Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Natl Cancer Inst ; 111(6): 597-608, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30779852

ABSTRACT

BACKGROUND: Cancer cells from different origins exhibit various basal redox statuses and thus respond differently to intrinsic or extrinsic oxidative stress. These intricate characteristics condition the success of redox-based anticancer therapies that capitalize on the ability of reactive oxygen species to achieve selective and efficient cancer cell killing. METHODS: Redox biology methods, stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics, and bioinformatics pattern comparisons were used to decipher the underlying mechanisms for differential response of lung and breast cancer cell models to redox-modulating molecule auranofin (AUF) and to combinations of AUF and vitamin C (VC). The in vivo effect of AUF, VC, and two AUF/VC combinations on mice bearing MDA-MB-231 xenografts (n = 5 mice per group) was also evaluated. All statistical tests were two-sided. RESULTS: AUF targeted simultaneously the thioredoxin and glutathione antioxidant systems. AUF/VC combinations exerted a synergistic and hydrogen peroxide (H2O2)-mediated cytotoxicity toward MDA-MB-231 cells and other breast cancer cell lines. The anticancer potential of AUF/VC combinations was validated in vivo on MDA-MB-231 xenografts in mice without notable side effects. On day 14 of treatments, mean (SD) tumor volumes for the vehicle-treated control group and the two AUF/VC combination-treated groups (A/V1 and A/V2) were 197.67 (24.28) mm3, 15.66 (10.90) mm3, and 10.23 (7.30)mm3, respectively; adjusted P values of the differences between mean tumor volumes of vehicle vs A/V1 groups and vehicle vs A/V2 groups were both less than .001. SILAC proteomics, bioinformatics analysis, and functional experiments linked prostaglandin reductase 1 (PTGR1) expression levels with breast cancer cell sensitivity to AUF/VC combinations. CONCLUSION: The combination of AUF and VC, two commonly available drugs, could be efficient against triple-negative breast cancer and potentially other cancers with similar redox properties and PTGR1 expression levels. The redox-based anticancer activity of this combination and the discriminatory potential of PTGR1 expression are worth further assessment in preclinical and clinical studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Triple Negative Breast Neoplasms/drug therapy , A549 Cells , Animals , Ascorbic Acid/administration & dosage , Auranofin/administration & dosage , Cell Line, Tumor , Female , Glutathione/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Oxidative Stress/drug effects , Proteome/metabolism , Random Allocation , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
2.
J Natl Cancer Inst ; 2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30462268

ABSTRACT

BACKGROUND: Cancer cells from different origins exhibit various basal redox statuses and thus respond differently to intrinsic or extrinsic oxidative stress. These intricate characteristics condition the success of redox-based anticancer therapies that capitalize on the ability of reactive oxygen species to achieve selective and efficient cancer cell killing. METHODS: Redox biology methods, stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics, and bioinformatics pattern comparisons were used to decipher the underlying mechanisms for differential response of lung and breast cancer cell models to redox-modulating molecule auranofin (AUF) and to combinations of AUF and vitamin C (VC). The in vivo effect of AUF, VC, and two AUF/VC combinations on mice bearing MDA-MB-231 xenografts (n = 5 mice per group) was also evaluated. All statistical tests were two-sided. RESULTS: AUF targeted simultaneously the thioredoxin and glutathione antioxidant systems. AUF/VC combinations exerted a synergistic and hydrogen peroxide (H2O2)-mediated cytotoxicity toward MDA-MB-231 cells and other breast cancer cell lines. The anticancer potential of AUF/VC combinations was validated in vivo on MDA-MB-231 xenografts in mice without notable side effects. On day 14 of treatments, mean (SD) tumor volumes for the vehicle-treated control group and the two AUF/VC combination-treated groups (A/V1 and A/V2) were 197.67 (24.28) mm3, 15.66 (10.90) mm3, and 10.23 (7.30)mm3, respectively; adjusted P values of the differences between mean tumor volumes of vehicle vs A/V1 groups and vehicle vs A/V2 groups were both less than .001. SILAC proteomics, bioinformatics analysis, and functional experiments linked prostaglandin reductase 1 (PTGR1) expression levels with breast cancer cell sensitivity to AUF/VC combinations. CONCLUSION: The combination of AUF and VC, two commonly available drugs, could be efficient against triple-negative breast cancer and potentially other cancers with similar redox properties and PTGR1 expression levels. The redox-based anticancer activity of this combination and the discriminatory potential of PTGR1 expression are worth further assessment in preclinical and clinical studies.

3.
Oncotarget ; 7(50): 82511-82527, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27764776

ABSTRACT

Melanoma is a particularly virulent human cancer, due to its resistance to conventional treatments and high frequency of metastasis. Melanomas contain a fraction of cells, the melanoma-initiating cells (MICs), responsible for tumor propagation and relapse. Identification of the molecular pathways supporting MICs is, therefore, vital for the development of targeted treatments. One factor produced by melanoma cells and their microenvironment, insulin-like growth factor-1 (IGF- 1), is linked to epithelial-mesenchymal transition (EMT) and stemness features in several cancers.We evaluated the effect of IGF-1 on the phenotype and chemoresistance of B16-F10 cells. IGF-1 inhibition in these cells prevented malignant cell proliferation, migration and invasion, and lung colony formation in immunodeficient mice. IGF-1 downregulation also markedly inhibited EMT, with low levels of ZEB1 and mesenchymal markers (N-cadherin, CD44, CD29, CD105) associated with high levels of E-cadherin and MITF, the major regulator of melanocyte differentiation. IGF-1 inhibition greatly reduced stemness features, including the expression of key stem markers (SOX2, Oct-3/4, CD24 and CD133), and the functional characteristics of MICs (melanosphere formation, aldehyde dehydrogenase activity, side population). These features were associated with a high degree of sensitivity to mitoxantrone treatment.In this study, we deciphered new connections between IGF-1 and stemness features and identified IGF-1 as instrumental for maintaining the MIC phenotype. The IGF1/IGF1-R nexus could be targeted for the development of more efficient anti-melanoma treatments. Blocking the IGF-1 pathway would improve the immune response, decrease the metastatic potential of tumor cells and sensitize melanoma cells to conventional treatments.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition , Insulin-Like Growth Factor I/metabolism , Melanoma, Experimental/metabolism , Neoplastic Stem Cells/metabolism , Skin Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Female , Insulin-Like Growth Factor I/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/secondary , Mice, Inbred C57BL , Mitoxantrone/pharmacology , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Time Factors , Transfection , Tumor Microenvironment
4.
FEBS Lett ; 590(20): 3660-3668, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27616439

ABSTRACT

Cell-cell contacts coordinate the endothelial barrier function in response to external cues. To identify new mediators involved in cytokine-promoted endothelial permeability, we screened a siRNA library targeting E3 ubiquitin ligases. Here, we report that silencing of the late endosome/lysosomal membrane-associated RING-CH-3 (MARCH3) enzyme protects the endothelial barrier. Furthermore, transcriptome analysis unmasked the upregulation of the tight junction-encoding gene occludin (OCLN) in MARCH3-depleted cells. Indeed, MARCH3 silencing results in the strengthening of cell-cell contacts, as evidenced by the accumulation of junctional proteins. From a molecular standpoint, the FoxO1 forkhead transcription repressor was inactivated in the absence of MARCH3. This provides a possible molecular link between MARCH3 and the signaling pathway involved in regulating the expression of junctional proteins and barrier integrity.


Subject(s)
Blood-Brain Barrier/metabolism , Carrier Proteins/metabolism , Endothelium/metabolism , Forkhead Box Protein O1/metabolism , Membrane Proteins/metabolism , Occludin/genetics , Ubiquitin-Protein Ligases/metabolism , Carrier Proteins/genetics , Cell Line , Gene Expression Profiling/methods , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/metabolism , Membrane Proteins/genetics , Ubiquitin-Protein Ligases/genetics
5.
Oncotarget ; 7(13): 15507-24, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26551931

ABSTRACT

As rapidly developing patient-derived xenografts (PDX) could represent potential sources of cancer stem cells (CSC), we selected and characterized non-cultured PDX cell suspensions from four different renal carcinomas (RCC). Only the cell suspensions from the serial xenografts (PDX-1 and PDX-2) of an undifferentiated RCC (RCC-41) adapted to the selective CSC medium. The cell suspension derived from the original tumor specimen (RCC-41-P-0) did not adapt to the selective medium and strongly expressed CSC-like markers (CD133 and CD105) together with the non-CSC tumor marker E-cadherin. In comparison, PDX-1 and PDX-2 cells exhibited evolution in their phenotype since PDX-1 cells were CD133high/CD105-/Ecadlow and PDX-2 cells were CD133low/CD105-/Ecad-. Both PDX subsets expressed additional stem cell markers (CD146/CD29/OCT4/NANOG/Nestin) but still contained non-CSC tumor cells. Therefore, using different cell sorting strategies, we characterized 3 different putative CSC subsets (RCC-41-PDX-1/CD132+, RCC-41-PDX-2/CD133-/EpCAMlow and RCC-41-PDX-2/CD133+/EpCAMbright). In addition, transcriptomic analysis showed that RCC-41-PDX-2/CD133- over-expressed the pluripotency gene ERBB4, while RCC-41-PDX-2/CD133+ over-expressed several tumor suppressor genes. These three CSC subsets displayed ALDH activity, formed serial spheroids and developed serial tumors in SCID mice, although RCC-41-PDX-1/CD132+ and RCC-41-PDX-2/CD133+ displayed less efficiently the above CSC properties. RCC-41-PDX-1/CD132+ tumors showed vessels of human origin with CSC displaying peri-vascular distribution. By contrast, RCC-41-PDX-2 originated tumors exhibiting only vessels of mouse origin without CSC peri-vascular distribution.Altogether, our results indicate that PDX murine microenvironment promotes a continuous redesign of CSC phenotype, unmasking CSC subsets potentially present in a single RCC or generating ex novo different CSC-like subsets.


Subject(s)
Carcinoma, Renal Cell/pathology , Heterografts , Kidney Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Cell Culture Techniques/methods , Cell Separation/methods , Flow Cytometry/methods , Humans , Mice , Mice, SCID , Tumor Cells, Cultured
6.
Front Pharmacol ; 6: 281, 2015.
Article in English | MEDLINE | ID: mdl-26635611

ABSTRACT

Glioblastoma multiforme (GBM) constitutes the most common and the most aggressive type of human tumors affecting the central nervous system. Prognosis remains dark due to the inefficiency of current treatments and the rapid relapse. Paralleling other human tumors, GBM contains a fraction of tumor initiating cells with the capacity to self-renew, initiate and maintain the tumor mass. These cells were found in close proximity to brain vasculature, suggesting functional interactions between brain tumor-initiating cells (BTICs) and endothelial cells within the so-called vascular niche. However, the mechanisms by which these cells impact on the endothelium plasticity and function remain unclear. Using culture of BTICs isolated from a cohort of 14 GBM patients, we show that BTICs secretome promotes brain endothelial cell remodeling in a VEGF-independent manner. Gene array analysis unmasked that BTICs-released factors drove the expression of Ptch2 in endothelial cells. Interestingly, BTICs produce desert hedgehog (DHH) ligand, enabling a paracrine DHH/Ptch2 signaling cascade that conveys elevated permeability and angiogenesis. Finally, DHH silencing in BTICs dramatically reduced tumor growth, as well as vascularization and intra-tumor permeability. Collectively, our data unveil a role for DHH in exacerbated tumor angiogenesis and permeability, which may ultimately favor glioblastoma growth, and thus place the DHH/Ptch2 nexus as a molecular target for novel therapies.

7.
Neoplasia ; 17(6): 509-17, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26152359

ABSTRACT

Intrarenal interleukin-15 (IL-15) participates to renal pathophysiology, but the role of its different membrane-bound isoforms remains to be elucidated. In this study, we reassess the biology of membrane-bound IL-15 (mb-IL-15) isoforms by comparing primary cultures of human renal proximal tubular epithelial cells (RPTEC) to peritumoral (ptumTEC), tumoral (RCC), and cancer stem cells (CSC/CD105(+)). RPTEC express a 14 to 16 kDa mb-IL-15, whose existence has been assumed but never formally demonstrated and likely represents the isoform anchored at the cell membrane through the IL-15 receptor α (IL-15Rα) chain, because it is sensitive to acidic treatment and is not competent to deliver a reverse signal. By contrast, ptumTEC, RCC, and CSC express a novel N-hyperglycosylated, short-lived transmembrane mb-IL-15 (tmb-IL-15) isoform around 27 kDa, resistant to acidic shock, delivering a reverse signal in response to its soluble receptor (sIL-15Rα). This reverse signal triggers the down-regulation of the tumor suppressor gene E-cadherin in ptumTEC and RCC but not in CSC/CD105(+), where it promotes survival. Indeed, through the AKT pathway, tmb-IL-15 protects CSC/CD105(+) from non-programmed cell death induced by serum starvation. Finally, both mb-IL-15 and tmb-IL-15 are sensitive to metalloproteases, and the cleaved tmb-IL-15 (25 kDa) displays a powerful anti-apoptotic effect on human hematopoietic cells. Overall, our data indicate that both mb-IL-15 and tmb-IL-15 isoforms play a complex role in renal pathophysiology downregulating E-cadherin and favoring cell survival. Moreover, "apparently normal" ptumTEC cells, sharing different properties with RCC, could contribute to organize an enlarged peritumoral "preneoplastic" environment committed to favor tumor progression.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Membrane/metabolism , Interleukin-15/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/pathology , Neoplastic Stem Cells/pathology , Apoptosis , Blotting, Western , Carcinoma, Renal Cell/metabolism , Cell Proliferation , Cells, Cultured , Epithelial-Mesenchymal Transition , Flow Cytometry , Humans , Kidney Neoplasms/metabolism , Kidney Tubules, Proximal/metabolism , Neoplastic Stem Cells/metabolism , Protein Isoforms , Tumor Microenvironment
8.
Cell Commun Signal ; 13: 11, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25889342

ABSTRACT

BACKGROUND: The viral G protein-coupled receptor (vGPCR) is proposed to act as one of the predominant mediators of Kaposi's sarcoma (KS), a human herpes virus 8 (HHV8)-elicited disease. The actions of vGPCR manifest pathogenesis, in part, through increased permeability of endothelial cells. Endothelial cell-cell junctions have indeed emerged as an instrumental target involved in the vasculature defects observed within the tumor microenvironment. The pathway leading to adherens junction destabilization has been shown to involve the activation of the small GTPase Rac, in the context of either latent infection or the sole expression of vGPCR. However, the precise molecular mechanisms governed by vGPCR in vascular leakage require further elucidation. FINDINGS: Guanine exchange factors (GEFs) function as critical molecular switches that control the activation of small GTPases. We therefore screened the effects of 80 siRNAs targeting GEFs on vGPCR-driven endothelial permeability and identified switch-associated protein 70 (SWAP70) as necessary for its elevating effects. Pull-down experiments further showed that Rac activation by vGPCR was dependent on SWAP70. Examination of tissues and cells from HHV8-positive patients revealed that SWAP70 was ubiquitously expressed. Furthermore, SWAP70 was found to be crucial for vGPCR-driven endothelial tube formation and endothelial sprouting in vitro. CONCLUSIONS: SWAP70 appears to act as a molecular intermediate between vGPCR and endothelial activation. Because of the important role of vGPCR-mediated endothelial plasticity in KS pathogenesis, inhibition of SWAP70 function could be of interest for blocking vGPCR-driven activities in HHV8-defined diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Herpesvirus 8, Human/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Nuclear Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Viral Proteins/metabolism , DNA-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Herpesvirus 8, Human/genetics , Humans , Minor Histocompatibility Antigens , Nuclear Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Viral Proteins/genetics
9.
BMC Cell Biol ; 15: 38, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25339290

ABSTRACT

BACKGROUND: The interleukin-8 chemokine (IL-8) G-protein coupled receptor CXCR2 governs pro-inflammatory and pro-angiogenic responses in leukocytes and endothelial cells. At a molecular standpoint, CXCR2 is widely reported to operate through calcium flux, phosphoinoisitide 3 kinase (PI3K) and mitogen-activated protein kinase (MAPK). While CXCR2 trafficking is suspected to be intertwined with its signaling, the exact mechanism is not fully elucidated. RESULTS: Here, we identified the lysine 327 within the CXCR2 C-terminal tail as a key residue for ubiquitination, internalization, and signaling. First, the substitution to an arginine of K327 mutation was associated with a reduction in CXCR2 poly-ubiquitination. While WT CXCR2 was rapidly internalized following IL-8 administration, K327R mutant remained at the plasma membrane. Finally, K327R mutant failed to promote the recruitment of ß-arrestin2, as estimated by imagery and bioluminescence resonance transfer. As a consequence, the activation of intracellular signaling, including both early events such as ERK phosphorylation and the increase in calcium flux, and the latter activation of the AP1 and NF-κB transcription factors, was blunted. CONCLUSIONS: Overall, our results demonstrate that CXCR2 ubiquitination on K327 residue modulates agonist-activated CXCR2 cell sorting and intracellular signaling. Thus, the inhibition of K327 ubiquitination might emerge as an effective mean to curb exacerbated CXCR2 signaling in several pathological conditions, such as inflammatory diseases and cancer.


Subject(s)
Lysine/analysis , Receptors, Interleukin-8B/metabolism , Ubiquitination , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Interleukin-8/metabolism , Lysine/metabolism , Molecular Sequence Data , Protein Transport , Receptors, Interleukin-8B/chemistry , Sequence Alignment , Signal Transduction
10.
Med Sci (Paris) ; 30(4): 408-14, 2014 Apr.
Article in French | MEDLINE | ID: mdl-24801036

ABSTRACT

Tumor initiation and progression is a complex process in which cells acquire aberrant proliferation, survival and migration properties, and the ability to form a dedicated blood vessel network. Recent studies highlighted the existence of an active crosstalk between endothelial cells and the tumor mass. Indeed, cancer stem-like cells have been identified and found in the vicinity of blood vessels, and the latter has been proposed to act as a feeding bed for tumors, especially governing the fate of cancer stem-like cells. Here, we present an overview of the direct interplay between endothelial and cancer cells. We will first introduce the mechanisms involved in tumor angiogenesis. How the microenvironment impacts on endothelial plasticity will next be detailed, focusing on the tumor main secreted factors. Finally, the role of the vascular niche and its angiocrine factors in tumorigenesis will be addressed.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic , Humans , Neoplasms/pathology , Neovascularization, Pathologic/pathology
11.
Front Oncol ; 3: 211, 2013.
Article in English | MEDLINE | ID: mdl-23967403

ABSTRACT

The endothelial barrier strictly maintains vascular and tissue homeostasis, and therefore modulates many physiological processes such as angiogenesis, immune responses, and dynamic exchanges throughout organs. Consequently, alteration of this finely tuned function may have devastating consequences for the organism. This is particularly obvious in cancers, where a disorganized and leaky blood vessel network irrigates solid tumors. In this context, vascular permeability drives tumor-induced angiogenesis, blood flow disturbances, inflammatory cell infiltration, and tumor cell extravasation. This can directly restrain the efficacy of conventional therapies by limiting intravenous drug delivery. Indeed, for more effective anti-angiogenic therapies, it is now accepted that not only should excessive angiogenesis be alleviated, but also that the tumor vasculature needs to be normalized. Recovery of normal state vasculature requires diminishing hyperpermeability, increasing pericyte coverage, and restoring the basement membrane, to subsequently reduce hypoxia, and interstitial fluid pressure. In this review, we will introduce how vascular permeability accompanies tumor progression and, as a collateral damage, impacts on efficient drug delivery. The molecular mechanisms involved in tumor-driven vascular permeability will next be detailed, with a particular focus on the main factors produced by tumor cells, especially the emblematic vascular endothelial growth factor. Finally, new perspectives in cancer therapy will be presented, centered on the use of anti-permeability factors and normalization agents.

12.
Cell Commun Signal ; 11(1): 37, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23714586

ABSTRACT

BACKGROUND: The endothelial specific cell-cell adhesion molecule, VE-cadherin, modulates barrier function and vascular homeostasis. In this context, we have previously characterized that VEGF (vascular endothelial growth factor) leads to VE-cadherin phosphorylation, ß-arrestin2 recruitment and VE-cadherin internalization in mouse endothelial cells. However, exactly how this VE-cadherin/ß-arrestin complex contributes to VEGF-mediated permeability in human endothelial cells remains unclear. In this study, we investigated in-depth the VE-cadherin/ß-arrestin interactions in human endothelial cells exposed to VEGF. FINDINGS: First, we demonstrated that VEGF induces VE-cadherin internalization in a clathrin-dependent manner in human umbilical vein endothelial cells (HUVEC). In addition to the classical components of endocytic vesicles, ß-arrestin1 was recruited and bound to phosphorylated VE-cadherin. Molecular mapping of this interaction uncovered that the C-terminus tail of ß-arrestin1, that comprises amino acids 375 to 418, was sufficient to directly interact with the phosphorylated form of VE-cadherin. Interestingly, the expression of the C-terminus tail of ß-arrestin1 induced loss of surface exposed-VE-cadherin, promoted monolayer disorganization and enhanced permeability. Finally, this effect relied on decreased VE-cadherin expression at the transcriptional level, through inhibition of its promoter activity. CONCLUSIONS: Altogether, our results demonstrate that ß-arrestin1 might play multiple functions collectively contributing to endothelial barrier properties. Indeed, in addition to a direct implication in VE-cadherin endocytosis, ß-arrestin1 could also control VE-cadherin transcription and expression. Ultimately, understanding the molecular mechanisms involved in VE-cadherin function might provide therapeutic tools for many human diseases where the vascular barrier is compromised.

13.
Cytokine Growth Factor Rev ; 24(1): 13-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22981349

ABSTRACT

Experiments in IL-15(-/-) and IL-15Rα(-/-) mice show that intra-renal IL-15, through IL-15Rα behaves as an epithelial survival factor. Recent data highlight new functions of IL-15 in renal homeostasis mediated by IL-15Rγ (CD132). Indeed, in CD132+ renal epithelial tubular cells IL-15 preserves E-cadherin expression inhibiting epithelial-mesenchymal transition (EMT). By contrast, during allograft rejection, the increased intra-graft IL-15 expression favors tubular destruction facilitating the intraepithelial recruitment of CD8 T cells expressing the E-cadherin ligand CD103. In renal cancer, loss of CD132 by epithelial cells defines a tumoral microenvironment where IL-15 triggers E-cadherin down-regulation and EMT. Finally, in CD132+ renal cancer stem cells IL-15 induces the generation of non-tumorigenic epithelial cells sensitive to cytotoxic drugs. These findings are discussed in the light of IL-15-based immunotherapy for renal cancer.


Subject(s)
Homeostasis , Interleukin-15 Receptor alpha Subunit/metabolism , Interleukin-15/metabolism , Kidney Neoplasms/immunology , Kidney/physiology , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Cadherins/biosynthesis , Cellular Microenvironment , Epithelial-Mesenchymal Transition , Graft Rejection , Humans , Integrin alpha Chains/metabolism , Interleukin Receptor Common gamma Subunit/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Mice , Mice, Knockout , Neoplastic Stem Cells
14.
PLoS One ; 7(9): e45562, 2012.
Article in English | MEDLINE | ID: mdl-23029099

ABSTRACT

Glioblastoma constitutes the most aggressive and deadly of brain tumors. As yet, both conventional and molecular-based therapies have met with limited success in treatment of this cancer. Among other explanations, the heterogeneity of glioblastoma and the associated microenvironment contribute to its development, as well as resistance and recurrence in response to treatments. Increased vascularity suggests that tumor angiogenesis plays an important role in glioblastoma progression. However, the molecular crosstalk between endothelial and glioblastoma cells requires further investigation. To examine the effects of glioblastoma-derived signals on endothelial homeostasis, glioblastoma cell secretions were collected and used to treat brain endothelial cells. Here, we present evidence that the glioblastoma secretome provides pro-angiogenic signals sufficient to disrupt VE-cadherin-mediated cell-cell junctions and promote endothelial permeability in brain microvascular endothelial cells. An unbiased angiogenesis-specific antibody array screen identified the chemokine, interleukin-8, which was further demonstrated to function as a key factor involved in glioblastoma-induced permeability, mediated through its receptor CXCR2 on brain endothelia. This underappreciated interface between glioblastoma cells and associated endothelium may inspire the development of novel therapeutic strategies to induce tumor regression by preventing vascular permeability and inhibiting angiogenesis.


Subject(s)
Brain Neoplasms/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Glioblastoma/metabolism , Interleukin-8/metabolism , Receptors, Interleukin-8B/metabolism , Brain Neoplasms/genetics , Capillary Permeability/drug effects , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Interleukin-8/pharmacology , Receptors, Interleukin-8B/genetics
15.
PLoS One ; 7(2): e31624, 2012.
Article in English | MEDLINE | ID: mdl-22363690

ABSTRACT

The ability of Interleukin-15 (IL-15) to activate many immune antitumor mechanisms renders the cytokine a good candidate for the therapy of solid tumors, particularly renal cell carcinoma. Although IL-15 is being currently used in clinical trials, the function of the cytokine on kidney's components has not been extensively studied; we thus investigated the role of IL-15 on normal and tumor renal epithelial cells. Herein, we analyzed the expression and the biological functions of IL-15 in normal renal proximal tubuli (RPTEC) and in their neoplastic counterparts, the renal clear cell carcinomas (RCC). This study shows that RPTEC express a functional heterotrimeric IL-15Rαßγc complex whose stimulation with physiologic concentrations of rhIL-15 is sufficient to inhibit epithelial mesenchymal transition (EMT) commitment preserving E-cadherin expression. Indeed, IL-15 is not only a survival factor for epithelial cells, but it can also preserve the renal epithelial phenotype through the γc-signaling pathway, demonstrating that the cytokine possess a wide range of action in epithelial homeostasis. In contrast, in RCC in vitro and in vivo studies reveal a defect in the expression of γc-receptor and JAK3 associated kinase, which strongly impacts IL-15 signaling. Indeed, in the absence of the γc/JAK3 couple we demonstrate the assembly of an unprecedented functional high affinity IL-15Rαß heterodimer, that in response to physiologic concentrations of IL-15, triggers an unbalanced signal causing the down-regulation of the tumor suppressor gene E-cadherin, favoring RCC EMT process. Remarkably, the rescue of IL-15/γc-dependent signaling (STAT5), by co-transfecting γc and JAK3 in RCC, inhibits EMT reversion. In conclusion, these data highlight the central role of IL-15 and γc-receptor signaling in renal homeostasis through the control of E-cadherin expression and preservation of epithelial phenotype both in RPTEC (up-regulation) and RCC (down-regulation).


Subject(s)
Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/physiopathology , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-15/metabolism , Kidney Neoplasms/physiopathology , Kidney Tubules, Proximal/physiopathology , Signal Transduction , Cadherins/metabolism , Carcinoma, Renal Cell/pathology , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Humans , Immunohistochemistry , Interleukin-15/pharmacology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Signal Transduction/drug effects , Solubility/drug effects , Up-Regulation/drug effects
16.
J Natl Cancer Inst ; 103(24): 1884-98, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-22043039

ABSTRACT

BACKGROUND: Many renal cancer patients experience disease recurrence after immunotherapy or combined treatments due to persistence of cancer stem cells (CSCs). The identification of reliable inducers of CSC differentiation may facilitate the development of efficient strategies for eliminating CSCs. We investigated whether interleukin 15 (IL-15), a regulator of kidney homeostasis, induces the differentiation of CD105-positive (CD105(+)) CSCs from human renal cancers. METHODS: CD105(+) CSCs were cultured to preserve their stem cell properties and treated with recombinant human IL-15 (rhIL-15) to evaluate their ability to differentiate, to acquire sensitivity to chemotherapeutic drugs, and to form spheroids in vitro and tumors in vivo. Expression of stem cell and epithelial markers were studied by flow cytometry, immunocytochemistry, and immunoblotting. Identification of a CSC side population fraction and its sensitivity to chemotherapy drugs and expression of ATP-binding cassette (ABC) transporters and aldehyde dehydrogenase (ALDH) activities were determined by flow cytometry. Spheroid formation was determined in limiting dilution assay. Xenograft tumors were generated in severe combined immunodeficient mice (n = 12-18 mice per group). All statistical tests were two-sided. RESULTS: CD105(+) CSCs treated with rhIL-15 at 10 pg/mL differentiated into cells expressing epithelial markers. rhIL-15 induced epithelial differentiation of all CD105(+) CSCs subsets and blocked CSC self-renewal (sphere-forming ability) and their tumorigenic properties in severe combined immunodeficient mice. Vinblastine and paclitaxel induced statistically significant higher levels of apoptosis in rhIL-15-differentiated epithelial cells compared with CD105(+) CSCs (mean percentage of apoptotic cells, vinblastine: 33% vs 16.5%, difference = 16.5%, 95% confidence interval = 12.25% to 20.74%, P = .0025; paclitaxel: 35% vs 11.6%, difference = 23.4%, 95% confidence interval = 22.5% to 24.24%, P = .0015). The higher sensitivity of rhIL-15-differentiated epithelial cells to chemotherapeutic drugs was associated with loss of detoxifying mechanisms such as ALDH and ABC transporter activities. CONCLUSION: IL-15 directs the epithelial differentiation of renal CSCs and meets the criteria for a treatment strategy: CSC pool depletion and generation of differentiated nontumorigenic cells that are sensitive to chemotherapeutic agents.


Subject(s)
Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Interleukin-15/pharmacology , Kidney Neoplasms/drug therapy , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/drug effects , Receptors, Cell Surface/metabolism , ATP-Binding Cassette Transporters/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Endoglin , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Interleukin-15/therapeutic use , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , STAT5 Transcription Factor/drug effects , STAT5 Transcription Factor/metabolism , Secondary Prevention , Transplantation, Heterologous
17.
Bull Cancer ; 98(5): 32-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21609895

ABSTRACT

Primary human epithelial renal cells of normal (HRE), paratumoral (pTEC) and tumoral (RCC) origin display important differences, concerning the expression and biological effects of the IL-15/IL-15R system that deeply influences the evolution of the tumour microenvironment. A major distinguishing feature is represented in RCC and pTEC by the loss of the γc chain leading to the assembly of a IL-15Rαß heterodimer that in response to physiologic concentrations of IL-15 initiates the process of their epithelial-mesenchymal transition (EMT). In contrast, this treatment in HRE cells, which display the IL-15Rαßγc heterotrimer, causes opposite effects inhibiting their drift towards EMT. Thus, IL-15 at physiologic concentrations displays novel functions acting as a major regulator of renal epithelial homeostasis. As second distinguishing feature, RCC and pTEC but not HRE cells express a trans-membrane-bound IL-15 (tmb-IL-15) able to deliver a reverse signal in response to the soluble IL-15Rα chain inducing their EMT. In conclusion, comparison of primary normal (HRE) to primary pathological cells (pTEC and RCC) highlights two major issues: (1) IL-15 is a major regulator of epithelial homeostasis; (2) "apparently normal" pTEC cells, could contribute to organize a generalized "pre-neoplastic" environment committed to favour tumour progression.


Subject(s)
Carcinoma, Renal Cell/metabolism , Epithelial-Mesenchymal Transition/physiology , Interleukin-15/physiology , Kidney Neoplasms/metabolism , Kidney/metabolism , Cadherins/metabolism , Carcinoma, Renal Cell/pathology , Cell Communication , Epithelial Cells , Humans , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-15/pharmacology , Interleukin-15 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Kidney/cytology , Kidney Neoplasms/pathology , Recombinant Proteins/pharmacology , Tumor Microenvironment , Vimentin/metabolism
18.
PLoS One ; 5(12): e16001, 2010 Dec 30.
Article in English | MEDLINE | ID: mdl-21209864

ABSTRACT

Monoclonal antibodies (MAbas) constitute remarkable tools to analyze the relationship between the structure and the function of a protein. By immunizing a mouse with a 29mer peptide (K159) formed by residues 147 to 175 of the HIV-1 integrase (IN), we obtained a monoclonal antibody (MAba4) recognizing an epitope lying in the N-terminal portion of K159 (residues 147-166 of IN). The boundaries of the epitope were determined in ELISA assays using peptide truncation and amino acid substitutions. The epitope in K159 or as a free peptide (pep-a4) was mostly a random coil in solution, while in the CCD (catalytic core domain) crystal, the homologous segment displayed an amphipathic helix structure (α4-helix) at the protein surface. Despite this conformational difference, a strong antigenic crossreactivity was observed between pep-a4 and the protein segment, as well as K156, a stabilized analogue of pep-a4 constrained into helix by seven helicogenic mutations, most of them involving hydrophobic residues. We concluded that the epitope is freely accessible to the antibody inside the protein and that its recognition by the antibody is not influenced by the conformation of its backbone and the chemistry of amino acids submitted to helicogenic mutations. In contrast, the AA →Glu mutations of the hydrophilic residues Gln148, Lys156 and Lys159, known for their interactions with LTRs (long terminal repeats) and inhibitors (5CITEP, for instance), significantly impaired the binding of K156 to the antibody. Moreover, we found that in competition ELISAs, the processed and unprocessed LTR oligonucleotides interfered with the binding of MAba4 to IN and K156, confirming that the IN α4-helix uses common residues to interact with the DNA target and the MAba4 antibody. This also explains why, in our standard in vitro concerted integration assays, MAba4 strongly impaired the IN enzymatic activity.


Subject(s)
HIV Integrase/chemistry , HIV Integrase/genetics , HIV-1/genetics , Terminal Repeat Sequences/drug effects , Antibodies/chemistry , Antibodies, Monoclonal/chemistry , Antigens/chemistry , Catalysis , DNA/chemistry , Enzyme-Linked Immunosorbent Assay/methods , Epitope Mapping , Epitopes/chemistry , Mutation , Peptides/chemistry , Protein Binding , Protein Conformation
19.
Eur Cytokine Netw ; 19(3): 131-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18775806

ABSTRACT

Since the withdrawal of penicillin determinants from the market, in addition to the hazard of re-exposing the patient to the drug, skin testing for the diagnosis of penicillin allergy has become less accurate and less standardized. The assay currently used, the lymphocyte transformation test (LTT), lacks sufficient sensitivity, and requires the use of radioactive material. The objective of this study was to establish an accessible and reliable method for the safe diagnosis of penicillin allergy. Peripheral blood mononuclear cells (PBMC) were isolated from 18 patients who were allergic to penicillin and 12 control subjects using the Ficoll-Hypaque method. The isolated, sensitized cells were stimulated in vitro with amoxicillin (1 mg/mL). Stimulation with phytohemagglutinin A (PHA) was used as the positive control. Transcriptional expression of specific cytokines (IL-2, -4, -5 and -13, TGF-beta, TNF-alpha and IFN-gamma) was assessed by RT-PCR. IFN-gamma expression was also evaluated by ELISPOT. Secreted levels of IL-2, -5 and IFN-gamma were measured by ELISA. All of these assays were performed two or five days, post-stimulation. This study of the in vitro diagnosis of penicillin allergy by the measurement of cytokine concentration in the supernatants of sensitized lymphocytes cultures involved the largest number of patients to-date. The Delta values (difference in cytokine concentration in the supernatants before and after stimulation) were compared between cases and controls using different statistical tests (Student's t test and the Mann-Whitney rank test). Of the various tests performed in this study, measurement of secreted cytokines using ELISA was the most sensitive and specific (80% and 100% respectively). In vitro stimulation of human lymphocytes sensitized to amoxicillin is a safe and useful test for the diagnosis of penicillin allergy if the ELISA is used to measure cytokine expression. The advantages are that it can be performed by many laboratories since kits to determine cytokines are widely available, and it can be done without the need for particularly specialized equipment.


Subject(s)
Amoxicillin/pharmacology , Cytokines/genetics , Gene Expression Profiling , T-Lymphocytes/drug effects , Adolescent , Adult , Aged , Anti-Bacterial Agents , Cell Proliferation/drug effects , Cells, Cultured , Drug Hypersensitivity/blood , Drug Hypersensitivity/etiology , Drug Hypersensitivity/genetics , Female , Humans , Interferon-gamma/genetics , Interleukin-13/genetics , Interleukin-2/genetics , Interleukin-4/genetics , Interleukin-5/genetics , Male , Middle Aged , Penicillins/adverse effects , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...