Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 128(5): 918-927, 2023 03.
Article in English | MEDLINE | ID: mdl-36550208

ABSTRACT

BACKGROUND: Neuroendocrine prostate cancer (NEPC) is a multi-resistant variant of prostate cancer (PCa) that has become a major challenge in clinics. Understanding the neuroendocrine differentiation (NED) process at the molecular level is therefore critical to define therapeutic strategies that can prevent multi-drug resistance. METHODS: Using RNA expression profiling and immunohistochemistry, we have identified and characterised a gene expression signature associated with the emergence of NED in a large PCa cohort, including 169 hormone-naïve PCa (HNPC) and 48 castration-resistance PCa (CRPC) patients. In vitro and preclinical in vivo NED models were used to explore the cellular mechanism and to characterise the effects of castration on PCa progression. RESULTS: We show for the first time that Neuropilin-1 (NRP1) is a key component of NED in PCa cells. NRP1 is upregulated in response to androgen deprivation therapies (ADT) and elicits cell survival through induction of the PKC pathway. Downmodulation of either NRP1 protein expression or PKC activation suppresses NED, prevents tumour evolution toward castration resistance and increases the efficacy of docetaxel-based chemotherapy in preclinical models in vivo. CONCLUSIONS: This study reveals the NRP1/PKC axis as a promising therapeutic target for the prevention of neuroendocrine castration-resistant variants of PCa and indicates NRP1 as an early transitional biomarker.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/pathology , Neuropilin-1 , Prostatic Neoplasms, Castration-Resistant/pathology , Androgen Antagonists , Drug Resistance , Cell Differentiation , Cell Line, Tumor
2.
Int J Mol Sci ; 22(21)2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34768734

ABSTRACT

Prostate cancer is the most common cancer in men. For patients with advanced or metastatic prostate cancer, available treatments can slow down its progression but cannot cure it. The development of innovative drugs resulting from the exploration of biodiversity could open new therapeutic alternatives. Dermaseptin-B2, a natural multifunctional antimicrobial peptide isolated from Amazonian frog skin, has been reported to possess antitumor activity. To improve its pharmacological properties and to decrease its peripheral toxicity and lethality we developed a hormonotoxin molecule composed of dermaseptin-B2 combined with d-Lys6-LHRH to target the LHRH receptor. This hormonotoxin has a significant antiproliferative effect on the PC3 tumor cell line, with an IC50 value close to that of dermaseptin-B2. Its antitumor activity has been confirmed in vivo in a xenograft mouse model with PC3 tumors and appears to be better tolerated than dermaseptin-B2. Biophysical experiments showed that the addition of LHRH to dermaseptin-B2 did not alter its secondary structure or biological activity. The combination of different experimental approaches indicated that this hormonotoxin induces cell death by an apoptotic mechanism instead of necrosis, as observed for dermaseptin-B2. These results could explain the lower toxicity observed for this hormonotoxin compared to dermaseptin-B2 and may represent a promising targeting approach for cancer therapy.


Subject(s)
Amphibian Proteins/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cell Line, Tumor/drug effects , Amino Acid Sequence , Amphibian Proteins/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Immunologic Factors/metabolism , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Xenograft Model Antitumor Assays
3.
ChemMedChem ; 13(3): 259-269, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29314771

ABSTRACT

A member of the ribonuclease A superfamily, human angiogenin (hAng) is a potent angiogenic factor. Heteronuclear NMR spectroscopy combined with induced-fit docking revealed a dual binding mode for the most antiangiogenic compound of a series of ribofuranosyl pyrimidine nucleosides that strongly inhibit hAng's angiogenic activity in vivo. While modeling suggests the potential for simultaneous binding of the inhibitors at the active and cell-binding sites, NMR studies indicate greater affinity for the cell-binding site than for the active site. Additionally, molecular dynamics simulations at 100 ns confirmed the stability of binding at the cell-binding site with the predicted protein-ligand interactions, in excellent agreement with the NMR data. This is the first time that a nucleoside inhibitor is reported to completely inhibit the angiogenic activity of hAng in vivo by exerting dual inhibitory activity on hAng, blocking both the entrance of hAng into the cell and its ribonucleolytic activity.


Subject(s)
Pyrimidine Nucleosides/chemistry , Ribonuclease, Pancreatic/antagonists & inhibitors , Animals , Binding Sites , Cell Line , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Computer Simulation , Humans , Molecular Dynamics Simulation , Neovascularization, Physiologic/drug effects , Nuclear Magnetic Resonance, Biomolecular , Structure-Activity Relationship
4.
PLoS One ; 12(8): e0182926, 2017.
Article in English | MEDLINE | ID: mdl-28797092

ABSTRACT

Dermaseptin-B2 (DRS-B2) is a multifunctional cationic antimicrobial peptide (CAP) isolated from frog skin secretion. We previously reported that DRS-B2 possesses anticancer and antiangiogenic activities in vitro and in vivo. In the present study, we evaluated the antiproliferative activity of DRS-B2 on numerous tumor cell lines, its cell internalization and studies of its molecular partners as well as their influences on its structure. Confocal microscopy using ([Alexa594]-(Cys0)-DRS-B2) shows that in sensitive human tumor cells (PC3), DRS-B2 seems to accumulate rapidly at the cytoplasmic membranes and enters the cytoplasm and the nucleus, while in less sensitive tumor cells (U87MG), DRS-B2 is found packed in vesicles at the cell membrane. Furthermore FACS analysis shows that PC3 cells viability decreases after DRS-B2 treatment while U87 MG seems to be unaffected. However, "pull down" experiments performed with total protein pools from PC3 or U87MG cells and the comparison between the antiproliferative effect of DRS-B2 and its synthetic analog containing all D-amino acids suggest the absence of a stereo-selective protein receptor. Pretreatment of PC3 cells with sodium chlorate, decreases the antiproliferative activity of DRS-B2. This activity is partially restored after addition of exogenous chondroitin sulfate C (CS-C). Moreover, we demonstrate that at nanomolar concentrations CS-C potentiates the antiproliferative effect of DRS-B2. These results highlight the partial implication of glycosaminoglycans in the mechanism of antiproliferative action of DRS-B2. Structural analysis of DRS-B2 by circular dichroism in the presence of increasing concentration of CS-C shows that DRS-B2 adopts an α-helical structure. Finally, structure-activity-relationship studies suggest a key role of the W residue in position 3 of the DRS-B2 sequence for its antiproliferative activity.


Subject(s)
Amphibian Proteins/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Glycosaminoglycans/metabolism , Neoplasms/drug therapy , Amino Acid Sequence , Amphibian Proteins/chemistry , Animals , Antimicrobial Cationic Peptides/chemistry , Antineoplastic Agents/chemistry , Anura , Cell Line, Tumor , Humans , Neoplasms/metabolism , Structure-Activity Relationship
5.
Am J Pathol ; 186(2): 435-45, 2016 02.
Article in English | MEDLINE | ID: mdl-26687816

ABSTRACT

Activating mutations of anaplastic lymphoma kinase (ALK) have been identified as important players in neuroblastoma development. Our goal was to evaluate the significance of overall ALK activation in neuroblastoma. Expression of phosphorylated ALK, ALK, and its putative ligands, pleiotrophin and midkine, was screened in 289 neuroblastomas and 56 paired normal tissues. ALK was expressed in 99% of tumors and phosphorylated in 48% of cases. Pleiotrophin and midkine were expressed in 58% and 79% of tumors, respectively. ALK activation was significantly higher in tumors than in paired normal tissues, together with ALK and midkine expression. ALK activation was largely independent of mutations and correlated with midkine expression in tumors. ALK activation in tumors was associated with favorable features, including a younger age at diagnosis, hyperdiploidy, and detection by mass screening. Antitumor activity of the ALK inhibitor TAE684 was evaluated in wild-type or mutated ALK neuroblastoma cell lines and xenografts. TAE684 was cytotoxic in vitro in all cell lines, especially those harboring an ALK mutation. TAE684 efficiently inhibited ALK phosphorylation in vivo in both F1174I and R1275Q xenografts but demonstrated antitumor activity only against the R1275Q xenograft. In conclusion, ALK activation occurs frequently during neuroblastoma oncogenesis, mainly through mutation-independent mechanisms. However, ALK activation is not associated with a poor outcome and is not always a driver of cell proliferation and/or survival in neuroblastoma.


Subject(s)
Cell Proliferation/genetics , Cell Transformation, Neoplastic/drug effects , Neuroblastoma/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Adolescent , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Mutation/genetics , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/drug effects
6.
Neoplasia ; 17(8): 613-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26408254

ABSTRACT

Pleiotrophin (PTN) is a pleiotropic growth factor that exhibits angiogenic properties and is involved in tumor growth and metastasis. Although it has been shown that PTN is expressed in tumor cells, few studies have investigated its receptors and their involvement in cell migration and invasion. Neuropilin-1 (NRP-1) is a receptor for multiple growth factors that mediates cell motility and plays an important role in angiogenesis and tumor progression. Here we provide evidence for the first time that NRP-1 is crucial for biological activities of PTN. We found that PTN interacted directly with NRP-1 through its thrombospondin type-I repeat domains. Importantly, binding of PTN to NRP-1 stimulated the internalization and recycling of NRP-1 at the cell surface. Invalidation of NRP-1 by RNA interference in human carcinoma cells inhibited PTN-induced intracellular signaling of the serine-threonine kinase, mitogen-activated protein MAP kinase, and focal adhesion kinase pathways. Accordingly, NRP-1 silencing or blocking by antibody inhibited PTN-induced human umbilical vein endothelial cell migration and tumor cell invasion. These results suggest that NRP-1/PTN interaction provides a novel mechanism for controlling the response of endothelial and tumoral cells to PTN and may explain, at least in part, how PTN contributes to tumor angiogenesis and cancer progression.


Subject(s)
Carrier Proteins/metabolism , Cell Movement/physiology , Cytokines/metabolism , Neuropilin-1/metabolism , Signal Transduction/physiology , Animals , Binding Sites/genetics , CHO Cells , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Cricetinae , Cricetulus , Cytokines/genetics , Endocytosis/drug effects , Endocytosis/genetics , Endocytosis/physiology , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Immunoblotting , Microscopy, Fluorescence , Neoplasm Invasiveness , Neuropilin-1/genetics , Protein Binding/genetics , RNA Interference , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Biochimie ; 107 Pt B: 350-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25315978

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.


Subject(s)
Carrier Proteins/metabolism , Cytokines/metabolism , Fibroblast Growth Factor 2/metabolism , Amino Acid Sequence , Binding Sites , Biosensing Techniques , Carrier Proteins/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytokines/pharmacology , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Molecular Sequence Data , Neovascularization, Physiologic , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
8.
Cytokine ; 62(1): 44-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23481101

ABSTRACT

BACKGROUND: Pleiotrophin (PTN) is a heparin-binding growth factor involved in angiogenesis during development and tumor growth. Plasmid therapy with PTN also induces angiogenesis after myocardial infarction. During aging, angiogenesis is impaired and we therefore examined whether a growth factor therapy with PTN is able to restore neovascularization. METHODS: We evaluated the PTN effects on capillary-like endothelial sprouting in adult (n = 10) and senescent (n = 10) rats, using an ex vivo model of explanted aortic segments in culture. Freshly cut thoracic aortic rings from 3 and 24 month old (mo) rats (both n = 12) were cultured in a 3-dimensional collagen matrix with or without addition of recombinant human PTN (2.5-250 ng/ml) or Vascular Endothelial Growth Factor-165 (VEGF) (1-100 ng/ml) and the length of developed capillary network was quantified at day 3 and 6 by image analysis. RESULTS: After 6 days of culture, capillary-like tube formation was lower in control conditions in 24 mo aortic rings than in 3 mo rings. Addition of PTN increased dose-dependently the length of capillary-like tube formation in both 3 and 24 mo rings (P < 0.001 and P < 0.001 respectively). Age-associated impairment of capillary-like tube formation had been successfully restored in senescent aortic segments by PTN treatment. PTN induced development of capillary network similar to that observed with VEGF therapy with doses equal or superior to 10 ng/ml. CONCLUSION: PTN is able to induce ex vivo angiogenesis during aging and might be a new promising therapy to induce neovascularization in aged tissues as well as after age-associated cardiac, hindlimb or cerebral ischemia.


Subject(s)
Aging/physiology , Aorta/physiology , Carrier Proteins/pharmacology , Cytokines/pharmacology , Neovascularization, Physiologic/drug effects , Aging/drug effects , Animals , Aorta/drug effects , Humans , In Vitro Techniques , Male , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/pharmacology
9.
PLoS One ; 7(9): e44351, 2012.
Article in English | MEDLINE | ID: mdl-23028527

ABSTRACT

Recently, we have found that the skin secretions of the Amazonian tree frog Phyllomedusa bicolor contains molecules with antitumor and angiostatic activities and identified one of them as the antimicrobial peptide dermaseptin (Drs) B2. In the present study we further explored the in vitro and in vivo antitumor activity of this molecule and investigated its mechanism of action. We showed that Drs B2 inhibits the proliferation and colony formation of various human tumor cell types, and the proliferation and capillary formation of endothelial cells in vitro. Furthermore, Drs B2 inhibited tumor growth of the human prostate adenocarcinoma cell line PC3 in a xenograft model in vivo. Research on the mechanism of action of Drs B2 on tumor PC3 cells demonstrated a rapid increasing amount of cytosolic lactate dehydrogenase, no activation of caspase-3, and no changes in mitochondrial membrane potential. Confocal microscopy analysis revealed that Drs B2 can interact with the tumor cell surface, aggregate and penetrate the cells. These data together indicate that Drs B2 does not act by apoptosis but possibly by necrosis. In conclusion, Drs B2 could be considered as an interesting and promising pharmacological and therapeutic leader molecule for the treatment of cancer.


Subject(s)
Amphibian Proteins/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Antineoplastic Agents/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , L-Lactate Dehydrogenase/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Microscopy, Confocal , Xenograft Model Antitumor Assays
10.
Amino Acids ; 42(1): 385-95, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21132338

ABSTRACT

The discovery of new molecules with potential antitumor activity continues to be of great importance in cancer research. In this respect, natural antimicrobial peptides isolated from various animal species including humans and amphibians have been found to be of particular interest. Here, we report the presence of two anti-proliferative peptides active against cancer cells in the skin secretions of the South American tree frog, Phyllomedusa bicolor. The crude skin exudate was fractioned by size exclusion gel followed by reverse-phase HPLC chromatography. After these two purification steps, we identified two fractions that exhibited anti-proliferative activity. Sequence analysis indicated that this activity was due to two antimicrobial α-helical cationic peptides of the dermaseptin family (dermaseptins B2 and B3). This result was confirmed using synthetic dermaseptins. When tested in vitro, synthetic B2 and B3 dermaseptins inhibited the proliferation of the human prostatic adenocarcinoma PC-3 cell line by more than 90%, with an EC(50) of around 2-3 µM. No effect was observed on the growth of the NIH-3T3 non-tumor mouse cell line with Drs B2, whereas a slight inhibiting effect was observed with Drs B3 at high dose. In addition, the two fractions obtained after size exclusion chromatography also inhibited PC-3 cell colony formation in soft agar. Interestingly, inhibition of the proliferation and differentiation of activated adult bovine aortic endothelial cells was observed in cells treated with these two fractions. Dermaseptins B2 and B3 could, therefore, represent interesting new pharmacological molecules with antitumor and angiostatic properties for the development of a new class of anticancer drugs.


Subject(s)
Angiostatic Proteins/metabolism , Angiostatic Proteins/pharmacology , Antineoplastic Agents/pharmacology , Skin/chemistry , Skin/metabolism , Angiostatic Proteins/analysis , Angiostatic Proteins/isolation & purification , Animals , Antineoplastic Agents/analysis , Antineoplastic Agents/isolation & purification , Anura , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , NIH 3T3 Cells , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Am J Pathol ; 179(3): 1278-86, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21777561

ABSTRACT

Dry eye is a common disease that develops as a result of alteration of tear fluid, leading to osmotic stress and a perturbed epithelial barrier. Matrix metalloproteinase-9 (MMP-9) may be important in dry eye disease, as its genetic knockout conferred resistance to the epithelial disruption. We show that extracellular matrix metalloproteinase inducer (EMMPRIN; also termed CD147), an inducer of MMP expression, participates in the pathogenesis of dry eye through MMP-mediated cleavage of occludin, an important component of tight junctions. EMMPRIN expression was increased on the ocular surface of dry eye patients and correlated with those of MMP-9. High osmolarity in cell culture, mimicking dry eye conditions, increased both EMMPRIN and MMP-9 and resulted in the disruption of epithelial junctions through the cleavage of occludin. Exogenously added recombinant EMMPRIN had similar effects that were abrogated in the presence of the MMP inhibitor marimastat. Membrane occludin immunostaining was markedly increased in the apical corneal epithelium of both EMMPRIN and MMP-9 knock-out mice. Furthermore, an inverse correlation between EMMPRIN and occludin membrane staining was consistently observed both in vitro and in vivo as a function of corneal epithelial cells differentiation. These data suggest a possible role of EMMPRIN in regulating the amount of occludin at the cell surface in homeostasis beyond pathological situations such as dry eye disease, and EMMPRIN may be essential for the formation and maintenance of organized epithelial structure.


Subject(s)
Basigin/pharmacology , Dry Eye Syndromes/etiology , Matrix Metalloproteinase Inhibitors , Membrane Proteins/drug effects , Animals , Basigin/metabolism , Cell Differentiation , Dry Eye Syndromes/metabolism , Epithelium, Corneal/drug effects , Homeostasis , Humans , Mice , Mice, Knockout , Occludin , Osmolar Concentration , Recombinant Proteins/pharmacology
12.
BMC Cancer ; 11: 212, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21624116

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 RPTPß/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, RPTPß/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.


Subject(s)
Adenocarcinoma/metabolism , Carrier Proteins/pharmacology , Cytokines/pharmacology , Peptides/pharmacology , Prostatic Neoplasms/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Carrier Proteins/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/chemistry , Female , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Peptides/chemical synthesis , Prostatic Neoplasms/pathology , Protein Binding/drug effects , Xenograft Model Antitumor Assays
13.
Int J Oncol ; 38(1): 179-88, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21109939

ABSTRACT

We previously demonstrated, using the glioblastoma cell line U87MG as an experimental model, that the adenoviral mediated overexpression of the truncated protein HARPΔ111-136 inhibits the proliferation of these cells in vitro as well as tumor growth and angiogenesis in vivo. This study focused on identifying the underlying mechanisms for the observed antitumoral effect. The present study demonstrated that HARPΔ111-136 induced the ATF4/ATF3/CHOP cascade resulting in a strong expression of the proapoptotic protein CHOP, leading to tumor cell apoptosis as demonstrated by PARP cleavage and FACS analysis. siRNA-mediated CHOP gene silencing abolished Ad-HARPΔ111-136 induced apoptosis. Moreover, Ad-HARPΔ111-136 increased the expression of the death receptor DR5 and enhanced U87MG cells sensitivity in vitro to TRAIL a DR5 ligand with subsequent activation of caspase 8. Infection of U87MG cells with Ad-HARPΔ111-136 also enhanced radiation-induced apoptosis. In vivo, the combination of Ad-HARPΔ111-136 and radiation therapy resulted in a striking inhibition (92%) of the growth of U87MG xenografts, resulting from the potent effect on tumor angiogenesis and tumor cell apoptosis as determined by TUNEL analysis. Taken together, our results indicated that the inhibitor HARPΔ111-136 sensitized U87MG cells to apoptosis.


Subject(s)
Apoptosis/radiation effects , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , DNA Helicases/biosynthesis , Glioblastoma/pathology , Glioblastoma/radiotherapy , Peptide Fragments/biosynthesis , Animals , Apoptosis/physiology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , DNA Helicases/genetics , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunohistochemistry , Mice , Mice, Nude , Peptide Fragments/genetics , Transcription Factor CHOP , Transfection , Xenograft Model Antitumor Assays
14.
J Cell Physiol ; 226(1): 141-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20648565

ABSTRACT

Matrix metalloproteinases (MMPs) are thought to play an important role in skeletal muscle cell growth and differentiation. In view of the MMP inducing function of EMMPRIN/CD147, its role in myogenic cell differentiation was investigated. EMMPRIN level increased during differentiation of both rat primary myoblasts derived from satellite cells and mouse C2.7 myogenic cells and was associated with an alteration in its molecular forms. In parallel, expression of pro-MMP-9 gradually decreased and that of pro-MMP-2 and active MMP-2 increased. While small interfering RNA (siRNA) inhibition of EMMPRIN expression accelerated cell differentiation, exogenously added recombinant EMMPRIN inhibited differentiation by an MMP-mediated mechanism, as the MMP inhibitor marimastat abrogated EMMPRIN's effect. Our results further suggest that EMMPRIN regulates differentiation through an MMP activation of transforming growth factor beta (TGFß), a known inhibitor of myoblast's differentiation, as the increased activation and signaling of TGFß by EMMPRIN was attenuated in the presence of marimastat. EMMPRIN inhibition may thus represent a novel strategy in the treatment of muscular degenerative disorders.


Subject(s)
Basigin/metabolism , Cell Differentiation/physiology , Gene Silencing , Matrix Metalloproteinases/metabolism , Satellite Cells, Skeletal Muscle/cytology , Animals , Basigin/genetics , Cell Line , Cells, Cultured , Enzyme Induction , Extracellular Matrix/enzymology , Extracellular Matrix/physiology , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Mice , Muscle Development/physiology , Myoblasts/cytology , Myoblasts/physiology , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Satellite Cells, Skeletal Muscle/physiology , Transforming Growth Factor beta/metabolism
15.
Mol Cancer ; 9: 224, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20738847

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 RPTPß/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 RPTPß/ζ, but also interferes with other pleiotrophin receptors, as demonstrated by selective knockdown of pleiotrophin or RPTPß/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 RPTPß/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.


Subject(s)
Carrier Proteins/chemistry , Cytokines/chemistry , Peptide Fragments/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans
16.
Int J Cancer ; 127(5): 1038-51, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20013808

ABSTRACT

Glioblastoma is the most common primary brain tumor in human adults. Since existing treatments are not effective enough, novel therapeutic targets must be sought. The heparin-binding growth factor, heparin affin regulatory peptide (HARP), also known as pleiotrophin (PTN), could potentially represent such a target. We have previously shown that a mutant protein, HARPDelta111-136, which lacks HARP's C-terminal 26 amino acids, acts as a dominant negative HARP effector by heterodimerizing with the wild-type growth factor. The aim of our study was to evaluate the potential inhibitory activity of HARPDelta111-136 on the U87 MG human glioblastoma cell line. By overexpressing the truncated form of HARP in stably established clones of U87 MG cells, we observed an inhibition of proliferation under both anchorage-dependent and anchorage-independent conditions. We confirmed these results in an in vivo subcutaneous tumor xenograft model. In addition, we found that HARPDelta111-136 inhibited cell proliferation in a paracrine manner. Analysis of key cellular pathways revealed a decrease of cell adhesion in U87 MG cells that overexpressed the mutant protein, which could explain this inhibitory effect. A replication-defective adenovirus model that encoded HARPDelta111-136 supported a putative antiproliferative role for the truncated protein in vitro and in vivo. Interestingly, HARPDelta111-136 was also able to abolish angiogenic activity in HUVEC proliferation and in a Matrigel plug assay. These results demonstrate that considering its antiproliferative and angiostatic effects, HARPDelta111-136 could be of great interest when used in conjunction with standard treatments.


Subject(s)
Brain Neoplasms/pathology , Carrier Proteins/genetics , Cytokines/genetics , Glioblastoma/pathology , Mutation/genetics , Proto-Oncogene Proteins/genetics , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , CHO Cells , Cell Adhesion , Cell Movement , Cell Proliferation , Collagen/metabolism , Cricetinae , Cricetulus , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunoenzyme Techniques , Laminin/metabolism , Mice , Mice, Nude , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Proteoglycans/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Exp Hematol ; 37(9): 1072-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19539688

ABSTRACT

OBJECTIVE: Glycosaminoglycans (GAG) are major components of bone marrow extracellular matrix because they have the property to interact with cells and growth factors in hematopoietic niches. In this study, we investigated the effect of two different chemically defined GAG mimetics on mobilization of hematopoietic stem and progenitor cells (HSPCs) in mice peripheral blood. MATERIALS AND METHODS: Mobilization was achieved by intraperitoneal injection of GAG mimetics. Mobilized cells were characterized phenotypically by reverse transcription polymerase chain reaction and fluorescence-activated cell sorting analysis and functionally by colony-forming cell, cobblestone area-forming cell and long-term culture-initiating cell assays in vitro. Radioprotection assays were performed to confirm the functionality of primitive hematopoietic cells in vivo. Involvement of stromal-derived factor-1 (SDF-1) and matrix metalloproteinase-9 (MMP-9) were investigated. RESULTS: GAG mimetics treatment induces hyperleukocytosis and mobilization of HSPC. They synergize with the effects of granulocyte colony-stimulating factor or AMD3100 on hematopoietic progenitors mobilization. Reconstitution of lethally irradiated recipient mice with peripheral blood mononuclear cells from GAG mimetic-treated donor mice improves engraftment and survival. BiAcore studies indicate that the mimetics interact directly with SDF-1. In addition, GAG mimetics-induced mobilization is associated with increased levels of pro- and active MMP-9 from bone marrow cells and increased level of SDF-1 in peripheral blood. Finally, mobilization is partially inhibited by co-injection with anti-SDF-1 antibody. CONCLUSION: This study demonstrates that GAG mimetics induce efficient mobilization of HSPCs, associated with an activation of pro-MMP-9 and a modification in the SDF-1 concentration gradient between bone marrow and peripheral blood. We suggest that structural features of GAGs can modify the nature of mobilized cells.


Subject(s)
Biomimetic Materials/pharmacology , Chemokine CXCL12/blood , Glycosaminoglycans/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Matrix Metalloproteinase 9/blood , Animals , Anti-HIV Agents/agonists , Anti-HIV Agents/pharmacology , Benzylamines , Bone Marrow/metabolism , Cyclams , Drug Synergism , Glycosaminoglycans/agonists , Graft Survival/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/agonists , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/agonists , Heterocyclic Compounds/pharmacology , Male , Mice , Structure-Activity Relationship , Transplantation, Homologous
18.
Mol Cancer Ther ; 7(9): 2817-27, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790762

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.


Subject(s)
Breast Neoplasms/pathology , Carrier Proteins/pharmacology , Cytokines/pharmacology , Endothelial Cells/drug effects , Peptide Fragments/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/blood supply , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Endothelial Cells/pathology , Gene Expression Regulation, Neoplastic/drug effects , Mice , Mice, Nude , Molecular Weight , Mutant Proteins/metabolism , Neovascularization, Pathologic/pathology , Receptors, Cell Surface/metabolism , Xenograft Model Antitumor Assays
19.
J Cell Physiol ; 214(1): 250-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17607711

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.


Subject(s)
Carrier Proteins/chemistry , Cytokines/chemistry , Mitosis/drug effects , Neoplasms/drug therapy , Neovascularization, Physiologic/drug effects , Peptides/pharmacology , Thrombospondins/chemistry , Amino Acid Sequence , Animals , Breast Neoplasms/pathology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement , Cytokines/antagonists & inhibitors , Cytokines/genetics , Endothelium, Vascular/cytology , Female , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/genetics , Heparin/metabolism , Humans , Mice , Mice, Nude , Molecular Sequence Data , NIH 3T3 Cells , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Transplantation, Heterologous , Umbilical Veins/cytology
20.
Mol Cell Biol ; 27(24): 8454-65, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17908800

ABSTRACT

Matrix metalloproteinases (MMPs) exert both pro- and antiangiogenic functions by the release of cytokines or proteolytically generated angiogenic inhibitors from extracellular matrix and basement membrane remodeling. In the Mmp2-/- mouse neovascularization is greatly reduced, but the mechanistic aspects of this remain unclear. Using isotope-coded affinity tag labeling of proteins analyzed by multidimensional liquid chromatography and tandem mass spectrometry we explored proteome differences between Mmp2-/- cells and those rescued by MMP-2 transfection. Proteome signatures that are hallmarks of proteolysis revealed cleavage of many known MMP-2 substrates in the cellular context. Proteomic evidence of MMP-2 processing of novel substrates was found. Insulin-like growth factor binding protein 6, follistatin-like 1, and cystatin C protein cleavage by MMP-2 was biochemically confirmed, and the cleavage sites in heparin affin regulatory peptide (HARP; pleiotrophin) and connective tissue growth factor (CTGF) were sequenced by matrix-assisted laser desorption ionization-time of flight mass spectrometry. MMP-2 processing of HARP and CTGF released vascular endothelial growth factor (VEGF) from angiogenic inhibitory complexes. The cleaved HARP N-terminal domain increased HARP-induced cell proliferation, whereas the HARP C-terminal domain was antagonistic and decreased cell proliferation and migration. Hence the unmasking of cytokines, such as VEGF, by metalloproteinase processing of their binding proteins is a new mechanism in the control of cytokine activation and angiogenesis.


Subject(s)
Angiogenesis Inhibitors/metabolism , Carrier Proteins/metabolism , Cytokines/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Protein Processing, Post-Translational , Proteomics , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Movement , Cell Proliferation , Connective Tissue Growth Factor , Culture Media, Conditioned , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , Humans , Isotope Labeling , Mice , NIH 3T3 Cells , Peptides/metabolism , Protein Biosynthesis , Reproducibility of Results , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...