1
|
Yu B, Zhu HD, Shi XL, Chen PP, Sun XM, Xia GY, Fang M, Zhong YX, Tang XL, Zhang T, Pan HT. iTRAQ-based quantitative proteomic analysis of thoracic aortas from adult rats born to preeclamptic dams. Clin Proteomics 2021; 18:22. [PMID: 34418970 PMCID: PMC8379584 DOI: 10.1186/s12014-021-09327-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/09/2021] [Indexed: 01/04/2023] Open
Abstract
Background Preeclampsia and gestational hypertension can cause vascular function impairment in offspring. In our previous work, we described the protein expression profiles of umbilical artery tissues from patients with preeclampsia. Methods To gain insights into the mechanisms of vascular dysfunction in adult rats born to preeclamptic dams, we analyzed thoracic aorta tissues by using iTRAQ isobaric tags and 2D nano LC-MS/MS. Results By using the iTRAQ method, we analyzed 1825 proteins, of which 106 showed significantly different expression in the thoracic aortic. Ingenuity pathway analysis (IPA) showed that the majority of differentially expressed proteins (DEPs) were associated with cardiovascular function. Further analysis indicated that glucose-6-phosphate dehydrogenase (G6PD), which is inhibited by miR-423-5p and activated by TP53, had the strongest effect on cardiovascular function. The expression of G6PD was upregulated in thoracic aorta tissues, as confirmed by Western blotting. The expression of two other vascular function-related proteins, cysteine- and glycine-rich protein 2 (CSRP2) and tubulin alpha-4 A (TUBA4A), was upregulated, as demonstrated by mass spectrometry (MS). Conclusions Although the results require further functional validation, these data provide novel findings related to vascular function impairment in the adult offspring of preeclamptic mothers. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09327-9.
Collapse
Affiliation(s)
- Bin Yu
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Hong-Dan Zhu
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Xiao-Liang Shi
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Pan-Pan Chen
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Xiang-Mei Sun
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Gui-Yu Xia
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Min Fang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Yong-Xing Zhong
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Xiao-Li Tang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Tao Zhang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China. .,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.
| | - Hai-Tao Pan
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China. .,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.
| |
Collapse
|
2
|
Wang W, LeBlanc ME, Chen X, Chen P, Ji Y, Brewer M, Tian H, Spring SR, Webster KA, Li W. Pathogenic role and therapeutic potential of pleiotrophin in mouse models of ocular vascular disease. Angiogenesis 2017; 20:479-492. [PMID: 28447229 DOI: 10.1007/s10456-017-9557-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/18/2017] [Indexed: 01/06/2023]
Abstract
Angiogenic factors play an important role in the pathogenesis of diabetic retinopathy (DR), neovascular age-related macular degeneration (nAMD) and retinopathy of prematurity (ROP). Pleiotrophin, a well-known angiogenic factor, was recently reported to be upregulated in the vitreous fluid of patients with proliferative DR (PDR). However, its pathogenic role and therapeutic potential in ocular vascular diseases have not been defined in vivo. Here using corneal pocket assays, we demonstrated that pleiotrophin induced angiogenesis in vivo. To investigate the pathological role of pleiotrophin we used neutralizing antibody to block its function in multiple in vivo models of ocular vascular diseases. In a mouse model of DR, intravitreal injection of pleiotrophin-neutralizing antibody alleviated diabetic retinal vascular leakage. In a mouse model of oxygen-induced retinopathy (OIR), which is a surrogate model of ROP and PDR, we demonstrated that intravitreal injection of anti-pleiotrophin antibody prevented OIR-induced pathological retinal neovascularization and aberrant vessel tufts. Finally, pleiotrophin-neutralizing antibody ameliorated laser-induced choroidal neovascularization, a mouse model of nAMD, suggesting that pleiotrophin is involved in choroidal vascular disease. These findings suggest that pleiotrophin plays an important role in the pathogenesis of DR with retinal vascular leakage, ROP with retinal neovascularization and nAMD with choroidal neovascularization. The results also support pleiotrophin as a promising target for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Weiwen Wang
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Michelle E LeBlanc
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Xiuping Chen
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,Department of Ophthalmology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Ping Chen
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,Department of Ophthalmology, Renji Hospital of Jiaotong University, Shanghai, China
| | - Yanli Ji
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,Department of Ophthalmology, Zhengzhou Eye Hospital, Zhengzhou, Henan, China
| | - Megan Brewer
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Hong Tian
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA.,School of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Samantha R Spring
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Keith A Webster
- Vascular Biology Institute, University of Miami School of Medicine, Miami, FL, USA
| | - Wei Li
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL, USA. .,Vascular Biology Institute, University of Miami School of Medicine, Miami, FL, USA.
| |
Collapse
|
3
|
Dianat N, Le Viet B, Gobbo E, Auger N, Bièche I, Bennaceur-Griscelli A, Griscelli F. Midkine lacking its last 40 amino acids acts on endothelial and neuroblastoma tumor cells and inhibits tumor development. Mol Cancer Ther 2014; 14:213-24. [PMID: 25492619 DOI: 10.1158/1535-7163.mct-14-0226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Midkine (MDK) is a member of a new family of neurotrophic factors considered as rate-limiting growth and angiogenic factors implicated in the onset, invasion, and metastatic process of neuronal tumors, including neuroblastoma. We showed that all neuroblastoma cell lines highly expressed MDK, indicating that it is a critical player in tumor development, which may henceforth represent an attractive therapeutic target. We showed that the knockdown of MDK expression by siRNA led to a marked and significant decrease in neuroblastoma (IGR-N91 and SH-SY5Y) cell proliferation in vitro. Using a new strategy, we then evaluated the antitumor effect of a truncated MDK protein, lacking the C-terminal 81-121 portion of the molecule (MDKΔ81-121), which may act as a dominant-negative effector for its mitogenic, angiogenic, and tumorigenic activities by heterodimerizing with the wild-type protein. In vitro studies showed that MDKΔ81-121 selectively inhibited MDK-dependent tumor cells and was able to strongly reduce endothelial cell proliferation and migration and to induce ER stress-mediated apoptosis. We then investigated the effects of MDKΔ81-121 in vivo using electrotransfer of a plasmid encoding a secretable form of MDKΔ81-121 into tibialis cranialis muscles of nude mice. We showed that MDKΔ81-121 dramatically inhibited (up to 91%) tumor development and growth. This inhibition was correlated with the detection of the MDKΔ81-121 molecule in plasma and the suppression of intratumor neovascularization. Our findings demonstrate that MDK inhibition is a tractable therapeutic target for this lethal pediatric malignancy.
Collapse
Affiliation(s)
- Noushin Dianat
- INSERM U745, Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Barbara Le Viet
- INSERM U745, Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Emilie Gobbo
- INSERM U935, Human Embryonic Stem Cell Core Facility, Villejuif, France
| | - Nathalie Auger
- Départements de Biologie et Pathologie Médicale, Institute Gustave-Roussy, Villejuif, France
| | - Ivan Bièche
- Laboratoire d'Oncogénétique, Institut Curie, Hôpital René Huguenin, St-Cloud, France. Université Paris Descartes, Sorbonne Paris Cité, Faculté de Sciences Pharmaceutiques et Biologiques, Paris, France
| | - Annelise Bennaceur-Griscelli
- INSERM U935, Human Embryonic Stem Cell Core Facility, Villejuif, France. Université Paris-Sud 11, Campus Orsay, Orsay, France
| | - Frank Griscelli
- INSERM U935, Human Embryonic Stem Cell Core Facility, Villejuif, France. Départements de Biologie et Pathologie Médicale, Institute Gustave-Roussy, Villejuif, France. Université Paris Descartes, Sorbonne Paris Cité, Faculté de Sciences Pharmaceutiques et Biologiques, Paris, France.
| |
Collapse
|
4
|
Dos Santos C, Blanc C, Elahouel R, Prescott M, Carpentier G, Ori A, Courty J, Hamma-Kourbali Y, Fernig DG, Delbé J. Proliferation and migration activities of fibroblast growth factor-2 in endothelial cells are modulated by its direct interaction with heparin affin regulatory peptide. Biochimie 2014; 107 Pt B:350-7. [PMID: 25315978 DOI: 10.1016/j.biochi.2014.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 10/02/2014] [Indexed: 11/15/2022]
Abstract
Angiogenesis is the physiological process involving the growth of new blood vessels from pre-existing vessels. In normal or pathological angiogenesis, angiogenic growth factors activate cognate receptors on endothelial cells. Fibroblast growth factor-2 (FGF-2) and heparin affin regulatory peptide (HARP) are two heparin-binding growth factors and were described for their pro-angiogenic properties on human umbilical endothelial cells (HUVEC). We now show that HARP acts as a mediator of FGF-2's stimulatory effects, since it is able to inhibit the proliferation and migration of HUVEC induced by FGF-2. We demonstrate by ELISA and optical biosensor binding assay that HARP and FGF-2 interact through direct binding. We have adapted a previously developed structural proteomics method for the identification of residues involved in protein-protein interactions. Application of this method showed that two sequences in HARP were involved in binding FGF-2. One was in the C-thrombospondin type 1 repeat (C-TSR-1) domain and the other in the C-terminal domain of HARP. The identification of these regions as mediating the binding of FGF-2 was confirmed by ELISA using synthetic peptides, which are as well mediators of FGF-2-induced proliferation, migration and tubes formation on HUVEC in vitro. These results imply that besides a regulation of the proliferation, migration and angiogenesis of HUVEC by direct interaction of FGF-2 with its receptors, an alternative pathway exists involving its binding to growth factors such as HARP.
Collapse
Affiliation(s)
- Célia Dos Santos
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France.
| | - Charly Blanc
- IMRB INSERM, U955, Equipe 07, Faculté de Médecine, 8 rue du Général Sarrail, 94010 Créteil, France
| | - Rania Elahouel
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - Mark Prescott
- Department of Structural and Chemical Biology, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool L69 7ZB, UK
| | - Gilles Carpentier
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - Alessandro Ori
- Department of Structural and Chemical Biology, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool L69 7ZB, UK
| | - José Courty
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - Yamina Hamma-Kourbali
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | - David G Fernig
- Department of Structural and Chemical Biology, Institute of Integrative Biology, Biosciences Building, University of Liverpool, Liverpool L69 7ZB, UK
| | - Jean Delbé
- Laboratoire CRRET, CNRS, Université Paris Est, Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| |
Collapse
|
5
|
Mikelis C, Lamprou M, Koutsioumpa M, Koutsioubas AG, Spyranti Z, Zompra AA, Spiliopoulos N, Vradis AA, Katsoris P, Spyroulias GA, Cordopatis P, Courty J, Papadimitriou E. A peptide corresponding to the C-terminal region of pleiotrophin inhibits angiogenesis in vivo and in vitro. J Cell Biochem 2011; 112:1532-43. [PMID: 21344482 DOI: 10.1002/jcb.23066] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pleiotrophin (PTN) is a heparin-binding growth factor that plays a significant role in tumor growth and angiogenesis. We have previously shown that in order for PTN to induce migration of endothelial cells, binding to both α(ν) β(3) integrin and its receptor protein tyrosine phosphatase beta/zeta (RPTPβ/ζ) is required. In the present study we show that a synthetic peptide corresponding to the last 25 amino acids of the C-terminal region of PTN (PTN(112-136) ) inhibited angiogenesis in the in vivo chicken embryo chorioallantoic membrane (CAM) assay and PTN-induced migration and tube formation of human endothelial cells in vitro. PTN(112-136) inhibited binding of PTN to α(ν) β(3) integrin, and as shown by surface plasmon resonance (SPR) measurements, specifically interacted with the specificity loop of the extracellular domain of β(3) . Moreover, it abolished PTN-induced FAK Y397 phosphorylation, similarly to the effect of a neutralizing α(ν) β(3) -selective antibody. PTN(112-136) did not affect binding of PTN to RPTPβ/ζ in endothelial cells and induced β(3) Y773 phosphorylation and ERK1/2 activation to a similar extent with PTN. This effect was inhibited by down-regulation of RPTPβ/ζ by siRNA or by c-src inhibition, suggesting that PTN(112-136) may interact with RPTPβ/ζ. NMR spectroscopy studies showed that PTN(112-136) was characterized by conformational flexibility and absence of any element of secondary structure at room temperature, although the biologically active peptide segment 123-132 may adopt a defined structure at lower temperature. Collectively, our data suggest that although PTN(112-136) induces some of the signaling pathways triggered by PTN, it inhibits PTN-induced angiogenic activities through inhibition of PTN binding to α(ν) β(3) integrin.
Collapse
Affiliation(s)
- Constantinos Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
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
|
7
|
Diamantopoulou Z, Bermek O, Polykratis A, Hamma-Kourbali Y, Delbé J, Courty J, Katsoris P. A Pleiotrophin C-terminus peptide induces anti-cancer effects through RPTPβ/ζ. Mol Cancer 2010; 9:224. [PMID: 20738847 PMCID: PMC2936342 DOI: 10.1186/1476-4598-9-224] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 08/25/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pleiotrophin, also known as HARP (Heparin Affin Regulatory Peptide) is a growth factor expressed in various tissues and cell lines. Pleiotrophin participates in multiple biological actions including the induction of cellular proliferation, migration and angiogenesis, and is involved in carcinogenesis. Recently, we identified and characterized several pleiotrophin proteolytic fragments with biological activities similar or opposite to that of pleiotrophin. Here, we investigated the biological actions of P(122-131), a synthetic peptide corresponding to the carboxy terminal region of this growth factor. RESULTS Our results show that P(122-131) inhibits in vitro adhesion, anchorage-independent proliferation, and migration of DU145 and LNCaP cells, which express pleiotrophin and its receptor RPTPβ/ζ. In addition, P(122-131) inhibits angiogenesis in vivo, as determined by the chicken embryo CAM assay. Investigation of the transduction mechanisms revealed that P(122-131) reduces the phosphorylation levels of Src, Pten, Fak, and Erk1/2. Finally, P(122-131) not only interacts with RPTPβ/ζ, but also interferes with other pleiotrophin receptors, as demonstrated by selective knockdown of pleiotrophin or RPTPβ/ζ expression with the RNAi technology. CONCLUSIONS In conclusion, our results demonstrate that P(122-131) inhibits biological activities that are related to the induction of a transformed phenotype in PCa cells, by interacing with RPTPβ/ζ and interfering with other pleiotrophin receptors. Cumulatively, these results indicate that P(122-131) may be a potential anticancer agent, and they warrant further study of this peptide.
Collapse
Affiliation(s)
- Zoi Diamantopoulou
- Division of Genetics, Cell and Developmental Biology, Department of Biology, University of Patras, Greece
| | | | | | | | | | | | | |
Collapse
|
8
|
Santos CD, Karaky R, Renoir D, Hamma-Kourbali Y, Albanese P, Gobbo E, Griscelli F, Opolon P, Dalle S, Perricaudet M, Courty J, Delbé J. Antitumorigenic effects of a mutant of the heparin affin regulatory peptide on the U87 MG glioblastoma cell line. Int J Cancer 2009; 127:1038-51. [DOI: 10.1002/ijc.25110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
9
|
Degoutin J, Brunet-de Carvalho N, Cifuentes-Diaz C, Vigny M. ALK (Anaplastic Lymphoma Kinase) expression in DRG neurons and its involvement in neuron-Schwann cells interaction. Eur J Neurosci 2009; 29:275-86. [PMID: 19200234 DOI: 10.1111/j.1460-9568.2008.06593.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) transiently expressed in specific regions of the central and peripheral nervous systems. In this study, we focused on the rat developing dorsal root ganglion (DRG). This ganglion is composed of heterogeneous sensory neurons characterized by the expression of RTK for neurotrophic factors, such as the nerve growth factor receptor TrkA or the glial-derived neurotrophic factor family receptor Ret, which are specifically detected in nociceptive neurons. In DRG, ALK expression reached a maximum around birth. We showed that ALK is specifically present in a subtype of neurons during DRG development, and that the majority of these neurons co-expressed TrkA and Ret. Interestingly, we identified only one form (220 kDa) of ALK in DRG neurons both in vivo and in vitro. On the opposite, in transfected cells as well as in brain extracts, ALK was identified as two forms (220 and 140 kDa). The DRG is composed of neurons and glial cells, principally satellite Schwann cells. Thus, we hypothesized that the presence of satellite Schwann cells was involved in the absence of truncated ALK. Using two different cell types, HEK293 cells stably expressing ALK, and MSC80 cells, a previously described Schwann cell line, we showed that a factor secreted by the Schwann cells is likely involved in the absence of ALK cleavage. All these data hence open new perspectives concerning the role of ALK in the specification of nociceptive DRG neurons and in the neurons-Schwann cells interaction.
Collapse
Affiliation(s)
- Joffrey Degoutin
- UMR_S839 INSERM/UPMC, Institut du Fer à Moulin (IFM), 17 rue du Fer à Moulin, Paris, France
| | | | | | | |
Collapse
|
10
|
Webb TR, Slavish J, George RE, Look AT, Xue L, Jiang Q, Cui X, Rentrop WB, Morris SW. Anaplastic lymphoma kinase: role in cancer pathogenesis and small-molecule inhibitor development for therapy. Expert Rev Anticancer Ther 2009; 9:331-56. [PMID: 19275511 DOI: 10.1586/14737140.9.3.331] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Anaplastic lymphoma kinase (ALK), a receptor tyrosine kinase in the insulin receptor superfamily, was initially identified in constitutively activated oncogenic fusion forms - the most common being nucleophosmin-ALK - in anaplastic large-cell lymphomas, and subsequent studies have identified ALK fusions in diffuse large B-cell lymphomas, systemic histiocytosis, inflammatory myofibroblastic tumors, esophageal squamous cell carcinomas and non-small-cell lung carcinomas. More recently, genomic DNA amplification and protein overexpression, as well as activating point mutations, of ALK have been described in neuroblastomas. In addition to those cancers for which a causative role for aberrant ALK activity is well validated, more circumstantial links implicate the full-length, normal ALK receptor in the genesis of other malignancies - including glioblastoma and breast cancer - via a mechanism of receptor activation involving autocrine and/or paracrine growth loops with the reported ALK ligands, pleiotrophin and midkine. This review summarizes normal ALK biology, the confirmed and putative roles of ALK in the development of human cancers and efforts to target ALK using small-molecule kinase inhibitors.
Collapse
Affiliation(s)
- Thomas R Webb
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, 332 North Lauderdale Street, Mail Stop 1000, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Ducès A, Karaky R, Martel-Renoir D, Mir L, Hamma-Kourbali Y, Biéche I, Opolon P, Delbé J, Courty J, Perricaudet M, Griscelli F. 16-kDa fragment of pleiotrophin acts on endothelial and breast tumor cells and inhibits tumor development. Mol Cancer Ther 2008; 7:2817-27. [PMID: 18790762 DOI: 10.1158/1535-7163.mct-08-0301] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pleiotrophin (PTN) is a 136-amino acid secreted heparin-binding protein that is considered as a rate-limiting growth and an angiogenic factor in the onset, invasion, and metastatic process of many tumors. Its mitogenic and tumorigenic activities are mediated by the COOH-terminal residues 111 to 136 of PTN, allowing it to bind to cell surface tyrosine kinase-linked receptors. We investigated a new strategy consisting in evaluating the antitumor effect of a truncated PTN, lacking the COOH-terminal 111 to 136 portion of the molecule (PTNDelta111-136), which may act as a dominant-negative effector for its mitogenic, angiogenic, and tumorigenic activities by heterodimerizing with the wild-type protein. In vitro studies showed that PTNDelta111-136 selectively inhibited a PTN-dependent MDA-MB-231 breast tumor and endothelial cell proliferation and that, in MDA-MB-231 cells expressing PTNDelta111-136, the vascular endothelial growth factor-A and hypoxia-inducible factor-1alpha mRNA levels were significantly decreased by 59% and 71%, respectively, compared with levels in wild-type cells. In vivo, intramuscular electrotransfer of a plasmid encoding a secretable form of PTNDelta111-136 was shown to inhibit MDA-MB-231 tumor growth by 81%. This antitumor effect was associated with the detection of the PTNDelta111-136 molecule in the muscle and tumor extracts, the suppression of neovascularization within the tumors, and a decline in the Ki-67 proliferative index. Because PTN is rarely found in normal tissue, our data show that targeted PTN may represent an attractive and new therapeutic approach to the fight against cancer.
Collapse
Affiliation(s)
- Aurélie Ducès
- INSERM U745, Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris V, 4 avenue de l'Observatoire, 75006 Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Pleiotrophin, a multifunctional angiogenic factor: mechanisms and pathways in normal and pathological angiogenesis. Curr Opin Hematol 2008; 15:210-4. [PMID: 18391787 DOI: 10.1097/moh.0b013e3282fdc69e] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW This study seeks to integrate recent studies that identify new critical mechanisms through which the 136 amino acid secreted heparin-binding cytokine pleiotrophin (PTN, Ptn) stimulates both normal and pathological angiogenesis. RECENT FINDINGS Pleiotrophin is directly angiogenic; it initiates an angiogenic switch in different cancer models in vivo. It acts as an angiogenic factor through multiple mechanisms that include a unique signaling pathway that activates newly identified downstream tyrosine kinases through a unique mechanism, an interaction with endothelial cells to initiate proliferation, migration, and tube formation, the regulation of basic fibroblast growth factor and vascular endothelial growth factor signaling, the remodeling of the stromal microenvironment, and induction of transdifferentiation of monocytes into endothelial cells. Recently also, domains of PTN that stimulate angiogenesis and peptides that function to inhibit PTN signaling have been identified. SUMMARY Recent studies have identified new mechanisms dependent on activation of the PTN signaling pathway that regulate angiogenesis and new targets to use PTN to both stimulate angiogenesis and block its activity to control pathological angiogenesis.
Collapse
|
13
|
Destouches D, El Khoury D, Hamma-Kourbali Y, Krust B, Albanese P, Katsoris P, Guichard G, Briand JP, Courty J, Hovanessian AG. Suppression of tumor growth and angiogenesis by a specific antagonist of the cell-surface expressed nucleolin. PLoS One 2008; 3:e2518. [PMID: 18560571 PMCID: PMC2424174 DOI: 10.1371/journal.pone.0002518] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Accepted: 05/20/2008] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Emerging evidences suggest that nucleolin expressed on the cell surface is implicated in growth of tumor cells and angiogenesis. Nucleolin is one of the major proteins of the nucleolus, but it is also expressed on the cell surface where is serves as a binding protein for variety of ligands implicated in cell proliferation, differentiation, adhesion, mitogenesis and angiogenesis. METHODOLOGY/PRINCIPAL FINDINGS By using a specific antagonist that binds the C-terminal tail of nucleolin, the HB-19 pseudopeptide, here we show that the growth of tumor cells and angiogenesis are suppressed in various in vitro and in vivo experimental models. HB-19 inhibited colony formation in soft agar of tumor cell lines, impaired migration of endothelial cells and formation of capillary-like structures in collagen gel, and reduced blood vessel branching in the chick embryo chorioallantoic membrane. In athymic nude mice, HB-19 treatment markedly suppressed the progression of established human breast tumor cell xenografts in nude mice, and in some cases eliminated measurable tumors while displaying no toxicity to normal tissue. This potent antitumoral effect is attributed to the direct inhibitory action of HB-19 on both tumor and endothelial cells by blocking and down regulating surface nucleolin, but without any apparent effect on nucleolar nucleolin. CONCLUSION/SIGNIFICANCE Our results illustrate the dual inhibitory action of HB-19 on the tumor development and the neovascularization process, thus validating the cell-surface expressed nucleolin as a strategic target for an effective cancer drug. Consequently, the HB-19 pseudopeptide provides a unique candidate to consider for innovative cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Bernard Krust
- CNRS UPR 2228, Université Paris Descartes, Paris, France
| | | | | | - Gilles Guichard
- CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Jean Paul Briand
- CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - José Courty
- CNRS UMR 7149, Université Paris-Est, Créteil, France
| | | |
Collapse
|
14
|
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
|
15
|
Bermek O, Diamantopoulou Z, Polykratis A, Dos Santos C, Hamma-Kourbali Y, Burlina F, Delbé J, Chassaing G, Fernig DG, Katsoris P, Courty J. A basic peptide derived from the HARP C-terminus inhibits anchorage-independent growth of DU145 prostate cancer cells. Exp Cell Res 2007; 313:4041-50. [PMID: 17727841 DOI: 10.1016/j.yexcr.2007.07.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 07/31/2007] [Accepted: 07/31/2007] [Indexed: 12/27/2022]
Abstract
Heparin affin regulatory peptide (HARP) is an 18 kDa heparin-binding protein that plays a key role in tumor growth. We showed previously that the synthetic peptide P(111-136) composed of the last 26 HARP amino acids inhibited HARP-induced mitogenesis. Here, to identify the exact molecular domain involved in HARP inhibition, we investigated the effect of the shorter basic peptide P(122-131) on DU145 cells, which express HARP and its receptor protein tyrosine phosphatase beta/zeta (RPTPbeta/zeta). P(122-131) was not cytotoxic; it dose-dependently inhibited anchorage-independent growth of DU145 cells. Binding studies using biotinylated P(122-131) indicated that this peptide interfered with HARP binding to DU145 cells. Investigation of the mechanisms involved suggested interference, under anchorage-independent conditions, of P(122-131) with a HARP autocrine loop in an RPTPbeta/zeta-dependent fashion. Thus, P(122-131) may hold potential for the treatment of disorders involving RPTPbeta/zeta.
Collapse
Affiliation(s)
- Oya Bermek
- Laboratoire de recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), CNRS UMR 7149, Université Paris 12, 61 Avenue du Général de Gaulle, 94010 Créteil Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Hamma-Kourbali Y, Bernard-Pierrot I, Heroult M, Dalle S, Caruelle D, Milhiet PE, Fernig DG, Delbé J, Courty J. Inhibition of the mitogenic, angiogenic and tumorigenic activities of pleiotrophin by a synthetic peptide corresponding to its C-thrombospondin repeat-I domain. J Cell Physiol 2007; 214:250-9. [PMID: 17607711 DOI: 10.1002/jcp.21191] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pleiotrophin (PTN), is a heparin-dependent growth factor involved in angiogenesis and tumor growth. PTN contains a thrombospondin repeat-I (TSR-I) motif in its two beta-sheet domains that are involved in its binding to heparin and its neurite outgrowth activity. Based on the importance of the binding of PTN to heparin in its dimerization and biological activities, we have designed two synthetic peptides, P(13-39) and P(65-97) corresponding to a part of the N-terminal and C-terminal TSR-I motif of PTN, respectively. P(65-97) inhibited the mitogenic, tumorigenic and angiogenic activities of PTN, as well as the mitogenic and an angiogenic activity of fibroblast growth factor-2 (FGF-2). However, P(65-97) had no effect on the mitogenic activity of epidermal growth factor, which does not bind heparin. P(65-97) but not P(13-39) inhibited the binding of PTN and to a lesser extent of FGF-2 to heparin using an immunoassay and an optical biosensor assay and bound directly to heparin with a K(d) of 120 nM. These findings suggest that P(65-97), containing amino acids 65-97 of the TSR-I motif of the C-terminal domain of PTN, inhibits the activities of PTN and FGF-2 by virtue of its ability to bind heparin very effectively and so compete with the growth factors for their polysaccharide co-receptor.
Collapse
Affiliation(s)
- Yamina Hamma-Kourbali
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), CNRS UMR 7149, Université Paris XII, Créteil Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Hatziapostolou M, Delbe J, Katsoris P, Polytarchou C, Courty J, Papadimitriou E. Heparin affin regulatory peptide is a key player in prostate cancer cell growth and angiogenicity. Prostate 2005; 65:151-8. [PMID: 15924335 DOI: 10.1002/pros.20270] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The development and growth of human prostate cancer is mediated by many tumor cell-derived growth factors. Heparin affin regulatory peptide (HARP) seems to be involved in the progression of several tumors of diverse origin. In the present study, we sought to determine if HARP is implicated in human prostate cancer. METHODS An antisense strategy for inhibition of HARP expression in the human prostate cancer cell line LNCaP was used to study the role of HARP on cancer cell growth, migration, and angiogenic potential in vitro and in vivo. RESULTS Exogenous human recombinant HARP was mitogenic for LNCaP cells. By decreasing the expression of endogenous HARP, we found that HARP was essential for LNCaP cell migration, as well as anchorage-dependent and independent growth. Endothelial cell functions in vitro and blood vessel formation in vivo induced by LNCaP cells were also inhibited when HARP expression was diminished. CONCLUSIONS HARP seems to act as an important regulator of diverse biological activities in human prostate cancer cells.
Collapse
Affiliation(s)
- Maria Hatziapostolou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Greece
| | | | | | | | | | | |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Polykratis A, Delbé J, Courty J, Papadimitriou E, Katsoris P. Identification of heparin affin regulatory peptide domains with potential role on angiogenesis. Int J Biochem Cell Biol 2004; 36:1954-66. [PMID: 15203110 DOI: 10.1016/j.biocel.2004.02.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2003] [Revised: 02/12/2004] [Accepted: 02/13/2004] [Indexed: 11/23/2022]
Abstract
Heparin affin regulatory peptide (HARP) is a growth factor displaying high affinity for heparin. It is present in the extracellular matrix of many tissues, interacting with heparan sulfate and dermatan/chondroitin sulfate glycosaminoglycans. We have previously shown that HARP is implicated in the control of angiogenesis and its effects are mimicked, at least in part, by synthetic peptides that correspond to its N and C termini. In the present work, we show that HARP is cleaved by plasmin, leading to the production of five peptides that correspond to distinct domains of the molecule. Heparin, heparan sulfate and dermatan sulfate, at various HARP to glycosaminoglycan ratios, partially protect HARP from plasmin degradation. The molecules with higher affinity to HARP are the more protective, heparin being the most efficient. The peptides that are produced from cleavage of HARP by plasmin, affect in vivo and in vitro angiogenesis and modulate the angiogenic activity of vascular endothelial growth factor on human umbilical vein endothelial cells. Similar results were obtained in vitro with recombinant HARP peptides, identical to the peptides generated after treatment of HARP with plasmin. These results suggest that different regions of HARP may induce or inhibit angiogenesis.
Collapse
Affiliation(s)
- Apostolos Polykratis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, GR 26504, Greece
| | | | | | | | | |
Collapse
|
22
|
Bernard-Pierrot I, Delbé J, Heroult M, Rosty C, Soulié P, Barritault D, Milhiet PE, Courty J. Heparin affin regulatory peptide in milk: its involvement in mammary gland homeostasis. Biochem Biophys Res Commun 2004; 314:277-82. [PMID: 14715276 DOI: 10.1016/j.bbrc.2003.12.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
HARP (heparin affin regulatory peptide) is a heparin binding growth factor implicated in cellular growth and differentiation. Previously, HARP had been localized in the human mammary, in both alveolar epithelial and myoepithelial cells although HARP mRNAs were only expressed by myoepithelial cells [J. Histochem. Cytochem. 45 (1997) 1]. In the present study, we demonstrate that HARP is secreted in human mature milk with concentrations ranging from 17.68+/-6.4ng/ml in mature milk to 59.9+/-11.22ng/ml in colostrum. In vitro, HARP was found to be mitogenic on human mammary epithelial and myoepithelial cell lines and correlated with the expression of its high affinity receptor tyrosine kinase ALK (anaplastic lymphoma kinase). In vivo, ALK is expressed in both mammary epithelial and myoepithelial cells, suggesting that HARP could act in vivo as a paracrine and autocrine growth factor in the regulation of the mammary gland development and its homeostatic maintenance during pregnancy and lactation.
Collapse
Affiliation(s)
- Isabelle Bernard-Pierrot
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires FRE CNRS No. 2412, Université Paris Val de Marne Avenue du Général de Gaulle, Créteil, France
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Héroult M, Bernard-Pierrot I, Delbé J, Hamma-Kourbali Y, Katsoris P, Barritault D, Papadimitriou E, Plouet J, Courty J. Heparin affin regulatory peptide binds to vascular endothelial growth factor (VEGF) and inhibits VEGF-induced angiogenesis. Oncogene 2004; 23:1745-53. [PMID: 15001987 DOI: 10.1038/sj.onc.1206879] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Heparin affin regulatory peptide (HARP) is an heparin-binding molecule involved in the regulation of cell proliferation and differentiation. Here, we report that HARP inhibited the biological activity induced by the 165-amino-acid form of vascular endothelial growth factor (VEGF165) on human umbilical vein endothelial cells. Endothelial-cell proliferation induced by VEGF165 showed about 50% inhibition in the presence of HARP in a concentration of 3 nM. In similar range of concentrations, HARP blocked tube formation induced by VEGF165 in three-dimensional angiogenesis assay. In vivo studies showed that HARP inhibited the VEGF165-induced Matrigel trade mark infiltration of endothelial cells. We then investigated the mechanisms of this inhibition and shown that HARP inhibited the binding of 125I-VEGF165 to the VEGF receptors of endothelial cells. Additional studies using VEGF soluble receptors indicated that binding of 125I-VEGF165 to kinase insert domain-containing receptor and neuropilin receptor was inhibited by HARP, but conversely the binding of 125I-VEGF165 to fms-like tyrosine kinase I receptor was unaffected. A competitive affinity-binding assay demonstrated that HARP interacted directly with VEGF165 with a dissociation coefficient of 1.38 nM. Binding assay using deletion mutants of HARP revealed that the thrombospondin type-1 repeats domains were involved in this interaction. These data demonstrate for the first time that the angiogenic factor HARP can also negatively regulates the angiogenic activity of VEGF165.
Collapse
Affiliation(s)
- Mélanie Héroult
- Laboratoire de Recherche sur la Croissance Cellulaire, la Réparation et la Régénération Tissulaires (CRRET), FRE CNRS 2412, Université Paris XII-Val de Marne, Avenue du Général de Gaulle, 94010 Créteil, France
| | | | | | | | | | | | | | | | | |
Collapse
|