Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Int J Cell Biol ; 2019: 9151394, 2019.
Article in English | MEDLINE | ID: mdl-31360169

ABSTRACT

Renal tubulointerstitial fibrosis is the final common pathway in end-stage renal disease and is characterized by aberrant accumulation of extracellular matrix (ECM) components secreted by myofibroblasts. Tubular type 2 EMT, induced by TGF-ß, plays an important role in renal fibrosis, by participating directly or indirectly in myofibroblasts generation. TGF-ß1-induced apoptosis and fibrosis in experimental chronic murine kidney diseases are concomitantly associated with an intrarenal decreased expression of the IL-15 survival factor. Since IL-15 counteracts TGF-ß1 effects in different cell models, we analyzed whether (1) human chronic inflammatory nephropathies evolving towards fibrosis could be also characterized by a weak intrarenal IL-15 expression and (2) IL-15 could inhibit epithelial-mesenchymal transition (EMT) and excess matrix deposition in human renal proximal tubular epithelial cells (RPTEC). Our data show that different human chronic kidney diseases are characterized by a strong decreased expression of intrarenal IL-15, which is particularly relevant in diabetic nephropathy, in which type 2 tubular EMT plays an important role in fibrosis. Moreover, primary epithelial tubular cultures deprived of growth supplements rapidly produce active TGF-ß1 inducing a "spontaneous" EMT process characterized by the loss of membrane-bound IL-15 (mbIL-15) expression. Both "spontaneous" EMT and recombinant human (rh) TGF-ß1-induced EMT models can be inhibited by treating RPTEC and HK2 cells with rhIL-15. Through a long-lasting phospho-c-jun activation, IL-15 inhibits rhTGF-ß1-induced Snail1 expression, the master inducer of EMT, and blocks TGF-ß1-induced tubular EMT and downstream collagen synthesis. In conclusion, our data suggest that intrarenal IL-15 could be a natural inhibitor of TGF-ß in human kidney able to guarantee epithelial homeostasis and to prevent EMT process. Thus, both in vivo and in vitro an unbalance in intrarenal IL-15 and TGF-ß1 levels could render RPTEC cells more prone to undergo EMT process. Exogenous IL-15 treatment could be beneficial in some human nephropathies such as diabetic nephropathy.

2.
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
3.
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.

4.
Cell Death Differ ; 24(2): 343-356, 2017 02.
Article in English | MEDLINE | ID: mdl-27911441

ABSTRACT

Over the past decade, endoplasmic reticulum (ER) stress has emerged as an important mechanism involved in the pathogenesis of cardiovascular diseases including heart failure. Cardiac therapy based on ER stress modulation is viewed as a promising avenue toward effective therapies for the diseased heart. Here, we tested whether sirtuin-1 (SIRT1), a NAD+-dependent deacetylase, participates in modulating ER stress response in the heart. Using cardiomyocytes and adult-inducible SIRT1 knockout mice, we demonstrate that SIRT1 inhibition or deficiency increases ER stress-induced cardiac injury, whereas activation of SIRT1 by the SIRT1-activating compound STAC-3 is protective. Analysis of the expression of markers of the three main branches of the unfolded protein response (i.e., PERK/eIF2α, ATF6 and IRE1) showed that SIRT1 protects cardiomyocytes from ER stress-induced apoptosis by attenuating PERK/eIF2α pathway activation. We also present evidence that SIRT1 physically interacts with and deacetylates eIF2α. Mass spectrometry analysis identified lysines K141 and K143 as the acetylation sites on eIF2α targeted by SIRT1. Furthermore, mutation of K143 to arginine to mimic eIF2α deacetylation confers protection against ER stress-induced apoptosis. Collectively, our findings indicate that eIF2α deacetylation on lysine K143 by SIRT1 is a novel regulatory mechanism for protecting cardiac cells from ER stress and suggest that activation of SIRT1 has potential as a therapeutic approach to protect the heart against ER stress-induced injury.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Sirtuin 1/metabolism , Acetylation , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/drug effects , Carbazoles/pharmacology , Cell Line , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/genetics , Heat-Shock Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects , Up-Regulation/drug effects
5.
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
6.
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
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.
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
9.
J Urol ; 189(2): 643-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23260549

ABSTRACT

PURPOSE: We determined the location where sperm were identified during microdissection testicular sperm extraction and characterized the subset of patients for whom complete bilateral exploration was most beneficial. MATERIALS AND METHODS: A total of 900 men underwent a first attempt at microdissection testicular sperm extraction. Sperm extraction began with an initial wide incision in the larger testis. If no sperm were identified, the deeper tissue was extensively microdissected. A similar technique was used on the contralateral testis if no sperm were found on the initial side. RESULTS: In 474 men (52.6%) sperm were identified at the first microdissection testicular sperm extraction. Of these men 308 (65%) had sperm identified through the initial wide incision alone. In men with lower preoperative follicle-stimulating hormone, larger testicular volume, a varicocele history and hypospermatogenesis on preoperative or intraoperative diagnostic biopsy there was a greater chance of finding sperm in the initial wide incision alone (p <0.05). Only 40 of the 506 men (8%) who underwent bilateral testicular microdissection had sperm found on the contralateral side when no sperm were identified on the initial side. In men with Klinefelter syndrome and small testes the chance of sperm retrieval was higher on the contralateral side after negative unilateral microdissection (p <0.05). CONCLUSIONS: More than a third of the men with nonobstructive azoospermia required complete microdissection of the testes to identify sperm. Sperm were found on the contralateral side in up to 8% of the men in whom no sperm were identified in the initial testis.


Subject(s)
Azoospermia , Microdissection , Sperm Retrieval , Adult , Humans , Male , Retrospective Studies , Testis
10.
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
11.
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
12.
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
13.
PLoS One ; 6(4): e19366, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559410

ABSTRACT

B-cells can contribute to the pathogenesis of autoimmune diseases not only through auto-antibody secretion but also via cytokine production. Therapeutic depletion of B-cells influences the functions and maintenance of various T-cell subsets. The mechanisms governing the functional heterogeneity of B-cell subsets as cytokine-producing cells are poorly understood. B-cells can differentiate into two functionally polarized effectors, one (B-effector-1-cells) producing a Th-1-like cytokine pattern and the other (Be2) producing a Th-2-like pattern. IL-12 and IFN-γ play a key role in Be1 polarization, but the initial trigger of Be1 commitment is unclear. Type-I-interferons are produced early in the immune response and prime several processes involved in innate and adaptive responses. Here, we report that IFN-α triggers a signaling cascade in resting human naive B-cells, involving STAT4 and T-bet, two key IFN-γ gene imprinting factors. IFN-α primed naive B-cells for IFN-γ production and increased IFN-γ gene responsiveness to IL-12. IFN-γ continues this polarization by re-inducing T-bet and up-regulating IL-12Rß2 expression. IFN-α and IFN-γ therefore pave the way for the action of IL-12. These results point to a coordinated action of IFN-α, IFN-γ and IL-12 in Be1 polarization of naive B-cells, and may provide new insights into the mechanisms by which type-I-interferons favor autoimmunity.


Subject(s)
B-Lymphocytes/cytology , Gene Expression Regulation , Interferon-alpha/metabolism , Autoimmunity , B-Lymphocytes/immunology , Cell Differentiation , Cell Separation , Cytokines/metabolism , Dimerization , Humans , Interferon Type I/metabolism , Microscopy, Confocal/methods , Phenotype , STAT4 Transcription Factor/metabolism , T-Lymphocytes/immunology , Th2 Cells/cytology
14.
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
15.
Eur J Immunol ; 39(10): 2877-84, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19728309

ABSTRACT

NK lymphocytes and type I IFN (IFN-alpha/beta) are major actors of the innate anti-viral response that also influence adaptive immune responses. We evaluated type I IFN production by human NK cells in response to polyI:C, a potent type I IFN-inducing TLR3 agonist. PolyI:C plus IL-2/IL-12 induced IFN-beta (but not IFN-alpha) mRNA expression and protein production by highly pure human NK cells and by the human NK cell line NK92. Neutralizing anti-IFNAR1 or anti-IFN-beta Ab prevented the production of IFN-gamma induced by polyI:C plus IL-2/IL-12. Similarly, IFN-gamma production induced by polyI:C plus IL-12 was reduced in NK cells isolated from IFNAR1(-/-) compared with WT mice. The ability of polyI:C plus IL-12 to induce IFN-gamma production was related to an increase of TLR3, Mda5 and IFNAR expression and by an increase of STAT1 and STAT4 phosphorylation. Collectively, these data demonstrate that NK cells, in response to polyI:C plus IL-2/IL-12, produce IFN-beta that induce, in an autocrine manner, the production of IFN-gamma and thereby highlight that NK cells may control the outcome of protective or injurious immune responses through type I IFN secretion.


Subject(s)
Autocrine Communication/immunology , Interferon-beta/metabolism , Interferon-gamma/metabolism , Interleukin-12/pharmacology , Interleukin-2/pharmacology , Killer Cells, Natural/metabolism , Poly I-C/pharmacology , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Line , Cells, Cultured , DEAD-box RNA Helicases/genetics , Gene Expression/drug effects , Gene Expression/genetics , Humans , Interferon-Induced Helicase, IFIH1 , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/pharmacology , Interferon-gamma/genetics , Killer Cells, Natural/drug effects , Kinetics , Mice , Mice, Inbred Strains , Mice, Knockout , Phosphorylation/drug effects , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptors, Interleukin-12/genetics , STAT1 Transcription Factor/metabolism , STAT4 Transcription Factor/metabolism , Toll-Like Receptor 3/genetics
16.
J Pediatr Hematol Oncol ; 31(10): 782-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19755920

ABSTRACT

Erdheim-Chester disease is a rare, non-Langerhans systemic histiocytosis characterized by bilateral sclerosis of the metaphyseal regions of the long bones and infiltration in other organs. The histopathologic hallmark is defined by a mononuclear infiltrate of foamy histiocytes and rare pathognomonic Touton giant cells with extensive fibrosis. This condition is exceptional in children. We report here a case of Erdheim-Chester disease in a 10-year-old girl with retroperitoneal infiltration and bone involvement, for whom the diagnosis was only established after a 3-year course with multiple biopsies. It is also the first pediatric case successfully treated with interferon-alpha suggesting that interferon-alpha can be a safe and efficient first-line therapy for this disease in children.


Subject(s)
Erdheim-Chester Disease/diagnosis , Erdheim-Chester Disease/drug therapy , Bone Diseases , Cell Movement , Child , Female , Humans , Interferon-alpha/therapeutic use , Remission Induction
17.
J Biol Chem ; 284(36): 24328-40, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19561067

ABSTRACT

Type I interferons (IFNs) bind IFNAR receptors and activate Jak kinases and Stat transcription factors to stimulate the transcription of genes downstream from IFN-stimulated response elements. In this study, we analyze the role of protein palmitoylation, a reversible post-translational lipid modification, in the functional properties of IFNAR. We report that pharmacological inhibition of protein palmitoylation results in severe defects of IFN receptor endocytosis and signaling. We generated mutants of the IFNAR1 subunit of the type I IFN receptor, in which each or both of the two cysteines present in the cytoplasmic domain are replaced by alanines. We show that cysteine 463 of IFNAR1, the more proximal of the two cytoplasmic cysteines, is palmitoylated. A thorough microscopic and biochemical analysis of the palmitoylation-deficient IFNAR1 mutant revealed that IFNAR1 palmitoylation is not required for receptor endocytosis, intracellular distribution, or stability at the cell surface. However, the lack of IFNAR1 palmitoylation affects selectively the activation of Stat2, which results in a lack of efficient Stat1 activation and nuclear translocation and IFN-alpha-activated gene transcription. Thus, receptor palmitoylation is a previously undescribed mechanism of regulating signaling activity by type I IFNs in the Jak/Stat pathway.


Subject(s)
Antiviral Agents/pharmacology , Interferon-alpha/pharmacology , Palmitic Acid/metabolism , Protein Processing, Post-Translational/drug effects , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Amino Acid Substitution , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Endocytosis/drug effects , Endocytosis/genetics , Humans , Mice , Mutation, Missense , Protein Processing, Post-Translational/genetics , Protein Structure, Tertiary/genetics , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics , Signal Transduction/drug effects
18.
Ann Neurol ; 65(5): 499-509, 2009 May.
Article in English | MEDLINE | ID: mdl-19475668

ABSTRACT

OBJECTIVE: T-helper 1 (Th1) and Th17 lymphocytes are involved in experimental autoimmune encephalomyelitis, the model of multiple sclerosis (MS). We characterized the Th1/Th17 cell populations in peripheral blood (PB), their interferon (IFN) receptor expression sensitivity to IFN-beta in MS patients. METHODS: In 30 untreated patients with active MS (AMS) and 32 with inactive MS (IMS), and in 22 healthy subjects, we measured intracellular cytokine expression, interleukin-17-producing myelin basic protein-stimulated PB lymphocytes, surface IFN type I receptor chain1 (IFN-alphaR1) expression, IFN-beta-dependent signal transducer and activator of transcription 1 (STAT1) phosphorylation, and apoptosis of anti-CD3 monoclonal antibody-stimulated PB lymphocytes. RESULTS: Th17 cell percentage increased around sevenfold in AMS compared with IMS or healthy subjects, but there was no change in Th1 cells. Th17 cells in AMS were myelin basic protein specific. The longitudinal follow-up of 18 MS patients shifting between AMS and IMS showed that the percentage of Th17 but not Th1 cells always increased in AMS. IFN-alphaR1 expression, IFN-beta-induced STAT1 activation, and apoptosis were significantly greater in Th17 than Th1 cells. IFN-alphaR1 expression and IFN-beta-dependent STAT1 activation progressively increased in vitro with a highly significant positive correlation only in developing Th17 but not in Th0 or Th1 cells. INTERPRETATION: Evidence that an expansion of peripheral Th17 cells, a Th subset that can infiltrate brain parenchyma and damage cells, is associated with disease activity in MS. The greater IFN-alphaR1 level expressed by Th17 compared with Th1 cells might make them a selective target for IFN-beta therapy.


Subject(s)
Immunologic Factors/pharmacology , Interferon-beta/pharmacology , Interleukin-17/metabolism , Multiple Sclerosis/pathology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , Adult , Annexin A5/metabolism , Antigens, CD/metabolism , Apoptosis/drug effects , Cell Differentiation/physiology , Chi-Square Distribution , Dactinomycin/analogs & derivatives , Dactinomycin/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Interferon-beta/metabolism , Leukocytes, Mononuclear/pathology , Male , Multiple Sclerosis/blood , Receptor, Interferon alpha-beta/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Time Factors
19.
Cancer Res ; 69(4): 1561-9, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19190330

ABSTRACT

Although interleukin-15 (IL-15) is a powerful immunomodulatory factor that has been proposed for cancer immunotherapy, its intratumoral expression may be correlated with tumor progression and/or poor clinical outcome. Therefore, neoplasias potentially sensitive to immunotherapy should be checked for their IL-15 expression and function before choosing immunotherapy protocols. Primary human renal cancer cells (RCC) express a novel form of membrane-bound IL-15 (mb-IL-15), which displays three major original properties: (a) It is expressed as a functional membrane homodimer of 27 kDa, (b) it is shed in the extracellular environment by the metalloproteases ADAM17 and ADAM10, and (c) its stimulation by soluble IL-15 receptor alpha (s-IL-15Ralpha) chain triggers a complex reverse signal (mitogen-activated protein kinases, FAK, pMLC) necessary and sufficient to ~induce epithelial-mesenchymal transdifferentiation (EMT), a crucial process in tumor progression whose induction is unprecedented for IL-15. In these cells, complete EMT is characterized by a dynamic reorganization of the cytoskeleton with the subsequent generation of a mesenchymal/contractile phenotype (alpha-SMA and vimentin networks) and the loss of the epithelial markers E-cadherin and ZO-1. The retrosignaling functions are, however, hindered through an unprecedented cytokine/receptor interaction of mb-IL-15 with membrane-associated IL-15Ralpha subunit that tunes its signaling potential competing with low concentrations of the s-IL-15Ralpha chain. Thus, human RCC express an IL-15/IL-15R system, which displays unique biochemical and functional properties that seem to be directly involved in renal tumoral progression.


Subject(s)
Carcinoma, Renal Cell/immunology , Epithelial Cells/immunology , Interleukin-15/physiology , Kidney Neoplasms/immunology , Mesoderm/immunology , Receptors, Interleukin-15/immunology , Antibodies, Monoclonal/therapeutic use , Carcinoma, Renal Cell/pathology , Carcinoma, Small Cell/immunology , Carcinoma, Small Cell/pathology , Cell Differentiation , Cell Line, Tumor , Cell Membrane/immunology , Epithelial Cells/pathology , Flow Cytometry , Humans , Immunotherapy/methods , Interleukin-15/immunology , Interleukin-15/therapeutic use , Kidney Neoplasms/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mesoderm/pathology , Neoplasm Metastasis/immunology , Sodium Acetate/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
20.
Eur J Immunol ; 38(4): 1058-64, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18350548

ABSTRACT

B cell activating factor (BAFF) plays a key role in promoting B lymphocyte activation. We investigated whether danger signals induce BAFF secretion by cultured salivary gland epithelial cells (SGEC), which are the target of primary Sjögren's syndrome, a prototypic systemic autoimmune disease. SGEC cultures were established from minor salivary glands obtained from ten patients with pSS or sicca symptoms. BAFF mRNA and protein were measured after stimulation of the different Toll-like receptors (TLR) by agonists or viruses. The expression of TLR2, -3, and -7 was detected in SGEC. Poly (I:C) (a synthetic TLR3 agonist) and reovirus-1 (a dsRNA virus) induced high expression of BAFF mRNA (multiplied by a factor of 246 +/- 39 (SEM) and 347 +/- 66, respectively) and of BAFF protein secretion (58.49 +/- 4.34 pg/mL and 69.73 +/- 5.67). Inhibition of both the endosomal (by chloroquine) and IFN (by anti-IFNAR antibody) pathways partly inhibited BAFF expression. Treatment with both dsRNA virus and poly (I:C) induced high levels of BAFF mRNA and protein expression by SGEC, through pathways dependent on and independent of TLR and dependent on and independent of IFN. BAFF induction by target organs of autoimmune diseases after viral infection may be a link between innate immunity and autoimmunity.


Subject(s)
B-Cell Activating Factor/metabolism , Epithelial Cells/immunology , Interferon Type I/metabolism , Salivary Glands/immunology , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Animals , Autoimmunity/immunology , B-Cell Activating Factor/genetics , Cells, Cultured , Chlorocebus aethiops , Epithelial Cells/metabolism , Gene Expression Regulation , Herpesvirus 1, Human/immunology , Humans , Immunity, Innate/immunology , Ligands , Orthoreovirus, Mammalian/immunology , Salivary Glands/metabolism , Vero Cells , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...