-6123(01)32085-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- L Haynes
- School of Biological Sciences, University of Bristol, Bristol BS8 1UG, UK
| | | |
Collapse
|
41
|
Sakurai H, Bush KT, Nigam SK. Identification of pleiotrophin as a mesenchymal factor involved in ureteric bud branching morphogenesis. Development 2001; 128:3283-93. [PMID: 11546745 DOI: 10.1242/dev.128.17.3283] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Branching morphogenesis is central to epithelial organogenesis. In the developing kidney, the epithelial ureteric bud invades the metanephric mesenchyme, which directs the ureteric bud to undergo repeated branching. A soluble factor(s) in the conditioned medium of a metanephric mesenchyme cell line is essential for multiple branching morphogenesis of the isolated ureteric bud. The identity of this factor had proved elusive, but it appeared distinct from factors such as HGF and EGF receptor ligands that have been previously implicated in branching morphogenesis of mature epithelial cell lines. Using sequential column chromatography, we have now purified to apparent homogeneity an 18 kDa protein, pleiotrophin, from the conditioned medium of a metanephric mesenchyme cell line that induces isolated ureteric bud branching morphogenesis in the presence of glial cell-derived neurotrophic factor. Pleiotrophin alone was also found to induce the formation of branching tubules in an immortalized ureteric bud cell line cultured three-dimensionally in an extracellular matrix gel. Consistent with an important role in ureteric bud morphogenesis during kidney development, pleiotrophin was found to localize to the basement membrane of the developing ureteric bud in the embryonic kidney. We suggest that pleiotrophin could act as a key mesenchymally derived factor regulating branching morphogenesis of the ureteric bud and perhaps other embryonic epithelial structures.
Collapse
Affiliation(s)
- H Sakurai
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0693, USA
| | | | | |
Collapse
|
42
|
Aigner A, Juhl H, Malerczyk C, Tkybusch A, Benz CC, Czubayko F. Expression of a truncated 100 kDa HER2 splice variant acts as an endogenous inhibitor of tumour cell proliferation. Oncogene 2001; 20:2101-11. [PMID: 11360194 DOI: 10.1038/sj.onc.1204305] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2000] [Revised: 10/04/2000] [Accepted: 01/22/2001] [Indexed: 11/09/2022]
Abstract
Overexpression of the HER2 (neu/c-erbB-2) oncogene frequently coincides with an aggressive clinical course of certain human adenocarcinomas. Expression and secretion of aberrant HER2 splice variants has been reported in various cell lines and tissues and can interfere with the oncogenic HER2 activity. Here we demonstrate, using two different approaches, that expression of a truncated 100 kDa HER2 variant which encodes the extracellular domain of HER2 (HER-ECD) inhibits growth factor-mediated tumour cell proliferation. A HER2-ECD cDNA encoding the truncated variant was overexpressed in MCF7 breast cancer cells. HER2-ECD overexpression decreased spontaneous proliferation of MCF7 cells as well as heregulin-mediated soft agar colony formation. Concomitantly, heregulin-induced phosphorylation of HER4 as well as downstream activation of p44/p42 MAP-kinases was decreased. To confirm these data, ribozymes were targeted to the 3'-untranslated region of the 2.3 kb HER2-ECD mRNA which is spontaneously expressed in MKN7 gastric cancer cells. HER2-ECD-targeted ribozymes downregulated HER2-ECD expression and enhanced EGF-mediated soft agar colony formation of MKN7 cells. In parallel, EGF-induced activation of p44/p42 MAP-kinases and activation of c-Fos expression were increased in ribozyme-transfected MKN7 cells. Finally, in RT-PCR we found a trend towards a progressive loss of 2.3 kb HER2-ECD mRNA expression in more advanced gastric tumours. These data show that the HER2-ECD variant inhibits growth factor-mediated tumour cell proliferation suggesting an important role during the progression of human cancer.
Collapse
MESH Headings
- Alternative Splicing
- Base Sequence
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Division/drug effects
- Cell Division/physiology
- DNA, Complementary/genetics
- Down-Regulation
- Doxycycline/pharmacology
- Epidermal Growth Factor/antagonists & inhibitors
- Epidermal Growth Factor/pharmacology
- Genes, erbB-2/genetics
- Growth Inhibitors/biosynthesis
- Growth Inhibitors/genetics
- Growth Inhibitors/physiology
- Humans
- MAP Kinase Signaling System/physiology
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3
- Mitogen-Activated Protein Kinases/metabolism
- Molecular Sequence Data
- Neuregulin-1/antagonists & inhibitors
- Neuregulin-1/pharmacology
- Protein Structure, Tertiary
- RNA, Catalytic/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/physiology
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A Aigner
- Department of Pharmacology and Toxicology, Philipps-University Marburg, Karl-von-Frisch-Strasse 1, D-35033 Marburg, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Stoica GE, Kuo A, Aigner A, Sunitha I, Souttou B, Malerczyk C, Caughey DJ, Wen D, Karavanov A, Riegel AT, Wellstein A. Identification of anaplastic lymphoma kinase as a receptor for the growth factor pleiotrophin. J Biol Chem 2001; 276:16772-9. [PMID: 11278720 DOI: 10.1074/jbc.m010660200] [Citation(s) in RCA: 294] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Pleiotrophin (PTN) is a secreted growth factor that induces neurite outgrowth and is mitogenic for fibroblasts, epithelial, and endothelial cells. During tumor growth PTN can serve as an angiogenic factor and drive tumor invasion and metastasis. To identify a receptor for PTN, we panned a phage display human cDNA library against immobilized PTN protein as a bait. From this we isolated a phage insert that was homologous to an amino acid sequence stretch in the extracellular domain (ECD) of the orphan receptor tyrosine kinase anaplastic lymphoma kinase (ALK). In parallel with PTN, ALK is highly expressed during perinatal development of the nervous system and down-modulated in the adult. Here we show in cell-free assays as well as in radioligand receptor binding studies in intact cells that PTN binds to the ALK ECD with an apparent Kd of 32 +/- 9 pm. This receptor binding is inhibited by an excess of PTN, by the ALK ECD, and by anti-PTN and anti-ECD antibodies. PTN added to ALK-expressing cells induces phosphorylation of both ALK and of the downstream effector molecules IRS-1, Shc, phospholipase C-gamma, and phosphatidylinositol 3-kinase. Furthermore, the growth stimulatory effect of PTN on different cell lines in culture coincides with the endogenous expression of ALK mRNA, and the effect of PTN is enhanced by ALK overexpression. From this we conclude that ALK is a receptor that transduces PTN-mediated signals and propose that the PTN-ALK axis can play a significant role during development and during disease processes.
Collapse
Affiliation(s)
- G E Stoica
- Lombardi Cancer Center, Georgetown University, Washington, DC 20007, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Qi M, Ikematsu S, Maeda N, Ichihara-Tanaka K, Sakuma S, Noda M, Muramatsu T, Kadomatsu K. Haptotactic migration induced by midkine. Involvement of protein-tyrosine phosphatase zeta. Mitogen-activated protein kinase, and phosphatidylinositol 3-kinase. J Biol Chem 2001; 276:15868-75. [PMID: 11340082 DOI: 10.1074/jbc.m005911200] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Midkine, a heparin-binding growth factor, plays a critical role in cell migration causing suppression of neointima formation in midkine-deficient mice. Here we have determined the molecules essential for midkine-induced migration. Midkine induced haptotaxis of osteoblast-like cells, which was abrogated by the soluble form of midkine or pleiotrophin, a midkine-homologous protein. Chondroitin sulfate B, E, chondroitinase ABC, B, and orthovanadate, an inhibitor of protein-tyrosine phosphatase, suppressed the migration. Supporting these data, the cells examined expressed PTPzeta, a receptor-type protein-tyrosine phosphatase that exhibits high affinity to both midkine and pleiotrophin and harbors chondroitin sulfate chains. Furthermore, strong synergism between midkine and platelet-derived growth factor in migration was detected. The use of specific inhibitors demonstrated that mitogen-activated protein (MAP) kinase and protein-tyrosine phosphatase were involved in midkine-induced haptotaxis but not PDGF-induced chemotaxis, whereas phosphatidylinositol 3 (PI3)-kinase and protein kinase C were involved in both functions. Midkine activated both PI3-kinase and MAP kinases, the latter activation was blocked by a PI3-kinase inhibitor. Midkine further recruited PTPzeta and PI3-kinase. These results indicate that PTPzeta and concerted signaling involving PI3-kinase and MAP kinase are required for midkine-induced migration and demonstrate for the first time the synergism between midkine and platelet-derived growth factor in cell migration.
Collapse
Affiliation(s)
- M Qi
- Department of Biochemistry, Nagoya University School of Medicine, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Bernard-Pierrot I, Delbe J, Caruelle D, Barritault D, Courty J, Milhiet PE. The lysine-rich C-terminal tail of heparin affin regulatory peptide is required for mitogenic and tumor formation activities. J Biol Chem 2001; 276:12228-34. [PMID: 11150308 DOI: 10.1074/jbc.m010913200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparin affin regulatory peptide (HARP) is a 18-kDa heparin-binding polypeptide that is highly expressed in developing tissues and in several primary human tumors. It seems to play a key role in cellular growth and differentiation. In vitro, HARP displays mitogenic, angiogenic, and neurite outgrowth activities. It is a secreted protein that is organized in two beta-sheet domains, each domain containing a cluster of basic residues. To assess determinants involved in the biological activities of HARP, C-terminally truncated proteins were produced in Chinese hamster ovary-K1 cells and tested for their mitogenic, tumor formation in nude mice and neurite outgrowth activities. Our data clearly indicate that the residues 111-136 of the lysine-rich C-terminal domain are involved in the mitogenic and tumor formation activities of HARP. Correlatively, no signal transduction was detected using the corresponding mutant, suggesting the absence of HARP binding to its high affinity receptor. However, this C-terminal domain of HARP is not involved in the neurite outgrowth activity. We also demonstrate that HARP signal peptide cleavage could led to two maturated forms that are both but differentially mitogenic.
Collapse
Affiliation(s)
- I Bernard-Pierrot
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires, CNRS UPRES-A 7053, Université Paris XII, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | | | | | | | | | | |
Collapse
|
46
|
Souttou B, Raulais D, Vigny M. Pleiotrophin induces angiogenesis: involvement of the phosphoinositide-3 kinase but not the nitric oxide synthase pathways. J Cell Physiol 2001; 187:59-64. [PMID: 11241349 DOI: 10.1002/1097-4652(2001)9999:9999<00::aid-jcp1051>3.0.co;2-f] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Pleiotrophin (PTN) is a developmentally regulated protein that has been shown to be involved in tumor growth and metastasis presumably by activating tumor angiogenesis. To clarify the potential angiogenic activity of PTN and to analyze the signaling pathways involved in this process, we used an in vitro model of Human Umbilical Vein Endothelial Cells (HUVEC). We show that PTN was mitogenic toward a variety of endothelial cells including HUVEC, stimulated HUVEC migration across a reconstituted basement membrane and induced the formation of capillary-like structures by HUVEC grown as 3D-cultures in Matrigel or collagen. The signaling pathways triggered following endothelial cell stimulation by PTN were studied by using pharmacological inhibitors of the Phosphoinositide-3 kinase (PI3K) and endothelial Nitric Oxide Synthase (eNOS), two enzymes that have been shown to be crucial in the angiogenic response to Vascular Endothelial Growth Factor (VEGF). Whereas wortmannin (a PI3K inhibitor) and L-NAME (an eNOS inhibitor) dramatically reduced HUVEC growth induced by VEGF, only the former inhibitor reduced the growth induced by PTN and to a lesser extent that stimulated by basic Fibroblast Growth Factor. Thus, our results indicate that PTN induces angiogenesis and utilizes PI3K- but not eNOS-dependent pathways for its angiogenic activity.
Collapse
Affiliation(s)
- B Souttou
- INSERM Unité 440/Université Paris 6, Paris, France
| | | | | |
Collapse
|
47
|
Papadimitriou E, Polykratis A, Courty J, Koolwijk P, Heroult M, Katsoris P. HARP induces angiogenesis in vivo and in vitro: implication of N or C terminal peptides. Biochem Biophys Res Commun 2001; 282:306-13. [PMID: 11264008 DOI: 10.1006/bbrc.2001.4574] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
HARP (heparin affin regulatory peptide) is a growth factor displaying high affinity for heparin. In the present work, we studied the ability of human recombinant HARP as well as its two terminal peptides (HARP residues 1-21 and residues 121-139) to promote angiogenesis. HARP stimulates endothelial cell tube formation on matrigel, collagen and fibrin gels, stimulates endothelial cell migration and induces angiogenesis in the in vivo chicken embryo chorioallantoic membrane assay. The two HARP peptides seem to be involved in most of the angiogenic effects of HARP. They both stimulate in vivo angiogenesis and in vitro endothelial cell migration and tube formation on matrigel. We conclude that HARP has an angiogenic activity when applied exogenously in several in vitro and in vivo models of angiogenesis and its NH(2) and COOH termini seem to play an important role.
Collapse
Affiliation(s)
- E Papadimitriou
- Laboratory of Molecular Pharmacology, University of Patras, Patras, Greece
| | | | | | | | | | | |
Collapse
|
48
|
Papadimitriou E, Heroult M, Courty J, Polykratis A, Stergiou C, Katsoris P. Endothelial cell proliferation induced by HARP: implication of N or C terminal peptides. Biochem Biophys Res Commun 2000; 274:242-8. [PMID: 10903925 DOI: 10.1006/bbrc.2000.3126] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
HARP (Heparin Affin Regulatory Peptide) is a 18-kDa secreted protein displaying high affinity for heparin. It has neurite outgrowth-promoting activity, while there are conflicting results regarding its mitogenic activity. In the present work, we studied the effect of human recombinant HARP expressed in bacterial cells as well as two peptides (HARP residues 1-21 and residues 121-139) on the proliferation of three endothelial cell types derived from human umbilical vein (HUVEC), rat adrenal medulla (RAME), and bovine brain capillaries (BBC) either added as a soluble form in the cell culture medium or coated onto the culture plate. HARP added in a soluble form in the culture medium had no effect on the proliferation of BBC, HUVEC, and RAME cells. However, when immobilized onto the cell culture plate, HARP had a concentration-dependent mitogenic effect on both BBC cells and HUVEC. The peptides presented as soluble factor induced a significant concentration-dependent mitogenic effect on BBC cells but only a small effect on HUVEC and RAME cells. When they were immobilized onto the cell culture plate, the mitogenic effect was much greater. The most responsive cells were BBC that expressed and secreted in the culture medium the higher amounts of HARP.
Collapse
Affiliation(s)
- E Papadimitriou
- Laboratory of Molecular Pharmacology, University of Patras, Patras, GR26504, Greece
| | | | | | | | | | | |
Collapse
|
49
|
Hsieh SS, Malerczyk C, Aigner A, Czubayko F. ERbB-2 expression is rate-limiting for epidermal growth factor-mediated stimulation of ovarian cancer cell proliferation. Int J Cancer 2000; 86:644-51. [PMID: 10797285 DOI: 10.1002/(sici)1097-0215(20000601)86:5<644::aid-ijc7>3.0.co;2-t] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Over-expression of the ErbB-2 proto-oncogene frequently coincides with an aggressive clinical course of certain human adenocarcinomas. The ErbB-2 receptor is a member of the ErbB family of growth factor receptors, and within this complex signaling network, ErbB-2-containing heterodimers are preferentially formed. To assess whether ErbB-2 is a critical component in epidermal growth factor (EGF)-mediated stimulation of tumor cell proliferation, we used as a model SK-OV-3 ovarian cancer cells, which over-express EGF receptor (EGFR) and ErbB-2 receptors. In these cells, we reduced ErbB-2 mRNA and protein expression by transfection with ErbB-2-targeted hammerhead ribozymes and generated cell lines expressing different levels of ErbB-2. In SK-OV-3 cells, ErbB-2 expression conferred a growth advantage and soft agar experiments revealed that ErbB-2 was rate-limiting for anchorage-independent growth. The induction of colony formation by EGF was completely abrogated in ErbB-2-depleted cells, despite unchanged expression levels and tyrosine phosphorylation of the EGFR. The duration of EGF-mediated c-Fos mRNA up-regulation was decreased in parallel with loss of ErbB-2 expression. Furthermore, the rate of spontaneous apoptosis was increased in ErbB-2-depleted cells. Our results demonstrate that in human ovarian cancer cells the EGFR-ErbB-2 heterodimer, and not the EGFR homodimer, can be rate-limiting for EGF-mediated proliferation, thus suggesting that the oncogenic activity of ErbB-2 in human tumors is due in part to its ability to increase the growth response to stroma-derived EGF-like growth factors.
Collapse
Affiliation(s)
- S S Hsieh
- Department of Pharmacology and Toxicology, Philipps-University Marburg, Germany
| | | | | | | |
Collapse
|
50
|
Harris VK, Coticchia CM, Kagan BL, Ahmad S, Wellstein A, Riegel AT. Induction of the angiogenic modulator fibroblast growth factor-binding protein by epidermal growth factor is mediated through both MEK/ERK and p38 signal transduction pathways. J Biol Chem 2000; 275:10802-11. [PMID: 10753873 DOI: 10.1074/jbc.275.15.10802] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Fibroblast growth factor-binding protein (FGF-BP) is a secreted protein that binds and activates fibroblast growth factors (FGF-1 and FGF-2) and induces angiogenesis in some human cancers. FGF-BP is expressed at high levels in squamous cell carcinoma (SCC) cell lines and tumor samples and has been shown to be rate-limiting for the growth of SCC tumors in vivo. In this study, we examine the regulation of FGF-BP by epidermal growth factor (EGF) and the signal transduction mechanisms that mediate this effect. We found that EGF treatment of the ME-180 SCC cell line caused a rapid induction of FGF-BP gene expression. This induction was mediated transcriptionally through the AP-1 (c-Fos/JunD) and CCAAT/enhancer-binding protein elements as well as through an E-box repressor site in the proximal regulatory region of the FGF-BP promoter. Pharmacological inhibition of protein kinase C and mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 (MEK1/2) completely blocked EGF induction of FGF-BP mRNA, whereas inhibition of phosphatidylinositol 3-kinase had no effect. Additionally, both EGF- and anisomycin-induced FGF-BP mRNA was abrogated by inhibition of p38 mitogen-activated protein kinase, demonstrating a role for p38 in the regulation of FGF-BP. Co-transfection of the FGF-BP promoter with dominant negative forms of MEK2, extracellular signal-regulated kinase 2, and p38 significantly decreased the level of EGF induction, whereas expression of a dominant negative c-Jun N-terminal kinase mutant or expression of c-Jun N-terminal kinase inhibitory protein had no effect. Similarly, activation of the p38 pathway by overexpression of wild-type p38 or MKK6 enhanced FGF-BP transcription. These results demonstrate that EGF induction of FGF-BP occurs selectively through dual activation of the stress-activated p38 and the MEK/extracellular signal-regulated kinase mitogen-activated protein kinase pathways, which ultimately leads to activation of the promoter through AP-1 and CCAAT/enhancer-binding protein sites.
Collapse
Affiliation(s)
- V K Harris
- Department of Oncology, Vincent T. Lombardi Cancer Center, Georgetown University, Washington, D.C. 20007, USA
| | | | | | | | | | | |
Collapse
|