Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Neurooncol ; 122(1): 63-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25563813

ABSTRACT

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are neuropeptides acting through VPAC1, VPAC2 and PAC1 receptors (referred here as the VIP-receptor system). In the central nervous system, VIP and PACAP are involved in neurogenesis, cell differentiation and migration, suggesting that they could be implicated in the development of glioblastoma (GBM). The infiltrative nature of GBM remains a major problem for the therapy of these tumors. We previously demonstrated that the VIP-receptor system regulated cell migration of the human cell lines M059J and M059K, derived from a single human GBM. Here, we evaluated the involvement of the VIP-receptor system in GBM cell invasion. In Matrigel invasion assays, M059K cells that express more the VIP-receptor system than M059J cells were less invasive. Invasion assays performed in the presence of agonists, antagonists or anti-PACAP antibodies as well as experiments with transfected M059J cells overexpressing the VPAC1 receptor indicated that the more the VIP-receptor system was expressed and activated, the less the cells were able to invade. Western immunoblotting experiments revealed that the VIP-receptor system inactivated the signaling protein AKT. Invasion assays carried out in the presence of an AKT inhibitor demonstrated the involvement of this signaling kinase in the regulation of cell invasion by the VIP-receptor system in M059K cells. The inhibition by VIP of invasion and AKT was also observed in U87 cells. In conclusion, VIP and PACAP act as anti-invasive factors in different GBM cell lines, a function mediated by VPAC1 inhibition of AKT signaling in M059K cells.


Subject(s)
Cell Movement/drug effects , Glioblastoma/drug therapy , Neuroprotective Agents/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
2.
J Neurochem ; 120(1): 93-105, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22007767

ABSTRACT

Increasing evidences suggest that neuropeptide Y (NPY) may act as a key modulator of the cross-talk between the brain and the immune system in health and disease. In the present study, we dissected the possible inhibitory role of NPY upon inflammation-associated microglial cell motility. NPY, through activation of Y(1) receptors, was found to inhibit lipopolysaccharide (LPS)-induced microglia (N9 cell line) motility. Moreover, stimulation of microglia with LPS was inhibited by IL-1 receptor antagonist (IL-1ra), suggesting the involvement of endogenous interleukin-1 beta (IL-1ß) in this process. Direct stimulation with IL-1ß promoted downstream p38 mitogen-activated protein kinase mobilization and increased microglia motility. Moreover, consistently, p38 mitogen-activated protein kinase inhibition decreased the extent of actin filament reorganization occurring during plasma membrane ruffling and p38 phosphorylation was inhibited by NPY, involving Y(1) receptors. Significantly, the key inhibitory role of NPY on LPS-induced motility of CD11b-positive cells was further confirmed in mouse brain cortex explants. In summary, we revealed a novel functional role for NPY in the regulation of microglial function that may have important implications in the modulation of CNS injuries/diseases where microglia migration/motility might play a role.


Subject(s)
Cell Movement/drug effects , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/pharmacology , Microglia/drug effects , Neuropeptide Y/pharmacology , Animals , Blotting, Western , CD11b Antigen/metabolism , Cell Line , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Data Interpretation, Statistical , Immunohistochemistry , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Receptors, Neuropeptide Y/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
4.
Int J Oncol ; 33(5): 1081-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18949372

ABSTRACT

Neuroblastoma is a pediatric tumor which can spontaneously regress or differentiate into a benign tumor. MYCN oncogene amplification occurs in 22% of neuroblastomas and is associated with poor prognosis. Retinoic acid (RA), a molecule able to induce differentiation and to decrease MYCN expression, is used in the therapy of neuroblastomas. The neuropeptide vasoactive intestinal peptide (VIP) is known to control proliferation or differentiation of numerous cancer cells. In vitro, VIP induces differentiation of neuroblastoma cells. To determine whether VIP could modulate MYCN expression, we carried out real-time quantitative RT-PCR and Western immunoblot analyses in human neuroblastoma SH-SY5Y and IMR-32 cells. The results indicated that VIP reduced MYCN mRNA and protein expression, especially in the MYCN-amplified IMR-32 cells, with a maximal and transient decrease by approximately 50% after few hours of treatment with VIP at 10(-6) M. This effect was compared to that of RA at 10(-5) M, which induced a diminution of MYCN mRNA expression by approximately 25% after few days of treatment. This indicated that VIP and RA display complementary kinetics. Cotreatments showed that VIP and RA had synergistic effects on regulation of expression of MYCN proteins. VIP and RA cotreatments regulated also expression of two MYCN target genes, SKP2 and TP53INP1. These results suggest that VIP, in combination with RA may have a potential therapeutic benefit in neuroblastomas with MYCN amplification, a genetic abnormality associated with poor prognosis.


Subject(s)
Antineoplastic Agents/pharmacology , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Tretinoin/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Shape/drug effects , Down-Regulation , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/metabolism , Humans , Kinetics , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , RNA, Messenger/metabolism , S-Phase Kinase-Associated Proteins/metabolism
5.
Oncotarget ; 8(69): 113360-113372, 2017 Dec 26.
Article in English | MEDLINE | ID: mdl-29371916

ABSTRACT

Interleukin 17B (IL-17B) is a pro-inflammatory cytokine that belongs to the IL-17 cytokines family and binds to IL-17 receptor B (IL-17RB). Here we found that high expression of IL-17B and IL-17RB is associated with poor prognosis in patients with breast cancer and that IL-17B expression upregulation is specifically associated with poorer survival in patients with basal-like breast cancer. We thus focused on IL-17B role in breast cancer by using luminal and triple negative (TN)/basal-like tumor cell lines. We found that IL-17B induces resistance to conventional chemotherapeutic agents. In vivo, IL-17B induced resistance to paclitaxel and treatment with an anti-IL-17RB neutralizing antibody completely restored breast tumor chemosensitivity, leading to tumor shrinkage. We next focused on the signaling pathways activated in human breast cancer cell lines upon incubation with IL-17B. We observed that IL-17B induces ERK1/2 pathway activation, leading to upregulation of anti-apoptotic proteins of the BCL-2 family. IL-17B-induced chemoresistance was completely abolished by incubation with PD98059, an inhibitor of the MAPK/ERK pathway, indicating that the ERK pathway plays a crucial role. Altogether our results emphasize the role of the IL-17B/IL-17RB signaling pathway in breast tumors and identify IL-17B and its receptor as attractive therapeutic targets for potentiating breast cancer chemotherapy.

6.
Sci Rep ; 5: 11874, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26154409

ABSTRACT

Pro-inflammatory IL-17 cytokines were initially described for their pathogenic role in chronic inflammatory diseases and subsequent accumulating evidence indicated their involvement in carcinogenesis. In the present study we report that IL-17A and IL-17E receptors subunits mRNA expressions are upregulated in breast cancers versus normal samples. IL-17E, which is undetectable in most normal breast tissues tested, seems more expressed in some tumors. Investigation of the molecular signaling following stimulation of human breast cancer cell lines with IL-17A and IL-17E showed that both cytokines induced the phosphorylation of c-RAF, ERK1/2 and p70 S6 Kinase were involved in the proliferation and survival of tumor cells. Accordingly, IL-17A and IL-17E promoted resistance to Docetaxel and failed to induce apoptosis as previously reported for IL-17E. Interestingly, we also revealed that both cytokines induced the generation of tumorogenic low molecular weight forms of cyclin E (LMW-E), which high levels correlated strongly with a poor survival in breast cancer patients. These results show for the first time some of the molecular pathways activated by IL-17A and IL-17E that may participate to their pro-oncogenic activity in breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Cyclin E/metabolism , Interleukin-17/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Biopsy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Humans , Interleukin-17/genetics , Interleukin-17/pharmacology , Phosphorylation , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism
7.
Cancer Immunol Res ; 3(3): 254-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25403716

ABSTRACT

The ectonucleotidases CD39 and CD73 hydrolyze extracellular adenosine triphosphate (ATP) and adenosine diphosphate (ADP) to generate adenosine, which binds to adenosine receptors and inhibits T-cell and natural killer (NK)-cell responses, thereby suppressing the immune system. The generation of adenosine via the CD39/CD73 pathway is recognized as a major mechanism of regulatory T cell (Treg) immunosuppressive function. The number of CD39⁺ Tregs is increased in some human cancers, and the importance of CD39⁺ Tregs in promoting tumor growth and metastasis has been demonstrated using several in vivo models. Here, we addressed whether CD39 is expressed by tumor cells and whether CD39⁺ tumor cells mediate immunosuppression via the adenosine pathway. Immunohistochemical staining of normal and tumor tissues revealed that CD39 expression is significantly higher in several types of human cancer than in normal tissues. In cancer specimens, CD39 is expressed by infiltrating lymphocytes, the tumor stroma, and tumor cells. Furthermore, the expression of CD39 at the cell surface of tumor cells was directly demonstrated via flow cytometry of human cancer cell lines. CD39 in cancer cells displays ATPase activity and, together with CD73, generates adenosine. CD39⁺CD73⁺ cancer cells inhibited the proliferation of CD4 and CD8 T cells and the generation of cytotoxic effector CD8 T cells (CTL) in a CD39- and adenosine-dependent manner. Treatment with a CD39 inhibitor or blocking antibody alleviated the tumor-induced inhibition of CD4 and CD8 T-cell proliferation and increased CTL- and NK cell-mediated cytotoxicity. In conclusion, interfering with the CD39-adenosine pathway may represent a novel immunotherapeutic strategy for inhibiting tumor cell-mediated immunosuppression.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine/immunology , Apyrase/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/immunology , Adenosine Triphosphatases/immunology , Antigens, CD/immunology , Apyrase/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Receptors, Purinergic P1/immunology
8.
Sci Rep ; 3: 3456, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-24316750

ABSTRACT

The proinflammatory cytokine Interleukin 17A (hereafter named IL-17A) or IL-17A producing cells are elevated in breast tumors environment and correlate with poor prognosis. Increased IL-17A is associated with ER(-) or triple negative tumors and reduced Disease Free Survival. However, the pathophysiological role of IL-17A in breast cancer remains unclear although several studies suggested its involvement in cancer cell dissemination. Here we demonstrated that a subset of breast tumors is infiltrated with IL-17A-producing cells. Increased IL-17A seems mainly associated to ER(-) and triple negative/basal-like tumors. Isolation of tumor infiltrating T lymphocytes (TILs) from breast cancer biopsies revealed that these cells secreted significant amounts of IL-17A. We further established that recombinant IL-17A recruits the MAPK pathway by upregulating phosphorylated ERK1/2 in human breast cancer cell lines thereby promoting proliferation and resistance to conventional chemotherapeutic agents such as docetaxel. We also confirmed here that recombinant IL-17A stimulates migration and invasion of breast cancer cells as previously reported. Importantly, TILs also induced tumor cell proliferation, chemoresistance and migration and treatment with IL-17A-neutralizing antibodies abrogated these effects. Altogether these results demonstrated the pathophysiological role of IL-17A-producing cell infiltrate in a subset of breast cancers. Therefore, IL-17A appears as potential therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Interleukin-17/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , MAP Kinase Signaling System , Antineoplastic Agents/pharmacology , Biopsy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Docetaxel , Female , Humans , Interleukin-17/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Receptors, Estrogen/metabolism , Taxoids/pharmacology
9.
Neuropeptides ; 44(5): 373-83, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20638719

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain tumor in adults. This cancer has an infiltrative nature and the median survival of patients is about one year. Vasoactive intestinal peptide (VIP) belongs to a structurally related family of polypeptides and is a major regulatory factor in the central and peripheral nervous systems. VIP regulates proliferation of astrocytes and of numerous cancer cell lines and modulates migration in prostatic and colonic cancer cell lines. Little is known about the involvement of VIP and its receptors (VIP-receptor system) in proliferation or migration of GBM cells. The effects of VIP, PACAP and of synthetic VIP antagonists were tested in two human GBM cell lines, M059K and M059J, established from two different parts of a single tumor. In these cells, the data revealed that the VIP-receptor system did not affect proliferation but controlled cell migration. Indeed, in M059K cells which express components of the VIP receptor system, the VIP receptor antagonists and a PACAP antibody enhanced migration. The VIP receptor antagonists increased generation of typical migration-associated processes: filopodia and lamellipodia, and activation of Rac1 and Cdc42 GTPases. Reciprocally, in M059J cells which poorly express the VIP-receptor system, treatments with the agonists VIP and PACAP resulted in decreased cell migration. Furthermore, the peptides appeared to act through a subclass of binding sites displaying an uncommon very high affinity for these ligands. Taken together, these observations suggest that components of the VIP-receptor system negatively regulate cell migration, thus showing potential anti-oncogenic properties.


Subject(s)
Cell Movement/physiology , Receptors, Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Humans , Immunohistochemistry , Polymerase Chain Reaction , Receptors, Vasoactive Intestinal Peptide/agonists , Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL