102 research outputs found

    A Pleiotrophin C-terminus peptide induces anti-cancer effects through RPTPβ/ζ

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>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.</p> <p>Results</p> <p>Our results show that P(122-131) inhibits <it>in vitro </it>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 <it>in vivo</it>, 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 Erk<sup>1</sup>/<sub>2</sub>. 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.</p> <p>Conclusions</p> <p>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.</p

    Pleiotrophin over-expression provides trophic support to dopaminergic neurons in parkinsonian rats

    Get PDF
    Background: Pleiotrophin is known to promote the survival and differentiation of dopaminergic neurons in vitro and is up-regulated in the substantia nigra of Parkinson's disease patients. To establish whether pleiotrophin has a trophic effect on nigrostriatal dopaminergic neurons in vivo, we injected a recombinant adenovirus expressing pleiotrophin in the substantia nigra of 6-hydroxydopamine lesioned rats. Results: The viral vector induced pleiotrophin over-expression by astrocytes in the substantia nigra pars compacta, without modifying endogenous neuronal expression. The percentage of tyrosine hydroxylase-immunoreactive cells as well as the area of their projections in the lesioned striatum was higher in pleiotrophin-treated animals than in controls. Conclusions: These results indicate that pleiotrophin over-expression partially rescues tyrosine hydroxylase-immunoreactive cell bodies and terminals of dopaminergic neurons undergoing 6-hydroxydopamine-induced degeneration

    Suppression of Tumor Growth and Angiogenesis by a Specific Antagonist of the Cell-Surface Expressed Nucleolin

    Get PDF
    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

    Tumor stiffening reversion through collagen crosslinking inhibition improves T cell migration and anti-PD-1 treatment

    Full text link
    Only a fraction of cancer patients benefits from immune checkpoint inhibitors. This may be partly due to the dense extracellular matrix (ECM) that forms a barrier for T cells. Comparing five preclinical mouse tumor models with heterogeneous tumor microenvironments, we aimed to relate the rate of tumor stiffening with the remodeling of ECM architecture and to determine how these features affect intratumoral T cell migration. An ECM-targeted strategy, based on the inhibition of lysyl oxidase, was used. In vivo stiffness measurements were found to be strongly correlated with tumor growth and ECM crosslinking but negatively correlated with T cell migration. Interfering with collagen stabilization reduces ECM content and tumor stiffness leading to improved T cell migration and increased efficacy of anti-PD-1 blockade. This study highlights the rationale of mechanical characterizations in solid tumors to understand resistance to immunotherapy and of combining treatment strategies targeting the ECM with anti-PD-1 therapy

    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

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>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 <it>in vitro </it>and <it>in vivo </it>the growth of a human tumour cell line PC-3 which possess an HARP autocrine loop.</p> <p>Methods</p> <p>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. <it>In vitro</it>, the P111-136 effect on HARP autocrine loop in PC-3 cells was determined by colony formation in soft agar. <it>In vivo</it>, 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. <it>In vivo </it>anti-angiogenic effect was confirmed using a mouse Matrigel™ plug assay.</p> <p>Results</p> <p>Using pull down experiments, we identified the HARP receptors RPTPβ/ζ, ALK and nucleolin as P111-136 binding proteins. <it>In vitro</it>, 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 <it>in vivo </it>Matrigel™ plug assay in mice</p> <p>Conclusions</p> <p>Our results demonstrate that P111-136 strongly inhibits the mitogenic effect of HARP on <it>in vitro </it>and <it>in vivo </it>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). <it>In vivo</it>, 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.</p

    Targeting surface nucleolin with a multivalent pseudopeptide delays development of spontaneous melanoma in RET transgenic mice

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>The importance of cell-surface nucleolin in cancer biology was recently highlighted by studies showing that ligands of nucleolin play critical role in tumorigenesis and angiogenesis. By using a specific antagonist that binds the C-terminal tail of nucleolin, the HB-19 pseudopeptide, we recently reported that HB-19 treatment markedly suppressed the progression of established human breast tumor cell xenografts in the athymic nude mice without apparent toxicity.</p> <p>Methods</p> <p>The <it>in vivo </it>antitumoral action of HB-19 treatment was assessed on the spontaneous development of melanoma in the RET transgenic mouse model. Ten days old RET mice were treated with HB-19 in a prophylactic setting that extended 300 days. In parallel, the molecular basis for the action of HB-19 was investigated on a melanoma cell line (called TIII) derived from a cutaneous nodule of a RET mouse.</p> <p>Results</p> <p>HB-19 treatment of RET mice caused a significant delay in the onset of cutaneous tumors, several-months delay in the incidence of large tumors, a lower frequency of cutaneous nodules, and a reduction of visceral metastatic nodules while displaying no toxicity to normal tissue. Moreover, microvessel density was significantly reduced in tumors recovered from HB-19 treated mice compared to corresponding controls. Studies on the melanoma-derived tumor cells demonstrated that HB-19 treatment of TIII cells could restore contact inhibition, impair anchorage-independent growth, and reduce their tumorigenic potential in mice. Moreover, HB-19 treatment caused selective down regulation of transcripts coding matrix metalloproteinase 2 and 9, and tumor necrosis factor-α in the TIII cells and in melanoma tumors of RET mice.</p> <p>Conclusions</p> <p>Although HB-19 treatment failed to prevent the development of spontaneous melanoma in the RET mice, it delayed for several months the onset and frequency of cutaneous tumors, and exerted a significant inhibitory effect on visceral metastasis. Consequently, HB-19 could provide a novel therapeutic agent by itself or as an adjuvant therapy in association with current therapeutic interventions on a virulent cancer like melanoma.</p

    Potentialisation des propriétés de cellules souches mésenchymateuses par des mimétiques de glycosaminoglycannes et leur application en thérapie osseuse en association à des biomatériaux.

    No full text
    Résumé français manquantScientific background: GAGs mimetics properties on regenerative process.Glycosaminoglycans (GAGs) are sulfated polysaccharides actually considered as major structural components of the extracellular matrix as well as regulators of cells functions during homeostatic and pathological processes. These GAGs activities are based on their ability to interact with heparin binding growth-factors (HBGF), chemokines and enzymes, to protect them from proteolytic degradation and to potentialyze their interaction with cell surface specific receptors and/or other components of the ECM. GAGs are characterized by their extensive structural diversity, based on the number and location of sulfate or acetylate groups, that would determine specific biological interactions.As comparative tool to study the relationship between the complexity of GAGs chemical structures and their biological functions, we used synthetic GAGs mimetics, derivate from a polymer of dextran and functionalized with carboxylate, sulfate and/or acetate groups. They are structurally and functionally related to natural heparan sulfates. These compounds improved both the rate and quality of regenerative process in numerous animal models of injury after topical treatment.Our hypothesize is that specific HS cooperative interactions with HBGF and ECM compounds could influence both therapeutic progenitors and stem cells properties by compartmentalizing them to specific microenvironment niches, and protecting them against deleterious signals. Such abilities to modulate stem cell biology could be a new way to explain and to take advantage of regenerative properties of these compounds. The principal aim of this work was to demonstrate the effects of GAGs mimetics on Mesenchymal Stem Cells (MSC) properties for application in bone repair. GAGs mimetics as new potentializing agents of mesenchymal stem cells propertiesDuring osteogenesis, a controlled expression of functional HS is required to interact and regulate the activity of growth promoting and osteogenic differentiation factors. However effects of GAGs on MSC properties remain to be analyzed. We focus on two GAGs mimetics leader molecules [OTR4131] and [OTR4120], with distinct chemical characteristics, since sulfated mimetic [OTR4120] was previously shown to stimulate bone repair in vivo. We demonstrate that its acetylated and sulfated counterpart [OTR4131] enhances proliferation, whereas [OTR4120] clearly stimulates migration and osteogenic differentiation properties of rat MSC in vitro, that could explain its bone regenerative effect in vivo. This indicates that GAGs mimetics would be of great interest for potential application in therapy, since according to their structural signature they could modulate specific activities of progenitors and stem cells, and represent an alternative to exogenous growth factor treatments. New matricial strategy for bone repair associating GAGs mimetics to biomaterials and human MSCCell based therapy associated to biomaterials for repair of bone defects are promising but not enough efficient. We proposed to develop matricial strategy, associating efficient micro-environment molecules such as GAGs mimetics, to optimize cell therapeutic approaches. First we validated that GAGs mimetics are effective on human MSC proliferation, migration and differentiation properties in vitro. We demonstrated that colonization efficiency of hydroxyapatite/b-tricalcium phosphate biomaterial scaffolds by human MSC was improved when scaffolds are functionalized with GAGs mimetics in vitro. Finally osteoformation in vivo was evaluated after ectopic transplantation of functionalized and/or cellularized biomaterials in nude mice: few effects were observed on bone formation, whereas osteoclastogenesis and vascularization were clearly modulated by GAGs mimetics immobilized. GAGs mimetics as new mobilizing agents of stem cells...PARIS-EST-Université (770839901) / SudocSudocFranceF
    corecore